WO2023150186A1 - Dgk targeting compounds and uses thereof - Google Patents

Dgk targeting compounds and uses thereof Download PDF

Info

Publication number
WO2023150186A1
WO2023150186A1 PCT/US2023/012145 US2023012145W WO2023150186A1 WO 2023150186 A1 WO2023150186 A1 WO 2023150186A1 US 2023012145 W US2023012145 W US 2023012145W WO 2023150186 A1 WO2023150186 A1 WO 2023150186A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
substituted
ring
methyl
Prior art date
Application number
PCT/US2023/012145
Other languages
French (fr)
Inventor
Jesus Raul Medina
Jennifer MACALUSO
Original Assignee
Arvinas Operations, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Arvinas Operations, Inc. filed Critical Arvinas Operations, Inc.
Publication of WO2023150186A1 publication Critical patent/WO2023150186A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic 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
    • C07D513/02Heterocyclic 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
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • SUMMARY Disclosed herein are compounds that have activity as inhibitors of diacylglycerol kinase alpha (DGK ⁇ ). Additionally, in embodiments, the disclosed compounds cause the degradation of DGK ⁇ . In embodiments, compounds described herein have desirable efficacy, stability, bioavailability, therapeutic index, and toxicity values that are important to their use as pharmaceuticals.
  • Ring A is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaromatic ring fused to ring B; Ring C is a heterocyclic ring, which may be further substituted in addition to R 3 and R 4 ;
  • R 1 is C 1 –C 4 alkyl;
  • R 2 is H or an electron withdrawing functional group;
  • each R 3 is independently NR 5 R 6 ;
  • each R 4 is independently C 1 –C 4 alkyl or C 1 –C 4 alkoxy, or one R 3 and one R 4 , together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring;
  • R 5 and R 6 are each independently H, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or R 5 and R 6 , together with the nitrogen to which they
  • the compound has the structure of Formula (II): (II) or a pharmaceutically acceptable salt thereof, wherein R 9 is H or C 1 –C 4 alkyl.
  • R 9 is straight or branched C 1 –C 4 alkyl.
  • R 9 is methyl.
  • the compound has the structure of Formula (III): (III) or a pharmaceutically acceptable salt thereof, wherein R 10 is C 1 –C 4 alkyl or an electron withdrawing functional group. In embodiments of Formula (III), R 10 is straight or branched C 1 –C 4 alkyl. In embodiments of the compound of Formula (III), the compound has the structure of Formula (IIIa): (IIIa) or a pharmaceutically acceptable salt thereof.
  • R 10 is -Cl. In embodiments of the compound of Formula (III) and (IIIa), R 10 is -C ⁇ N. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R 1 is straight or branched C 1 –C 4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R 1 is methyl.
  • R 2 is H. In embodiments of the compounds of Formula (I), (II), (III), and (IIIa), R 2 is -C ⁇ N. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R 4 is C 1 – C4 alkyl (e.g., straight or branched C 1 –C 4 alkyl). In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R 4 is C 1 – C 4 alkoxy.
  • ring C is further substituted by one or more substituents selected from R 15 , R 18 , R 19 , R 20 , R 21 , and R 22 .
  • ring C is selected from ; ; ; and , wherein: R 18 and R 19 are each, independently H, OH, C 1 –C 4 alkoxy, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), or -NR 23 R 24 , or R 18 and R 19 , together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R 20 is H or C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), or R 18 and R 20 , or R 19 and R 20 , together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring; R 21 is H, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl
  • R 21 is straight or branched C 1 –C 4 alkyl.
  • R 22 is straight or branched C 1 –C 4 alkyl.
  • R 23 and R 24 are each, independently H, straight or branched C 1 –C 4 alkyl, aryl, or heteroaryl.
  • R 15 is straight or branched C 1 –C 4 alkyl.
  • ring C is , wherein R 18 and R 19 , together with the carbon atom to which they are attached, form a 5- or 6-membered substituted or unsubstituted carbocyclic ring or a 5- or 6- membered substituted or unsubstituted heterocyclic ring.
  • ring C is , wherein R 18 and R 20 , or R 19 and R 20 , together with the carbon atoms to which they are attached form a substituted or unsubstituted 5-, 6-, or 7-membered carbocyclic ring or a substituted or unsubstituted 5-, 6-, or 7-membered heterocyclic ring.
  • R 20 is straight or branched C 1 –C 4 alkyl.
  • ring C is selected from ; ; ; ; ; ; ; and in embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is wherein R 21 is , wherein: R 25 is selected from OH, halogen, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), C 1 –C 4 alkoxy, C 1 –C 4 haloalkyl, C 1 –C 4 haloalkoxy, aryl, and heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R 21 to N.
  • R 25 is selected from OH, halogen, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), C 1 –C 4 alkoxy, C 1 –C 4 haloalkyl, C 1 –C 4 haloalkoxy, aryl, and
  • ring C is , wherein R is , wherein: R 25 is selected from OH, halogen, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), C 1 –C 4 alkoxy, C 1 –C 4 haloalkyl, C 1 –C 4 haloalkoxy, aryl, and heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R 21 to N.
  • R 25 is selected from OH, halogen, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), C 1 –C 4 alkoxy, C 1 –C 4 haloalkyl, C 1 –C 4 haloalkoxy, aryl, and heteroaryl
  • t is 0, 1, or 2; and indicates the point of attachment of R 21 to N.
  • each R 25 is independently selected from OH, halogen, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), C 1 –C 4 alkoxy, C 1 –C 4 haloalkyl, C 1 –C 4 haloalkoxy, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • ring C is , wherein R 21 is selected from In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is w 21 herein R is selected from ; ; ; ; ; ; and In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from ; ; ; ; ; ; and In embodiments of Ring C, R 22 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 22 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 22 is straight or branched C 1 –C 4 alkyl.
  • ring C is , wherein R 18 and R 19 are each independently selected from -H, -OH, C 1 – C4 alkoxy, C 1 –C 4 alkyl (straight or branched C 1 –C 4 alkyl), and , wherein: R 26 is H, C 1 –C 4 alkyl (straight or branched C 1 –C 4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R 27 is -OH, C 1 –C 4 alkyl (straight or branched C 1 –C 4 alkyl), halogen, C 1 –C 4 alkoxy, or C 1 –C 4 haloalkoxy, or two adjacent R 27 groups, and the carbon atoms two which
  • ring C is selected from and , wherein R 18 and R 19 are each independently selected from -H, -OH, C 1 –C 4 alkyl, and , wherein: R 26 is H, C 1 –C 4 alkyl (straight or branched C 1 –C 4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R 27 is -OH, C 1 –C 4 alkyl (straight or branched C 1 –C 4 alkyl), halogen, C 1 –C 4 alkoxy, or C 1 –C 4 haloalkoxy, or two adjacent R 27 groups, and the carbon atoms two which they are attached, form a substituted or unsubstituted 5- or 6-membered ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R 18 and R 19 are each independently selected from -H, -OH, C 1 –C 4 alkyl, and
  • R 26 is straight or branched C 1 –C 4 alkyl.
  • each R 27 is -OH, straight or branched C 1 –C 4 alkyl, C 1 –C 4 alkoxy, or C 1 –C 4 haloalkoxy.
  • ring C is selected from ; ; ; ; ; ; ; ; In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from .
  • each R 28 is independently selected from -OH, halogen, straight or branched C 1 –C 4 alkyl, C 1 –C 4 haloalkyl, C 1 –C 4 alkoxy, and C 1 –C 4 haloalkoxy.
  • R 29 is straight or branched C 1 –C 4 alkyl.
  • ring C is selected from ; ; ; ; ; ; and In embodiments of any one of the compounds of Formula (I), (II), (III), and (IIIa), ring C is selected from ; and wherein R 34 is C 1 –C 4 alkyl. In embodiments of Ring C, R 34 is straight or branched C 1 –C 4 alkyl.
  • ring C is , wherein R 32 is In embodiments of the compounds of Formula (I), (II), (III), and (IIIa), ring C is ; ; ; ; ; o , wherein R 22 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. In embodiments of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from
  • ring C is or .
  • disclosed herein are compounds selected from:
  • X is CH 2 , O, or NR 29 ; and R 29 is H or C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl).
  • R 29 is H or C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl).
  • compositions comprising one or more compounds disclosed herein and a pharmaceutically acceptable excipient.
  • the pharmaceutical compositions further comprise one or more additional anti-cancer agents.
  • at least one of the additional anti- cancer agents is an immune checkpoint inhibitor.
  • methods treating cancer comprising administering a therapeutically effective amount of one or more compounds disclosed herein, or a pharmaceutical composition thereof, to a subject in need thereof.
  • the cancer is selected from colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma.
  • DGK ⁇ diacylglycerol kinase alpha
  • methods of inhibiting the activity of diacylglycerol kinase alpha comprising administering a therapeutically effective amount one or more compounds disclosed herein, or a pharmaceutical composition thereof, to a subject in need thereof.
  • DGKs diacylglycerol kinases
  • DGKs are lipid kinases that mediate the conversion of diacylglycerol to phosphatidic acid thereby terminating T-cell functions propagated through the TCR signaling pathway.
  • DGKs serve as intracellular checkpoints, and inhibition of DGKs is expected to enhance T-cell signaling pathways and T-cell activation.
  • DGK ⁇ may be viewed as a target for cancer immunotherapy (Riese M. J. et al. Front Cell Dev Biol. (2016) 4: 108; Chen, S. S.
  • the disclosed compounds are selective inhibitors for DGK ⁇ over other diacylglycerol kinases (e.g., DGK ⁇ ). In embodiments, the disclosed compounds cause the degradation of DGK ⁇ . In embodiments, the disclosed compounds cause the selective degradation of DGK ⁇ . In embodiments, the disclosed compounds cause the selective degradation of DGK ⁇ over other diacylglycerol kinases (e.g., DGK ⁇ ).
  • the disclosed compounds can be used to treat certain diseases or disorders.
  • the disclosed compounds can be used for the treatment of certain cancers (e.g., colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma).
  • cancers e.g., colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma.
  • an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal.
  • an in vitro cell can be a cell in a cell culture.
  • an in vivo cell is a cell living in an organism such as a mammal.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • “contacting” the DGK ⁇ enzyme with a compound of the invention includes the administration of a compound of the present invention to a subject, such as a human, having DGK ⁇ , as well as, for example, introducing a of compound the invention into a sample containing a cellular or purified preparation containing the DGK ⁇ enzyme.
  • DGK ⁇ degrader refers to an agent that targets the DGK ⁇ enzyme resulting in its degradation.
  • DGK ⁇ inhibitor refers to an agent capable of inhibiting the enzymatic activity of diacylglycerol kinase alpha (DGK ⁇ ) in T-cells resulting in enhanced T-cell stimulation.
  • the DGK ⁇ inhibitor can be a reversible DGK ⁇ inhibitor.
  • a reversible DGK ⁇ inhibitor is a compound that reversibly inhibits DGK ⁇ enzyme activity either at the catalytic site or at a non-catalytic site.
  • patient “subject,” and “individual” are used interchangeably herein and refer to either a human or a non-human animal.
  • DGK ⁇ diacylglycerol kinases
  • selective degraders of DGK ⁇ refers to a compound’s to selectively degrade DGK ⁇ as compared to other diacylglycerol kinases (e.g., DGK ⁇ ).
  • Treating a condition or patient, and “treatment” refer to taking steps to obtain beneficial or desired results, including clinical results, via administration of a compound or composition of the present invention.
  • Beneficial or desired results include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total. “Treatment” also can mean prolonging survival as compared to expected survival if not receiving treatment. “Administering” or “administration of” a compound or a composition of the invention to a subject can be carried out using any of a variety of routes and methods known to those skilled in the art based on the directions of an attending healthcare provider.
  • a compound or pharmaceutical composition of the invention can be administered to a subject in need of such treatment by any of the following routes: intravenously, arterially, intradermally, intramuscularly, intraperitoneally, subcutaneously, ocularly, sublingually, buccally, orally (by ingestion), intranasally (by inhalation), intravaginally, intraspinally, intrathecally, intracerebrally, and transdermally (by absorption through the skin).
  • routes can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, delayed, or controlled release of the compound or composition.
  • Administering also can be performed, for example, once, a plurality of times, and/or over one or more extended periods. Appropriate methods of administering a compound or pharmaceutical composition of the invention to the subject will also depend on a variety of factors such as, for example, age, weight, gender, and physical condition of the subject, as well as the chemical and biological properties of the compound or pharmaceutical composition (e.g., solubility, absorption, bioavailability, metabolism, stability, and toxicity).
  • a compound or pharmaceutical composition of present invention is administered orally, e.g., to a subject by ingestion.
  • the orally administered compound or pharmaceutical composition is in a controlled release (e.g., a delayed release, extended release, or slow release) formulation.
  • the phrase “conjoint administration” refers to any form of administration of two or more different therapeutic agents such that the second agent is administered while the previously administered therapeutic agent is still effective in the body (e.g., the two agents are simultaneously effective in the patient, which may include synergistic effects of the two agents).
  • the different therapeutic compounds can be administered either in the same formulation or in separate formulations, either simultaneously or sequentially.
  • a “therapeutically effective amount” or a “therapeutically effective dose” of a compound or pharmaceutical composition of the invention is an amount of the drug or composition that, when administered to a subject, will have the intended therapeutic effect.
  • a therapeutically effective amount may be administered in one or more administrations.
  • the effective amount needed for a subject will depend, for example, upon the subject’s age, weight, health, gender, and the nature and extent of the condition (e.g., cancer) being treated.
  • the attending healthcare provider will generally determine the effective amount for a given situation according to these and other factors.
  • alkyl refers to saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl-substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups.
  • a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g., C 1 –C 6 for straight chains, C 3 –C 6 for branched chains), and more preferably four or less carbons in its backbone.
  • alkoxy refers to an alkyl group having an oxygen attached thereto, represented by -O-alkyl.
  • C 1 –C 4 alkoxy includes methoxy, ethoxy, propoxy, isopropoxy, and butoxy.
  • Cx–Cy when used in conjunction with a chemical moiety (e.g, alkyl, alkenyl, alkoxy) is meant to include groups that contain from x to y carbons in the chain.
  • a C 1 –C 6 alkyl group for example, contains from one to six carbon atoms in the chain; a C 1 –C 4 -alkyl contains from one to four carbon atoms in the chain.
  • aryl as used herein, includes substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon.
  • the ring is a 5- to 7-membered ring.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings and wherein at least one of the rings is aromatic, i.e., the other cyclic rings can be selected from cycloalkyls, cycloalkenyls, aryls, heteroaryls, heterocyclyls, and combinations thereof.
  • Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like.
  • carbocyclic ring refers to a non-aromatic saturated or unsaturated ring in which each atom of the ring is carbon that is substituted or unsubstituted.
  • a carbocycle ring contains from 3 to 10 atoms, more preferably from 3 to 8 atoms, including 5 to 7 atoms, such as, for example, 5 or 6 atoms.
  • electron withdrawing group means an atom or functional group that draws electron density from neighboring atoms towards itself, typically by resonance or inductive effects.
  • Exemplary electron withdrawing groups include, but are not limited to, trifluoromethylsulfonyl groups (-SO 2 CF 3 ), substituted or unsubstituted ammonium groups (-NR3 + , wherein each R is H or alkyl), nitro groups (-NO 2 ), sulfonic acid groups (-SO 3 H), sulfonyl groups (SO 2 R, wherein R is alkyl), cyano groups (-C ⁇ N), haloformyl groups (-COX, wherein X is Cl, Br, or I), formyl groups (-CHO), acyl groups (-COR, wherein R is alkyl), carboxyl groups (-CO 2 H), alkoxycarboxyl groups (-CO 2 R, wherein R is alkyl), substituted or unsubstituted amino carbonyl groups (-CONH 2 , -CONHR, -CONR 2 , wherein R is alkyl), halogen groups, and nitroso groups (-
  • halo refers to a substituent group from Group 17 of the periodic table of the elements and includes fluoro (-F), chloro (- Cl), bromo(-Br), and iodo (-I) substituent groups.
  • haloalkyl and haloalkoxy mean alkyl or alkoxy, respectively, substituted with one or more halogen atoms.
  • heteroaryl or “heteroaromatic” includes substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms.
  • heteroaryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be selected from cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and heterocyclyls.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur.
  • heterocyclic ring refers to substituted or unsubstituted non-aromatic ring structure, preferably 3- to 10-membered rings, more preferably 3- to 7-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms.
  • heterocyclic ring also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be selected from cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and heterocyclyls.
  • Heterocyclyl groups include, for example, diazinane, imidazolidine, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, octahydropyrrolo[3,4-c]pyrrole, and the like.
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched, and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic mo
  • aromatic ring includes aryl and heteroaryl rings.
  • ring includes aryl, cycloalkyl, heterocyclic, carbocyclic, and heteroaryl, ring systems.
  • Certain compounds described herein may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. When a disclosed compound is named or depicted by structure with or without indicating stereochemistry, it is understood that the name or structure encompasses all possible stereoisomers, geometric isomers, including essentially pure stereo or geometric isomers, as well as combination thereof.
  • the enrichment of the indicated configuration relative to the opposite configuration is greater than 50%, 60%, 70%, 80%, 90%, 99%, or 99.9%.
  • “Enrichment of the indicated configuration relative to the opposite configuration” is a mole percent and is determined by dividing the number of compounds with the indicated stereochemical configuration at the chiral center(s) by the total number of all the compounds with the same or opposite stereochemical configuration in a mixture.
  • stereochemical configuration at a chiral center in a compound having two or more chiral centers is depicted by its structure (e.g., the configuration is indicated by “wedge” bonds), and the configuration about the two or more chiral centers is labelled as “or1”, this means that compound comprises a mixture of stereoisomers and the stereochemistry is relative in the depicted configuration between the two or more stereocenters. For example, means that the compound comprises a mixture of and .
  • stereochemical configuration at a chiral center in a compound having two or more chiral centers is depicted by its structure (e.g., the configuration is indicated by “wedge” bonds), and the configuration about the two or more chiral centers is labelled as “abs”, this means that the compound is stereochemically enriched as the drawn stereochemistry or as the opposite, i.e., is absolute of one or the other stereoisomers.
  • the configuration is indicated by “wedge” bonds
  • abs the configuration about the two or more chiral centers
  • Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. “Enantiomer” means one of a pair of molecules that are mirror images of each other and are not superimposable. Diastereomers are stereoisomers that contain two or more asymmetrically substituted carbon atoms.
  • “Geometric isomers” are stereoisomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a carbocyclyl ring, or to a bridged bicyclic system. Enantiomeric and diastereomeric mixtures can be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral- phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Enantiomers and diastereomers can also be obtained from diastereomerically- or enantiomerically-pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
  • Compounds of the Invention Disclosed herein are compounds that have activity as inhibitors of DGK ⁇ . In preferred embodiments, the disclosed compounds selectively inhibit DGK ⁇ . In embodiments, the disclosed compounds are selective inhibitors of DGK ⁇ over other diacylglycerol kinases (e.g., DGK ⁇ ). In embodiments, the disclosed compounds cause the degradation of DGK ⁇ . In embodiments, the disclosed compounds cause the selective degradation of DGK ⁇ .
  • the disclosed compounds cause the selective degradation of DGK ⁇ over other diacylglycerol kinases (e.g., DGK ⁇ ).
  • the disclosed compounds have desirable efficacy, stability, bioavailability, therapeutic index, and toxicity values that are important to their use as pharmaceuticals.
  • Ring A is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaromatic ring fused to ring B;
  • Ring C is a substituted or unsubstituted carbocyclic ring or a substituted or unsubstituted heterocyclic ring;
  • R 1 is alkyl (e.g., straight or branched C 1 –C6 alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, isobutyl, neopentyl, pentyl, hexyl));
  • R 2 is H or an electron withdrawing functional group;
  • each R 3 is independently NR 5 R 6 ;
  • each R 4 is independently alkyl (e.g., straight or branched C 1 –C 6 alkyl (e.g.,
  • Ring A is a substituted or unsubstituted heteroaromatic ring fused to ring B
  • Ring C is a substituted or unsubstituted heterocyclic ring
  • R 1 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl);
  • R 2 is H or an electron withdrawing functional group;
  • each R 3 is independently NR 5 R 6 ;
  • each R 4 is independently C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl) or C 1 –C 4 alkoxy, or one R 3 and one R 4 , together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring;
  • R 5 and R 6 are each independently H, substituted or unsubstituted aryl, or substituted or
  • Ring A is a 5- or 6-membered substituted or unsubstituted heteroaromatic ring fused to ring B; Ring C is a 5- to 8-membered substituted or unsubstituted heterocyclic ring;
  • R 1 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl);
  • R 2 is H or an electron withdrawing functional group;
  • each R 3 is independently NR 5 R 6 ;
  • each R 4 is independently C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl) or C 1 –C 4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy), or one R 3 and
  • Ring A is selected from piperidine, diazinane, octahydropyrrolo[3,4-c]pyrrole, imidazolidine, and pyrrolidine.
  • R 9 is straight or branched C 1 –C 4 alkyl.
  • R 9 is methyl, ethyl, propyl, or butyl.
  • R 9 is methyl.
  • disclosed herein is a compound of Formula (A) or Formula (I), having a structure according to Formula (II): (II) or a pharmaceutically acceptable salt thereof, wherein R 9 is H, halogen, cyano (-CN), C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl such as methyl, ethyl, propyl, isopropyl, butyl), C 1 –C 4 haloalkyl (e.g., fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, fluoroethyl, difluoroethyl, trifluoroeth
  • R 9 is straight or branched C 1 –C 4 alkyl.
  • R 9 is methyl, ethyl, propyl, or butyl.
  • R 9 is methyl.
  • R 10 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl) or an electron withdrawing functional group.
  • R 10 is methyl, ethyl, propyl, isopropyl, butyl. In embodiments of Formula III), R 10 is methyl.
  • R 10 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, or butyl).
  • R 10 is straight or branched C 1 –C 4 alkyl.
  • R 10 is methyl (-CH 3 ).
  • halogen e.g.,
  • R 10 is -Cl. Alternatively, R 10 is -F. In embodiments, R 10 is -C ⁇ N. In embodiments, R 10 is -N(R 11 ) 2 , -SO 2 R 12 , or -C(R 13 ) 3 , -CSR 15 , wherein each of R 11 , R 12 , and R 15 is independently H or methyl. In embodiments, R 10 is -COR 14 , wherein R 14 is H or methyl. In embodiments, R 10 is -CHO. In embodiments, R 10 is -CO(CH 3 ). In embodiments, R 10 is -CO 2 (R 16 ), wherein R 16 is H or methyl.
  • R 1 is methyl, ethyl, propyl, or butyl. In embodiments of Formulae (A), (I), (II), (III), or (IIIa), wherein R 1 is methyl.
  • R 2 is selected from H, halogen, -SO 2 CF 3 , - N(R 32 ) 2 , -NO 2 , -SO
  • disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R 2 is H. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R 2 is -C ⁇ N.
  • ring C is further substituted by one or more substituents selected from R 15 , R 18 , R 19 , R 20 , R 21 , and R 22 .
  • ring C is further substituted by one or more substituents selected from R 15 , R 18 , R 19 , R 20 , R 21 , and R 22 wherein: R 15 is straight or branched C 1 –C 4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R 18 and R 19 are each, independently C 1 –C 4 alkoxy, C 1 –C 4 alkyl, or -NR 23 R 24 , or R 18 and R 19 , together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R 20 is straight or branched C 1 –C 4 alkyl, or an electron withdrawing group, or R 18 and R 20 , or R 19 and R 20 , together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring
  • R 4 is straight or branched C 1 –C 4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R 4 is C 1 – C 4 alkoxy.
  • ring C is selected from and , wherein: R 15 is straight or branched C 1 –C 4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R 18 and R 19 are each, independently H, OH, C 1 –C 4 alkoxy, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or -NR 23 R 24 , or R 18 and R 19 , together with the carbon atom to which they are attached, form a substituted or unsubstituted ring (e.g., a 5- or 6-membered substituted or unsubstituted carbocyclic ring or a 5- or 6-
  • ring C is selected from ; ; ; and , wherein: R 15 is straight or branched C 1 –C 4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R 18 and R 19 are each, independently OH, C 1 –C 4 alkoxy, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or -NR 23 R 24 , or R 18 and R 19 , together with the carbon atom to which they are attached, form a substituted or unsubstituted ring (e.g., a 5- or 6-membered substituted or unsubstituted carbocyclic ring or
  • any one of R 32 , R 26 , R 28 , R 29 , R 30 , or R 31 is straight or branched C 1 –C 4 alkyl.
  • ring C is selected from ; and , wherein: R 18 and R 19 are each, independently H, OH, C 1 –C 4 alkoxy, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or -NR 23 R 24 , or R 18 and R 19 , together with the carbon atom to which they are attached, form a substituted or unsubstituted ring (e.g., a 5- or 6-membered substituted or unsubstituted carbocyclic ring
  • R 21 is straight or branched C 1 –C 4 alkyl.
  • R 22 is straight or branched C 1 –C 4 alkyl.
  • R 15 is unsubstituted C 1 –C 4 alkyl.
  • disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is 18 , wherein R and R 19 , together with the carbon atom to which they are attached, form a substituted or unsubstituted 5- or 6-membered carbocyclic ring or a substituted or unsubstituted 5- or 6- membered heterocyclic ring.
  • ring C is selected from ; ; ; ; ; ; ; ; and .
  • ring C is R , wherein R 21 is , wherein Each R 25 is independently selected from OH, halogen (-Cl, -F), C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), C 1 –C 4 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), C 1 –C 4 haloalkyl (e.g., -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -(CH 2 ) 2 CH 2 F, -(CH 2 ) 2 CHF 2 , -(CH 2 ) 2 CHF 2 , -(CH 2 ) 2 CHF 2 , -(CH 2 ) 2 CHF 2 , -(CH
  • ring C is selected from ; ; ; ; ; ; ; ; ; ; ; ; and .
  • R 22 is straight or branched C 1 –C 4 alkyl.
  • ring C is , wherein R 18 and R 19 are each independently selected from -H, -OH, alkyl (e.g., straight or branched C 1 –C 4 alkyl) and , wherein: R 26 is H, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R 27 is -OH, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such methyl, ethyl, propyl, isopropyl, butyl), halogen (-Cl,
  • ring C is R18 R19 ; ; ; ; ; ; ; ; ; or , wherein R 18 and R 19 are each independently selected from -H, -OH, alkyl (e.g., straight or branched C 1 –C 4 alkyl), and , wherein: R 26 is H, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R 27 is -OH, C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as
  • R 26 is straight or branched C 1 –C 4 alkyl.
  • R 34 is straight or branched C 1 –C 4 alkyl.
  • disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is , 32 wherein R is ; ; ; or .
  • the disclosed herein is a compound having the structure of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is ; ; ; ; ; ; ; ; or , wherein R 22 is C 1 –C 4 alkyl (e.g., straight or branched C 1 –C 4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 22 is straight or branched C 1 –C 4 alkyl.
  • the disclosed herein is a compound having the structure of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C selected from ; ; ; ; ; ; ; ; ;
  • Example 312 Exemplary Synthesis of 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile
  • Step 1 Preparation of tert-butyl 5-[5-(trifluoromethoxy)- 2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate
  • 2-bromo-5-(trifluoromethoxy)pyridine (2.67 g, 11.05 mmol, 1 eq)
  • tert- butyl 2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine-1-carboxyl
  • Step 2 Preparation of tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate and tert-butyl (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-1-carboxylate Racemic tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-1-carboxylate (1.9 g, 4.90 mmol, 1 eq) was purified by SFC (column:
  • Step 3 Preparation of WC-ARV-JM-047-A-2a, (3aR,7aS)-5-[5-(trifluoromethoxy)-2- pyridyl]-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine
  • tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate 500 mg, 1.29 mmol, 1 eq
  • dichloromethane 5 mL
  • trifluoroacetic acid 7.70 g, 67.53 mmol, 5.00 mL, 52.32 eq.
  • Step 4 Preparation of 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile
  • 3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a- octahydropyrrolo[3,2-c]pyridine (517 mg, 1.29 mmol, 1 eq, trifluoroacetic acid) and 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (327 mg, 1.29 mmol, 1 eq) in acetonitrile (5 mL) was added
  • compositions Disclosed herein are pharmaceutical composition comprising a compound of the invention as defined hereinbefore, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable excipients.
  • compositions of the invention may be in a form suitable for oral use (for example, as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups, or elixirs), for topical use (for example, as creams, ointments, gels, or aqueous or oily solutions, suspensions, or transdermal patches), for administration by inhalation (for example, as a finely divided powder or a liquid aerosol), for administration by insufflation (for example, as a finely divided powder), or for parenteral administration (for example, as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular, intraperitoneal, or as a suppository for rectal or intravaginal dosing).
  • oral use for example, as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible
  • compositions of the invention can be obtained by conventional procedures in view of this disclosure using conventional pharmaceutical excipients well-known in the art.
  • compositions intended for oral use may contain, for example, one or more coloring, sweetening, flavoring, and/or preservative agents.
  • compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) (e.g., d-alpha-tocopherol, polyethyleneglycol 1000 succinate), surfactants used in pharmaceutical dosage forms (e.g., tweens), polyethoxylated castor oil (e.g., CREMOPHOR surfactant (BASF)) or other similar polymeric delivery matrices, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts), colloidal silica,
  • SEDDS self-emulsifying drug delivery
  • Cyclodextrins e.g., alpha-, beta-, and gamma-cyclodextrin
  • chemically modified derivatives thereof e.g., hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-cyclodextrins
  • other solubilized derivatives also may be advantageously used to enhance delivery of compounds of the compounds described herein.
  • the amount of the compound(s) that can be administered and the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention depends on a variety of factors, including the age, weight, sex, the medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed.
  • the dosage regimen can vary widely, but may be routinely determined using standard methods.
  • the daily dose can be administered in one to four doses divided per day.
  • Other dosing schedules include one dose per week and one dose per two-day cycle.
  • Methods of Treatment The compounds of the invention as defined hereinbefore, or a pharmaceutically acceptable salt thereof, are useful for the treatment of cancer.
  • the compounds of the invention, or a pharmaceutically acceptable salt thereof can be used in the treatment of diseases or disorders associated with DGK target inhibition in T-cells.
  • the compound of the invention is prepared in combination with one or more additional therapeutic agents for conjoint administration for treating diseases or disorders associated with DGK target inhibition in T- cells.
  • the compounds described herein may be used to treat or prevent viral infections and proliferative diseases such as cancer.
  • disease or conditions that are associated with DGK target inhibition in T cells include viral and other infections (e.g., skin infections, GI infection, urinary tract infections, genito-urinary infections, systemic infections), and proliferative diseases (e.g., cancer).
  • the subject is being treated for cancer.
  • Types of cancers that may be treated with a compound of the invention include, but are not limited to, brain cancers, skin cancers, bladder cancers, ovarian cancers, breast cancers, gastric cancers, pancreatic cancers, prostate cancers, colon cancers, blood cancers, lung cancers and bone cancers.
  • cancer types include neuroblastoma, intestine carcinoma such as rectum carcinoma, colon carcinoma, familiar adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, esophageal carcinoma, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, renal carcinoma, kidney parenchymal carcinoma, ovarian carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, pancreatic carcinoma, prostate carcinoma, testis carcinoma, breast carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, Hodgkin lymphoma, non-Hodgkin lymphoma, Burkitt lymphoma, acute lymphatic leuk
  • the compounds of the invention treat colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma.
  • the compounds of the invention can inhibit activity of the diacylglycerol kinase alpha (DGK ⁇ ).
  • DGK ⁇ diacylglycerol kinase alpha
  • the compounds of the invention can be used to inhibit activity of DGK ⁇ in a cell or in a subject in need of modulation of DGK ⁇ by administering a therapeutically effective amount of a compound of the invention or a salt thereof.
  • the compounds of the invention can cause the degradation of DGK ⁇ .
  • the compounds of the invention can be used to degrade DGK ⁇ in a cell or in a subject in need of said treatment by administering a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof.
  • methods of treating diseases or conditions associated with activity or expression, including abnormal activity and/or overexpression, of DGK ⁇ in a subject by administering to the subject in need of such treatment a therapeutically effective amount or dose of any one or more compounds of the invention or a pharmaceutical composition thereof.
  • Exemplary diseases can include any disease, disorder, or condition that is directly or indirectly linked to expression or activity of the DGK ⁇ enzyme, such as over expression or abnormal activity.
  • a DGK ⁇ -associated disease can also include any disease, disorder, or condition that can be prevented, ameliorated, or cured by modulating the DGK ⁇ enzyme activity.
  • Examples of DGK ⁇ associated diseases include cancer and viral infections such as HIV infection, hepatitis B, and hepatitis C.
  • Combination Therapies the compounds of the invention may be conjointly administered with an additional anticancer, such as an immuno-oncology agent.
  • the compounds of the invention are administered concurrently with the additional anticancer agent.
  • the compounds of the invention may be co-formulated with the anticancer agent.
  • Exemplary anticancer agents include, but are not limited to, a small molecule drugs, antibodies, or other biologics or small molecules.
  • Exemplary biologics include, but are not limited to, cancer vaccines, antibodies, and cytokines.
  • the antibody is a monoclonal antibody.
  • the monoclonal antibody is humanized or human.
  • the anticancer agent is an immuno-oncology agent.
  • the immuno-oncology agent is (i) an agonist of a stimulatory (including a co- stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co-inhibitory) signal on T cells, both of which result in amplifying antigen-specific T-cell responses (often referred to as immune checkpoint regulators).
  • Certain stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF).
  • B7 family which includes B7-1, B7-2, B7-H1 (PD-L1), B7- DC (PD-L2), B7-H 2 (ICOS-L), B7-H 3 , B7-H4, B7-H5 (VISTA), and B7-H6.
  • TNF family of molecules that bind to cognate TNF receptor family members which includes CD40 and CD4OL, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4- IBB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DRS, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTOR, LIGHT, DcR3, HVEM, VEGUTL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin ⁇ /TNF ⁇ , TNFR2, TNF ⁇ , LT ⁇ R, Lymphotoxin a 1 ⁇ 2, FAS
  • T-cell responses can be stimulated by a combination of a compound of the invention and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-IBB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX4OL, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H.
  • an antagonist of a protein that inhibits T cell activation e.g., immune checkpoint
  • agents that can be combined with the compounds of the invention for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells.
  • the compounds of the invention can be combined with antagonists of KIR, such as lirilumab.
  • agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (e.g., International Patent Publication Nos.
  • WO11/70024 WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044
  • FPA-008 e.g., International Patent Publication Nos. WO11/140249; WO13169264; WO14/036357.
  • the compounds of the invention can be used with one or more of agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD-1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites.
  • agonistic agents that ligate positive costimulatory receptors e.g., blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency
  • the additional anticancer agent is a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody.
  • Suitable CTLA-4 antibodies include, for example, YERVOY (ipilimumab), or tremelimumab.
  • the additional anticancer agent is a PD-1 antagonist, such as an antagonistic PD-1 antibody.
  • Suitable PD-1 antibodies include, for example, OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; e.g., International Patent Publication No. WO 2 012/145493).
  • the immuno-oncology agent may also include pidilizumab (CT-011), though its specificity for PD-1 binding has been questioned.
  • CT-011 pidilizumab
  • Another approach to target the PD-1 receptor is the recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgG1, called AMP-224.
  • the additional anticancer agent is a PD-L1 antagonist, such as an antagonistic PD-L1 antibody.
  • Suitable PD-L1 antibodies include, for example, MPDL3280A (RG7446; e.g., International Patent Publication No.
  • the additional anticancer agent is a LAG-3 antagonist, such as an antagonistic LAG-3 antibody.
  • LAG3 antibodies include, for example, BMS-986016 (e.g., International Patent Publication Nos. WO10/19570, WO14/08218), or IMP-731 or IMP-321 (e.g., International Patent Publication Nos. WO08/132601, WO09/44273).
  • the additional anticancer agent is a CD137 (4-1BB) agonist, such as an agonistic CD137 antibody.
  • Suitable CD137 antibodies include, for example, urelumab, and PF-05082566 (e.g., International Patent Publication No. WO12/32433).
  • the additional anticancer agent is a GITR agonist, such as an agonistic GITR antibody.
  • Suitable GITR antibodies include, for example, BMS-986153, BMS-986156, TRX- 518 (e.g., International Patent Publication Nos. WO06/105021, WO09/009116), and MK- 4166 (e.g., International Patent Publication No. WO11/028683).
  • the additional anticancer agent is an IDO antagonist.
  • IDO antagonists include, for example, INCB-024360 e.g., International Patent Publication Nos. (WO 2 006/122150, WO07/75598, WO08/36653, WO08/36642), indoximod, BMS-986205, or NLG-919 (e.g., International Patent Publication No. WO09/73620, WO09/1156652, WO11/56652, WO12/142237).
  • the additional anticancer agent is an OX40 agonist, such as an agonistic OX40 antibody.
  • Suitable OX40 antibodies include, for example, MEDI-6383 or MEDI-6469.
  • the additional anticancer agent is an OX4OL antagonist, such as an antagonistic OX40 antibody.
  • OX4OL antagonists include, for example, RG-7888 (e.g., International Patent Publication No. WO06/029879).
  • the additional anticancer agent is a CD40 agonist, such as an agonistic CD40 antibody.
  • the anticancer agent is a CD40 antagonist, such as an antagonistic CD40 antibody.
  • Suitable CD40 antibodies include, for example, lucatumumab or dacetuzumab.
  • the additional anticancer agent agent is a CD27 agonist, such as an agonistic CD27 antibody.
  • Suitable CD27 antibodies include, for example, varlilumab.
  • the additional anticancer agent is MGA271 (to B7H 3 ) (e.g., International Patent Publication No. WO11/109400).
  • Combination therapies, as disclosed herein, are intended to embrace conjoint administration of these therapeutic agents; for example, administration of said therapeutic agents in a sequential manner, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents in a substantially simultaneous manner.
  • Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents.
  • Sequential or substantially simultaneous administration of each therapeutic agent can be affected by any appropriate route including, but not limited to, oral routes, parental routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues.
  • the therapeutic agents can be administered by the same route or by different routes.
  • a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally.
  • all therapeutic agents may be administered orally, or both therapeutic agents may be administered by parentally, e.g., by intravenous injection.
  • Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g., surgery or radiation treatment).
  • the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved.
  • the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.
  • One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g., IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compounds of the invention for treatment of DGK ⁇ associated diseases, disorders, or conditions.
  • the agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • Suitable additional anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine, cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
  • alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes
  • alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazene
  • suitable additional agents for use in combination with the compounds of the invention include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the “Dartmouth regimen,” which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM.
  • DTIC dacarbazine
  • BCNU carmustine
  • cisplatin the “Dartmouth regimen,” which consists of DTIC, BCNU, cisplatin and tamoxifen
  • a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM.
  • immunotherapy drugs including cytokines such as interferon alpha, interleukin 2, and tumor necrosis
  • the compounds of the invention also can be used in combination with vaccine therapy in the treatment of cancer (e.g., melanoma).
  • Antimelanoma vaccines are, in some ways, similar to the anti-virus vaccines that are used to prevent diseases caused by viruses such as polio, measles, and mumps. Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body’s immune system to destroy melanoma cells.
  • Suitable additional anti-cancer agents also include, for example, anti-metabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6- mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.
  • anti-metabolites including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • Suitable additional anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxyco-formycin, mitomycin-C, L-asparaginase, interferons (especially IFN- a), etoposide, and teniposide.
  • certain natural products and their derivatives for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins
  • vinblastine vincristine, vindesine
  • bleomycin dactinomycin
  • daunorubicin daunorubicin
  • cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, and droloxafine.
  • cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors.
  • additional anti-cancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-10 or TGF- ⁇ ).
  • additional anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4.
  • additional anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T-cell transfer.
  • Additional anti-cancer agents also include anti-cancer vaccines, such as, for example, dendritic cells, synthetic peptides, DNA vaccines, and recombinant viruses.
  • the treatment methods of the invention may optionally include conjointly administering at least one signal transduction inhibitor (STI).
  • STI signal transduction inhibitor
  • a “signal transduction inhibitor” is an agent that selectively inhibits one or more vital steps in signaling pathways, in the normal function of cancer cells, thereby leading to apoptosis.
  • Suitable STIs include, but are not limited to: (i) bcr/abl kinase inhibitors such as, for example, STI 571 (GLEEVEC®); (ii) epidermal growth factor (EGF) receptor inhibitors such as, for example, kinase inhibitors ORES SA®, SSI-774) and antibodies (Imclone: C225 [Goldstein et al. Clin.
  • her-2/neu receptor inhibitors such as farnesyl transferase inhibitors (FTI) such as, for example, L-744,832 (Kohl et al. Nat. Med., 1(8):792–97 (1995));
  • FTI farnesyl transferase inhibitors
  • inhibitors of Akt family kinases or the Akt pathway such as, for example, rapamycin
  • cell cycle kinase inhibitors such as, for example, flavopiridol and UCN-01 (see, for example, Sausville Curr. Med. Chem.
  • At least one STI and at least one compound of Formula (I) may be in separate pharmaceutical compositions.
  • at least one compound of the invention and at least one STI may be administered to the patient conjointly.
  • At least one compound of the invention may be administered first or at least one STI may be administered first and the other is administered next; or at least one compound of the invention and at least one STI may be administered at the same time. Additionally, when more than one compound of invention and/or STI is used, the compounds may be administered in any order.
  • pharmaceutical compositions for the treatment of a chronic viral infections in a subject comprising administering a therapeutically effective amount of at least one compound of the invention, optionally, at least one chemotherapeutic drug, and, optionally, at least one antiviral agent, in a pharmaceutically acceptable carrier.
  • one or more compounds of the invention, one or more chemotherapeutic drugs, and/or one or more antiviral agents are administered conjointly.
  • At least one compound of the invention may be administered first or at least one chemotherapeutic agent may be administered first.
  • at least one compound of the invention and the at least one STI may be administered at the same time.
  • the compounds may be administered in any order.
  • any antiviral agent or STI may also be administered at any point in relation to the administration of the compound of the invention.
  • Chronic viral infections that may be treated using the present combinatorial treatment include, but are not limited to, diseases caused by hepatitis C virus (HCV), human papilloma virus (HPV), cytomegalovirus (CMV), herpes simplex virus (HSV), Epstein-Barr virus (EBV), varicella zoster virus, coxsackie virus, human immunodeficiency virus (HIV).
  • HCV hepatitis C virus
  • HPV human papilloma virus
  • CMV cytomegalovirus
  • HSV herpes simplex virus
  • EBV Epstein-Barr virus
  • varicella zoster virus coxsackie virus
  • coxsackie virus human immunodeficiency virus
  • HCV hepatitis C virus
  • HCV hepatitis C virus
  • HPV human papilloma virus
  • CMV cytomegalovirus
  • HSV herpes simplex virus
  • EBV Epstein-Barr virus
  • Suitable antiviral agents contemplated for use in combination with the compound of Formula (I) can comprise nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), non- nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors and other antiviral drugs.
  • NRTIs nucleoside and nucleotide reverse transcriptase inhibitors
  • NRTIs non- nucleoside reverse transcriptase inhibitors
  • protease inhibitors and other antiviral drugs.
  • NRTIs examples include zidovudine (AZT); didanosine (ddl); zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(P0M)- PMEA]; lobucavir; BCH-I0652; emitricitabine [(-)-FTC]; beta-L-FD4 (also called beta-L- D4C and named beta-L-2′,3′-dicleoxy-5-fluoro-cytidene); DAPD, (( ⁇ )-beta-D-2,6-diamino- purine dioxolane); and lodenosine (FddA).
  • ZT zidovudine
  • ddl didanosine
  • ddC zalcitabine
  • d4T stavudine
  • lamivudine lami
  • NNRTIs include nevirapine (BI- RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721; AG-1549; MKC-442 (1-(ethoxy-methyl)-5-(1-methylethyl)-6-(phenylmethyl)-(2,4(1H,3H)- pyrimidinedione); and (+)-calanolide A (NSC-675451) and B.
  • Typical suitable protease inhibitors include saquinavir (Ro 31-8959); ritonavir (ABT-538); indinavir (MK-639); nelfnavir (AG-1343); amprenavir (141W94); lasinavir; DMP-450; BMS-2322623; ABT-378; and AG-1549.
  • Other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and Yissum Project No.11607.
  • kits useful, for example, in the treatment or prevention of DGK ⁇ -associated diseases or disorders referred to herein which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • Methods of Preparation The compounds of the present invention may be synthesized by many methods available to those skilled in the art in view of the present disclosure.
  • Step 1 Preparation of tert-butyl 4-[(2,5-difluorophenyl)methyl]-4-hydroxy- piperidine-1-carboxylate
  • isopropyl magnesium chloride (2 M, 6.72 mL, 1.43 eq) in tetrahydrofuran was added a solution of 2-bromo-1,4-difluoro-benzene (1.81 g, 9.38 mmol, 1 eq) in tetrahydrofuran, and the resulting mixture was stirred at 25°C for 6 h.
  • Step 2 Preparation of 5-fluorospiro[3H-benzofuran-2,4'-piperidine]
  • a mixture of tert-butyl 4-[(2,5-difluorophenyl)methyl]-4-hydroxy-piperidine-1-carboxylate (1 g, 3.05 mmol, 1 eq) and potassium tert-butoxide (1 g, 9.16 mmol, 3 eq) in 1,2- dimethoxyethane (15 mL) was stirred at 60 °C for 12 h. The material was collected by filtration and then treated with concentrated hydrochloric acid.
  • Step 3 Preparation of 7-(5-fluorospiro[3H-benzofuran-2,4'-piperidine]-1'-yl)-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 3)
  • the title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 5- fluorospiro[3H-benzofuran-2,4'-piperidine]. (off-white solid).
  • Step 2 Preparation of (3S,4R)-N-(4-chlorophenyl)-N,3-dimethyl-piperidin-4-amine
  • a mixture of tert-butyl (3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.30 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 45.77 eq) in dichloromethane (1 mL) was stirred at 25 °C for 0.5 h.
  • Step 3 Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 6)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- chlorophenyl)-N,3-dimethyl-piperidin-4-amine. (yellow solid).
  • the reaction mixture was stirred at 80°C for 3 h under N 2 .
  • the mixture was diluted with water (6 mL) and extracted with ethyl acetate (6 mL*2).
  • the combined organic extracts were washed with brine (12 mL), dried over sodium sulfate, and concentrated under reduced pressure.
  • Step 2 Preparation of (3R,3aR,6aR)-3-methyl-5-[4-(trifluoromethoxy)phenyl]- 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • Step 3 Preparation of 4-[(3aR,4R,6aS)-4-methyl-2-[[4- (trifluoromethoxy)phenyl]methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]- 6-chloro-1-methyl-1,5-naphthyridin-2-one
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,3aR,6aR)-3- methyl-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole and (6-chloro-1-methyl-2-oxo-1,5-naphthyridin
  • Step 1 Preparation of tert-butyl (3aS,6aR)-2-[4-(trifluoromethoxy)phenyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • tert-butyl (3aS,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate 150 mg, 0.71 mmol, 1 eq
  • 1-bromo-4-(trifluoromethoxy)benzene 255 mg, 1.06 mmol, 0.16 mL, 1.5 eq
  • dioxane 3 mL
  • Step 2 Preparation of (3aR,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • Step 3 4-[(3aS,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 8)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-[4- (trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole.
  • Step 2 Preparation of (3aR,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • Step 3 Preparation of 4-[(3aS,6aR)-3a,6a-dimethyl-2-[4-(trifluoromethoxy)phenyl]- 1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 9)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-3a,6a- dimethyl-5-[4-(trifluoromethoxy)phenyl]-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrole.
  • Step 2 Preparation of ethyl 5-[(2-cyanoacetyl)-methyl-amino]-2-methyl-thiazole-4- carboxylate
  • 2-cyanoacetic acid 3.06 g, 35.95 mmol, 1.2 eq
  • acetic anhydride 32.70 g, 320.31 mmol, 30.00 mL, 10.69 eq
  • 80 °C 0.5 h
  • Ethyl 2-methyl-5- (methylamino)thiazole-4-carboxylate (6 g, 29.96 mmol, 1 eq) was then added, and the reaction mixture was stirred at 80 °C for 0.5 h under nitrogen.
  • Step 3 Preparation of 7-hydroxy-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6- carbonitrile
  • Step 4 Preparation of (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate
  • triethylamine (1.51 g, 14.92 mmol, 2.08 mL, 3 eq)
  • 4- dimethylaminopyridine 61 mg, 0.50 mmol, 0.1 eq
  • dichloromethane (10 mL) at 0°C was added trifluoromethylsulfonyl trifluoromethanesulfonate (2.81 g, 9.94 mmol, 1.64 mL, 2 eq), and the reaction mixture was stirred at 0°C for 1 h under nitrogen.
  • Step 2 Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3R,4S)-4-(4-chloro-2-methyl-anilino)-3- methyl-piperidine-1-carboxylate
  • the enantiomers were separated by prep-SFC(column: DAICEL CHIRALCEL OD(250mm*30mm,10um); mobile phase: 20% isopropanol (0.1% NH 4 OH) in CO 2 ). Absolute configuration was arbitrarily assigned.
  • Step 3 Preparation of (3S,4R)-N-(4-chloro-2-methyl-phenyl)-3-methyl-piperidin-4- amine
  • tert-butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-piperidine-1- carboxylate 100 mg, 0.29 mmol, 1 eq
  • dichloromethane 1 mL
  • trifluoroacetic acid 770 mg, 6.75 mmol, 0.5 mL, 23 eq
  • the resulting residue was purified by prep-HPLC (column: YMC Triart C18 70*250 mm*7 ⁇ m; mobile phase: [51-81% CH 3 CN in water (NH 4 HCO 3 )]) followed by SFC (column: DAICEL CHIRALPAK AD-H (250 mm*30 mm,5 um); mobile phase: 20% isopropanol (0.1% NH 4 OH) in CO 2 ) to afford the diastereomers.
  • Step 2 Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate tert-butyl (3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate
  • tert-butyl (3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate The enantiomers were separated by SFC (column: DAICEL CHIRALCEL OD (250 mm*30 mm,10 um); mobile phase: 20% isopropanol in CO 2 ). Absolute configuration was arbitrarily assigned.
  • tert-Butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1-carboxylate (731 mg, 2.06 mmol, 46% yield) was obtained as an off-white oil.
  • tert-Butyl (3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1-carboxylate (758 mg, 2.14 mmol, 48% yield) was obtained as an off-white oil.
  • Step 3 Preparation of (3S,4R)-N-(4-chloro-2-methoxy-phenyl)-3-methyl-piperidin-4- amine
  • a mixture of tert-butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.28 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 47.93 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes.
  • Step 1 Preparation of tert-butyl (3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3- methyl-piperidine-1-carboxylate
  • a mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.1 mmol, 1 eq) , 2- amino-5-fluoro-phenol (2.68 g, 21 mmol, 1.5 eq), and acetic acid (84.47 mg, 1.41 mmol, 0.08 mL, 0.1 eq) in CH 3 OH (30 mL) was stirred at 50 °C for 0.5 h.
  • Step 2 Preparation of 5-fluoro-2-[[(3S,4R)-3-methyl-4-piperidyl]amino]phenol
  • tert-butyl (3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-piperidine-1- carboxylate 400 mg, 1.23 mmol, 1 eq
  • dichloromethane 4 mL
  • trifluoroacetic acid 3.08 g, 27 mmol, 2 mL, 22 eq
  • Step 3 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid).
  • Step 4 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 21) and 6- chloro-4-[(3R,4S)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (Compound 20)
  • the enantiomers were separated by prep-HPLC (column: DAICEL CHIRALPAK AD(250mm*30mm, 10 ⁇ m); mobile phase: [0.1% NH 4 OH in ethanol]; B%: 50%-50%, 3.2; 30 min).
  • tert-Butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3- methyl-piperidine-1-carboxylate was further purified by SFC (column: DAICEL CHIRALPAK AD-H (250mm*30mm, 5 ⁇ m); mobile phase: 20% methanol (0.1% NH 4 OH) in CO 2 ).
  • tert-Butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate was purified by SFC (column: DAICEL CHIRALPAK AD-H (250 mm*30 mm, 5 ⁇ m); mobile phase: 25% methanol (0.1% NH 4 OH) in CO 2 ). Absolute configuration was arbitrarily assigned.
  • tert-Butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate 333 mg, 0.97 mmol, 7% yield) was obtained as an off-white oil.
  • Step 2 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 25)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- fluorophenyl)-3-methoxy-piperidin-4-amine. (off-white solid).
  • Step 2 Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • the enantiomer was separated by SFC (column: DAICEL CHIRALPAK AD(250mm*30mm, 10 um); mobile phase: 50% ethanol (0.1% NH 4 OH) in CO 2 ) followed by prep-HPLC (column: Welch Ultimate C18150*25mm*5 ⁇ m; mobile phase: [32-62% CH 3 CN in water (0.225% formic acid)]) to give 6-chloro-4-[(3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3- methyl-1-piperidyl]-1-methyl-2
  • Step 2 Preparation of tert-butyl 3-methoxy-4-oxo-piperidine-1-carboxylate
  • tert-butyl 3,4,4-trimethoxypiperidine-1-carboxylate 330 mg, 1.20 mmol, 1 eq
  • 4-methylbenzenesulfonic acid 41 mg, 0.24 mmol, 0.2 eq
  • acetone 10 mL
  • water 1 mL
  • the mixture was concentrated under vacuum to give tert- butyl 3-methoxy-4-oxo-piperidine-1-carboxylate (280 mg, crude) as an off-white oil.
  • Step 3 Preparation of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1- carboxylate
  • tert-butyl 3-methoxy-4-oxo-piperidine-1-carboxylate 280 mg, 1.22 mmol, 1 eq
  • 4-fluoroaniline 135.70 mg, 1.22 mmol, 0.12 mL, 1 eq
  • borane 2-methylpyridine (261 mg, 2.44 mmol, 2 eq) in methanol (1.5 mL) and acetic acid (1.5 mL) was stirred at 25 °C for 12 h.
  • Step 4 Preparation of tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy- piperidine-1-carboxylate
  • tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1-carboxylate 90 mg, 0.28 mmol, 1 eq
  • formaldehyde 68 mg, 0.83 mmol, 0.06 mL, 37% purity, 3 eq
  • sodium cyanoborohydride 52 mg, 0.83 mmol, 3 eq
  • Step 5 Preparation of (3S,4R)-N-(4-fluorophenyl)-3-methoxy-N-methyl-piperidin-4- amine
  • Step 6 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • Compound 31 A mixture of (3S,4R)-N-(4-fluorophenyl)-3-methoxy-N-methyl-piperidin-4-amine (100 mg, 0.28 mmol, 1 eq, TFA salt), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (50 mg, 196.79 ⁇ mol, 6.93e-1 eq), and diisopropylethylamine (371.00 mg, 2.87 mmol, 0.5 mL, 10.11 eq) in acetonitrile (1 mL) was stirred at 25 °C for 12 h.
  • Step 2 Preparation of tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy) anilino]piperidine-1-carboxylate.
  • Step 3 Preparation of (3S,4R)-N,3-dimethyl-N-[4- (trifluoromethoxy)phenyl]piperidin-4-amine.
  • a mixture of tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate (99 mg, 0.26 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 52.99 eq) in dichloromethane (1 mL) was stirred at 25 °C for 15 minutes.
  • Step 4 Preparation of 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 32)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N,3- dimethyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (yellow solid).
  • Step 1 Preparation of tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate and tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate
  • tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate 2 g, 9.38 mmol, 1 eq
  • 4-chloroaniline (1.20 g, 9.38 mmol, 1 eq) in methanol (20 mL)
  • sodium cyanoborohydride (1.47 g, 23.44 mmol, 2.5 eq
  • Step 2 Preparation of (3S,4R)-N-(4-chlorophenyl)-3-methyl-piperidin-4-amine
  • dichloromethane 1 mL
  • trifluoroacetic acid 1.54 g, 13.51 mmol, 1 mL, 43.87 eq
  • Step 3 Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile.
  • Step 4 Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 33) and 6- chloro-4-[(3R,4S)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 34)
  • the enantiomers were separated by SFC (column: DAICEL CHIRALCEL OJ(250mm*30mm,10 ⁇ m); mobile phase: 40% methanol (0.1%NH 4 OH) in CO 2 ) followed by prep-HPLC (column: Phenomenex luna C18150*25mm* 10 ⁇ m; mobile phase: [50-80% CH 3 CN in water (0.225% formic acid)]).
  • Step 2 Preparation of (3R,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine
  • Step 3 Preparation of 6-chloro-4-[4-(4-chloro-N-methyl-anilino)-1-piperidyl]-2-oxo- 1H-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from N-(4- chlorophenyl)-N-methyl-piperidin-4-amine. (yellow solid).
  • Step 2 Preparation of tert-butyl (3S,4R)-4-(4-fluoro-N,2-dimethyl-anilino)-3-methyl- piperidine-1-carboxylate
  • sodium cyanoborohydride 88 mg, 1.40 mmol, 3 eq
  • formaldehyde 189 mg, 2.33 mmol, 0.17 mL, 37% purity, 5 eq
  • acetic acid 105 mg, 1.75 mmol, 0.1 mL, 3.76 eq
  • Step 3 Preparation of (3S,4R)-N-(4-fluoro-2-methyl-phenyl)-N,3-dimethyl-piperidin- 4-amine
  • a mixture of tert-butyl (3S,4R)-4-(4-fluoro-N,2-dimethyl-anilino)-3-methyl-piperidine-1- carboxylate (90 mg, 0.27 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1.00 mL, 50.49 eq) in dichloromethane (1 mL) was stirred at 25 °C for 30 min under N 2 atmosphere.
  • Step 4 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N,2-dimethyl-anilino)-3- methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 37)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- fluoro-2-methyl-phenyl)-N,3-dimethyl-piperidin-4-amine.
  • Step 1 Preparation of O2-benzyl O5-tert-butyl (3aR,6aR)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-2,5-dicarboxylate
  • tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate 500 mg, 2.36 mmol, 1 eq
  • triethylamine 715 mg, 7.07 mmol, 0.98 mL, 3 eq
  • dichloromethane 10 mL
  • benzyl chloroformate 482 mg, 2.83 mmol, 0.4 mL, 1.2 eq
  • Step 2 Preparation of benzyl (3aS,6aS)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole-5-carboxylate
  • O 2 -benzyl O5-tert-butyl (3aR,6aR)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-2,5-dicarboxylate 95 mg, 0.27 ⁇ mol, 1 eq
  • dichloromethane (1 mL)
  • trifluoroacetic acid 770 mg, 6.75 mmol, 0.5 mL, 24.6 eq
  • Step 3 Preparation of benzyl (3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • Step 4 Preparation of (3aS,6aS)-5-(4-fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • benzyl (3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate 200 mg, 564.31 umol, 1 eq
  • CH 3 OH 5 mL
  • activated carbon catalyst 50 mg, 10% purity
  • Step 5 Preparation of 4-[(3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 38)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aS,6aS)-5-(4- fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole.
  • Step 1 Preparation of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl]piperazine-1- carboxylate
  • a mixture of tert-butyl piperazine-1-carboxylate (357 mg, 1.92 mmol, 1 eq), 4-chloro-2- hydroxy-benzaldehyde (300 mg, 1.92 mmol, 1 eq), and sodium cyanoborohydride (241 mg, 3.83 mmol, 2 eq) in methanol (3 mL) was stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the resulting crude product was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2).
  • Step 2 Preparation of 5-chloro-2-(piperazin-1-ylmethyl)phenol
  • a mixture of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl]piperazine-1-carboxylate (20 mg, 0.06 mmol, 1 eq) and trifluoroacetic acid (154 mg, 1.35 mmol, 0.1 mL, 22.07 eq) in dichloromethane (0.2 mL) was stirred at 20 °C for 0.5 h. The mixture was concentrated to give 5-chloro-2-(piperazin-1-ylmethyl)phenol (21 mg, crude, TFSA salt) as a yellow oil.
  • Step 3 Preparation of 7- ⁇ 4-[(4-chloro-2-hydroxyphenyl)methyl]piperazin-1-yl ⁇ -2,4- dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 39)
  • the title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 5- chloro-2-(piperazin-1-ylmethyl)phenol. (white solid).
  • Step 2 Preparation of (3aR,6aS)-5-(4-fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • 3aS,6aR tert-butyl-3aS,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (110 mg, 0.34 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 1 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes.
  • Step 3 Preparation of 6-chloro-4-((3aR,6aS)-5-(4-fluoro-2- methylphenyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile (Compound 40)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-(4- fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole.
  • Step 2 Preparation of benzyl 1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate
  • Step 4 Preparation of 1-(4-fluorophenyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2- c]pyridine
  • benzyl 1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate 108 mg, 0.31 mmol, 1 eq
  • CH 3 OH 3 mL
  • Step 5 Preparation of 4-[(3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 41)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from 1-(4- fluorophenyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine.
  • Step 1 Preparation of tert-butyl 4-(4-chloroanilino)piperidine-1-carboxylate
  • Step 2 Preparation of N-(4-chlorophenyl)piperidin-4-amine
  • a mixture of tert-butyl 4-(4-chloroanilino)piperidine-1-carboxylate (62 mg, 0.20 mmol, 1 eq) and trifluoroacetic acid (924 mg, 8.10 mmol, 0.6 mL, 83.96 eq) in dichloromethane (0.6 mL) was stirred at 25 °C for 15 minutes. The mixture was concentrated under vacuum to give N- (4-chlorophenyl)piperidin-4-amine (100 mg, crude, trifluoroacetic acid) as a red oil.
  • Step 3 Preparation of 6-chloro-4-[4-(4-chloroanilino)-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile
  • Step 1 Preparation of ethyl 3-acetamido-6-chloro-pyridine-2-carboxylate
  • Step 2 Preparation of ethyl 3-[acetyl(methyl)amino]-6-chloro-pyridine-2-carboxylate
  • Step 3 Preparation of 6-chloro-4-hydroxy-1-methyl-1,5-naphthyridin-2-one
  • ethyl 3-[acetyl(methyl)amino]-6-chloro-pyridine-2-carboxylate 110 mg, 0.43 mmol, 1 eq
  • tetrahydrofuran 1 mL
  • potassium bis(trimethylsilyl)amide 1 M, 0.64 mL, 1.5 eq
  • Step 5 Preparation of tert-butyl 2-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • 1-bromo-4-fluoro-benzene 185 mg, 1.06 mmol, 0.12 mL, 1.2 eq
  • Cs 2 CO 3 (864 mg, 2.65 mmol, 3 eq)
  • Step 6 Preparation of (3S,3aS,6aS)-5-(4-fluorophenyl)-3-methyl-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • Step 7 Preparation of 4-[(3aS,4S,6aR)-2-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-1,5-naphthyridin-2-one
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,3aS,6aS)-5- (4-fluorophenyl)-3-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole and (6-chloro- 1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulf
  • the mixture was diluted with water (40 mL) and extracted with ethyl acetate (40 mL*2). The combined organic extracts were washed with brine (100 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate). To the product was added trifluoroacetic acid (2 mL), and the resulting mixture was stirred at 20°C for 1 h.
  • Step 2 Preparation of 6-chloro-4-((3aR,6aS)-5-(4- fluorophenyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile (Compound 45)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-(4- fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole.
  • Step 2 Preparation of N,N-bis(4-fluorophenyl)piperidin-4-amine
  • tert-butyl 4-(4-fluoro-N-(4-fluorophenyl)anilino)piperidine-1-carboxylate 70 mg, 0.18 mmol, 1 eq
  • dichloromethane 1 mL
  • trifluoroacetic acid 1.20 g, 10.50 mmol, 0.78 mL, 58.29 eq
  • Step 3 Preparation of 6-chloro-4-[4-(4-fluoro-N-(4-fluorophenyl)anilino)-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from N,N-bis(4- fluorophenyl)piperidin-4-amine. (yellow solid).
  • Step 1 Preparation of tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate 48 mg, 0.23 mmol, 1.1 eq
  • Cs 2 CO 3 203 mg, 0.62 mmol, 3 eq
  • Step 2 Preparation of (3aS,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate 110 mg, 0.30 mmol, 1 eq
  • dichloromethane (1 mL)
  • trifluoroacetic acid (1.69 g, 14.86 mmol, 1.1 mL, 50.29 eq
  • Step 3 Preparation of 4-[(3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile
  • Step 1 Preparation of tert-butyl (3aR,6aS)-2-(4-fluoro-2-methyl-phenyl)-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • tert-butyl (3aR,6aS)-2-(4-fluoro-2-methyl-phenyl)-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate 343 mg, 1.43 mmol, 0.9 eq) in dioxane (6 mL) was added K2CO 3 (658 mg, 4.76 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1
  • Step 3 Preparation of 4-[(3aS,6aR)-2-(4-fluoro-2-methyl-phenyl)-3a,6a-dimethyl- 1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 49)
  • Compound 49 The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aS,6aR)-5-(4- fluoro-2-methyl-phenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrole.
  • the mixture was partitioned between ethyl acetate (100 mL) and water (100 mL). The layers were separated, and the aqueous phase was further extracted with ethyl acetate (100 mL x 2). The combined organic extract was dried over sodium sulfate and concentrated.
  • Step 2 Preparation of tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1- carboxylate
  • tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-hydroxy-piperidine-1-carboxylate 400 mg, 1.29 mmol, 1 eq
  • sodium hydride 129 mg, 3.22 mmol, 60% purity, 2.5 eq
  • Step 3 Preparation of (3R,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine
  • Step 4 Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 50)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,4R)-N-(4- fluorophenyl)-3-methoxy-piperidin-4-amine. (yellow solid).
  • Step 2 Preparation of (3R,4R)-N-(4-fluorophenyl)-3-methoxy-N-methylpiperidin-4- amine
  • tert-butyl (3R,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy-piperidine-1- carboxylate 120 mg, 0.35 mmol, 1 eq
  • dichloromethane 1 mL
  • trifluoroacetic acid (1.85 g, 16.21 mmol, 1.20 mL, 45.71 eq
  • Step 2 Preparation of benzyl (2S,5R)-2,5-dimethyl-4-oxo-piperidine-1-carboxylate
  • dichloromethane 10 mL
  • hydrogen chloride 4 M, 10 mL, 4.33 eq
  • This material was further purified by SFC [method: (column: DAICEL CHIRALPAK AY-H(250mm * 30mm,10um); mobile phase: 15% CH 3 OH in CO 2 ] to afford benzyl (2S,5R)-2,5-dimethyl-4-oxo-piperidine-1-carboxylate (350 mg, 1.34 mmol, 58% yield) as a yellow oil.
  • Step 3 Preparation of benzyl (2S,5R)-4-(4-fluoroanilino)-2,5-dimethyl-piperidine-1- carboxylate
  • benzyl (2S,5R)-2,5-dimethyl-4-oxo-piperidine-1-carboxylate 100 mg, 0.38 mmol, 1.00 eq
  • 4-fluoroaniline 63 mg, 0.57 mmol, 1.50 eq
  • sodium cyanoborohydride 48 mg, 0.76 mmol, 2.00 eq
  • acetic acid 2 mg, 38.27 umol, 0.10 eq
  • the reaction mixture was diluted with water 10 mL and extracted with ethyl acetate (10 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by semi-preparative reverse phase HPLC (70-80% acetonitrile + 0.225% formic acid in water, 7 min) to afford benzyl (2S,5R)- 4-(4-fluoroanilino)-2,5-dimethyl-piperidine-1-carboxylate (60 mg, 0.16 mmol, 43% yield) as a colorless oil.
  • Step 4 Preparation of benzyl (2S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl- piperidine-1-carboxylate
  • sodium cyanoborohydride 15 mg, 0.25 mmol, 1.50 eq
  • formaldehyde 136 mg, 1.68 mmol, 37% purity in water, 10.00 eq
  • Step 5 Preparation of (2S,5R)-N-(4-fluorophenyl)-N,2,5-trimethyl-piperidin-4-amine
  • benzyl (2S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-piperidine-1- carboxylate 62 mg, 0.16 mmol, 1.00 eq
  • methanol 4 mL
  • palladium on activated carbon catalyst (20 mg, 10% purity
  • the suspension was degassed and purged with hydrogen (3X), and the reaction mixture was stirred under hydrogen (15 Psi) at 25 °C for 2 h.
  • Step 1 Preparation of (3aR,6aR)-5-benzyl-3-methyl-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole
  • a mixture of tert-butyl (3aR,6aS)-2-benzyl-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (200 mg, 0.63 mmol, 1 eq) and HCl/dioxane (4 M, 1 mL, 6.33 eq) in dichloromethane (1 mL) was stirred at 20 °C for 1 h.
  • Step 2 Preparation of (3aR,6aS)-2-benzyl-5-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole
  • 3aR,6aR -5-benzyl-3-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole
  • 1-bromo-4-fluoro-benzene 122 mg, 0.69 mmol, 0.076 mL, 1.1 eq
  • Cs 2 CO 3 (619 mg, 1.90 mmol, 3 eq) in dioxane (3 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyr
  • Step 3 Preparation of (3aR,6aS)-5-(4-fluorophenyl)-4-methyl-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
  • the suspension was degassed under vacuum and purged with H 2 several times.
  • Step 4 Preparation of 4-[(3aR,6aS)-5-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile
  • Step 1 Preparation of tert-butyl (1R,3aS,6aR)-5-(6-chloro-3-cyano-1-methyl-2-oxo- 1,2-dihydro-1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate
  • tert-butyl (3aR,6aS)-4-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c] pyrrole-5-carboxylate (0.10 g, 0.44 mmol, 1 eq)
  • 4,6-dichloro-1-methyl-2-oxo -1,5- naphthyridine-3-carbonitrile (112 mg, 0.44 mmol, 1 eq) in acetonitrile (3 mL) was added diisopropylethylamine (171 mg, 1.33 m
  • Step 2 Preparation of 6-chloro-1-methyl-4-((3aR,4R,6aR)-4- methylhexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile
  • tert-butyl (1R,3aS,6aR)-5-(6-chloro-3-cyano-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate 50 mg, 0.11 mmol, 1 eq
  • dichloromethane 2 mL
  • trifluoroacetic acid 1 mL
  • Step 3 Preparation of 4-[(3aR,4R,6aR)-5-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile
  • 6-chloro-1-methyl-4-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4-c]pyrrol- 2(1H)-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile 50 mg, 0.11 mmol, 1 eq, trifluoroacetate
  • 4-chloro-2-hydroxy-benzaldehyde 34 mg, 0.22 mmol, 2 eq
  • Step 4 Preparation of 4-[(3aR,4R,6aR)-5-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 55) and 4-[(3aS,4S,6aS)-5 -[(4-chloro- 2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2- yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 56) The mixture of enantiomers (40 mg, 0.08
  • the second peak was purified by semi-preparative reverse phase (column: Unisil 3-100 C18 Ultra 150*50mm*3 um; mobile phase: [5-35% CH 3 CN in water (0.225% formic acid)]) to give 4-[(3aS,4S,6aS)-5-[(4-chloro-2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a - hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (9.6 mg, 0.02 mmol, 23% yield) (Compound 56) as a white solid.
  • Step 1 Preparation of tert-butyl (1S,3aR,6aS)-5-(4-chloro-2-hydroxybenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
  • tert-butyl (1R,3aS,6aR)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate (0.50 g, 2.21 mmol, 1 eq) and 4-chloro-2-hydroxy-benzaldehyde (371 mg, 2.37 mmol, 1.07 eq) in 1,2-dichloroethane (10 mL) were added sodium triacetoxyborohydride (1.40 g, 6.63 mmol, 3 eq) and triethylamine (447 mg, 4.42 mmol, 2 eq), and the reaction mixture was stirred at 60 °C for 12 h.
  • Step 2 Preparation of 5-chloro-2-(((3aS,4S,6aS)-4-methylhexahydropyrrolo[3,4- c]pyrrol-2(1H)-yl)methyl)phenol
  • tert-butyl (1S,3aR,6aS)-5-(4-chloro-2-hydroxybenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (0.30 g, 0.81 mmol, 1 eq) in dichloromethane (6 mL) was added trifluoroacetic acid (3 mL), and the reaction mixture was stirred at 25 °C for 0.5 h.
  • Step 3 Preparation of 4-[(3aS,4S,6aR)-2-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile
  • 4-6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (0.10 g, 0.39 mmol, 1 e
  • Step 4 Preparation of 4-[(3aS,4S,6aR)-2-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 57) and 4-[(3aR,4R,6aS)-2 -[(4-chloro- 2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5- yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 58) The mixture of enantiomers (0.15 g, 0.30 mmol, 1 eq) was separated by supercritical fluid chromatography (col
  • the second peak was purified further by semi-preparative reverse phase (column: Phenomenex C1875*30mm*3 ⁇ m; mobile phase: [12-42% CH 3 CN in water (formic acid)]) to give 4-[(3aR,4R,6aS)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a - hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (30.9 mg, 0.06 mmol, 77% yield) (Compound 58) as an off-white solid.
  • the reaction mixture was concentrated under reduced pressure.
  • the product was purified by prep-HPLC (column: Waters Xbridge 150*25 mm* 5 ⁇ m; mobile phase: [35-65% CH 3 CN in water (NH 4 HCO 3 )]) followed by lyophilization to afford 8-[(3aR,6aR)-2-[(4- chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (36.1 mg, 82.82 ⁇ mol, 37% yield) as a white solid.
  • the reaction mixture was neutralized by addition of 8 N aqueous NaOH (120 mL) at 0 °C, and the resulting mixture was extracted with ethyl acetate (100 mL ⁇ 3). The organic extract was dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure.
  • the crude product was purified by flash silica gel chromatography (gradient: of 0 ⁇ 10% ethyl acetate in petroleum ether) to give 1-benzyl-4-(2-bromoanilino) piperidine-4-carbonitrile (10.0 g, 27.0 mmol, 51% yield) as a white solid.
  • Step 2 Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]-3-imine
  • 1-benzyl-4-(2-bromoanilino)piperidine-4-carbonitrile 7.4 g, 19.98 mmol, 1 eq
  • AIBN 328 mg, 2.00 mmol, 0.1 eq
  • Bu3SnH 6.4 g, 21.98 mmol, 5.8 mL, 1.1 eq
  • Step 3 Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]-3-one
  • Step 4 Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]-3-ol
  • 1'-benzylspiro[indoline-2,4'-piperidine]-3-one 500 mg, 1.71 mmol, 1 eq
  • THF 5 mL
  • LiAlH4 1 M, 1.8 mL, 1.1 eq
  • the reaction mixture was stirred at 25°C for 1 hour.
  • the reaction quenched by sequential addition of water (0.3 mL), 15% aq. NaOH (0.3 mL), and water (0.6 mL).
  • the resulting suspension was dried over anhydrous Na 2 SO 4 and then filtered.
  • Step 5 Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]
  • a solution of 1'-benzylspiro[indoline-2,4'-piperidine]-3-ol (500 mg, 1.70 mmol, 1 eq) and Et 3 SiH (987 mg, 8.49 mmol, 1.3 mL, 5 eq) in CH 2 Cl 2 (10 mL) was treated with TFA (1.9 g, 16.9 mmol, 1.3 mL, 10 eq) at 25°C for 1 hour.
  • Step 6 Preparation of 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine]
  • 1'-benzylspiro[indoline-2,4'-piperidine] 450 mg, 1.62 mmol, 1 eq
  • formaldehyde 655 mg, 8.08 mmol, 601 ⁇ L, 37% purity, 5 eq
  • CH 3 OH 10 mL
  • AcOH 970 mg, 16.1 mmol, 924 ⁇ L, 10 eq
  • borane 2-methylpyridine 864 mg, 8.08 mmol, 5 eq
  • Step 7 Preparation of spiro[indoline-2,4'-piperidine]
  • a solution of 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine] 150 mg, 0.513 mmol, 1 eq
  • CH 3 OH 5 mL
  • Pd(OH) 2 62 mg, 0.442 mmol, 20% purity, 0.8 eq
  • AcOH 61 mg, 1.03 mmol, 58 ⁇ L, 2 eq
  • Step 8 Preparation of 6-chloro-1-methyl-4-(1-methylspiro[indoline-2,4'-piperidine]- 1'-yl)-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 62)
  • 4-6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile 87 mg, 0.346 mol, 1 eq
  • 1-methylspiro[indoline-2,4'-piperidine] 70 mg, 0.346 mmol, 1 eq
  • diisopropylethylamine 223 mg, 1.73 mmol, 305 ⁇ L, 5 eq
  • reaction mixture was cooled to room temperature and partitioned between ethyl acetate (200 mL) and water (30 mL). The organic layer was washed with brine (30 mL), dried over sodium sulfate, filtered, and concentrated.
  • Step 2 Preparation of 5-(4-fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole
  • Step 3 Preparation of 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 64)
  • Step 2 Preparation of 6-chloro-4-[4-(4-chloro-2-hydroxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • 6-chloro-4-[4-(4-chloro-2-hydroxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a solution of 5-chloro-2-[(3-methyl-4-piperidyl)amino]phenol (100 mg, 0.28 mmol, 1.00 eq, trifluoroacetate) in acetonitrile (5 mL) were added 4,6-dichloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (71 mg, 0.28 mmol, 1.00 eq) and N,N-diisopropylethylamine
  • Step 3 Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 65), 6- chloro-4-[(3R,4S)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (Compound 66), 6-chloro-4-[(3R,4R)-4-(4- chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile (Compound 67), and 6-chloro-4-[(3S,4S)-4-(4-chloro-2-hydroxy-anilino)-3-
  • Step 2 Preparation of 6-chloro-1-methyl-2-oxo-4-(3-oxospiro[indoline-2,4'- piperidine]-1'-yl)-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile starting from spiro[indoline-2,4'-piperidine]-3-one.
  • LC/MS (ESI) m/z: 419.9 [M+H] + .
  • Step 1 Preparation of tert-butyl (3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate and tert-butyl (3aS,6aS)-2-(4- fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • SFC columnumn: DAICEL CHIRALCEL OJ(250mm*30mm,10um); mobile phase: 10% CH 3 OH (0.1% NH 4 OH) in CO 2 ).
  • Step 2 Preparation of 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 69) and 4-[(3aS,6aS)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 43)
  • the title compounds were prepared in an analogous manner to 4-[(3aR,6aR)-2-(4- fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5
  • Step 2 Preparation of ethyl 6-chloro-3-[(2-cyanoacetyl)amino] pyridine-2- carboxylate
  • ethyl 3-amino-6-chloro-pyridine-2-carboxylate 8.00 g, 33.7 mmol, 1 eq, HCl
  • DMF 50 mL
  • triethylamine 17.1 g, 169 mmol, 5 eq
  • 2-cyanoacetic acid (8.61 g, 101 mmol, 3 eq)
  • 1-propanephosphonic anhydride (34.4 g, 54.0 mmol, 50% purity, 1.6 eq
  • Step 3 Preparation of 6-chloro-4-hydroxy-2-oxo-1H-1,5 –naphthyridine-3- carbonitrile
  • ethyl 6-chloro-3-[(2-cyanoacetyl)amino]pyridine-2-carboxylate 8.71 g, 32.5 mmol, 1 eq
  • THF 90 mL
  • potassium bis(trimethylsilyl)amide 1 M, 39.05 mL, 1.2 eq
  • the reaction mixture was stirred at -78°C under N 2 for 1 hour.
  • Step 4 Preparation of 6-chloro-4-hydroxy-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile
  • 6-chloro-4-hydroxy-2-oxo-1H-1,5-naphthyridine-3-carbonitrile 5.33 g, 20.9 mmol, 87% purity, 1 eq
  • DMF 80 mL
  • NaH 4.81 g, 120 mmol, 60% purity, 5.74 eq
  • Step 5 Preparation of 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a solution of 6-chloro-4-hydroxy-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (5.14 g, 21.8 mmol, 1 eq) and N,N-diisopropylethylamine (16.9 g, 131 mmol, 6 eq) in CH 3 CN (100 mL) were added benzyl(triethyl)ammonium chloride (9.94 g, 43.6 mmol, 2 eq) and POCl3 (16.7 g, 109 mmol, 5 eq) at 0 °C,
  • Step 6 Preparation of 6-chloro-1-methyl-2-oxo-4-spiro[indane-2,4'-piperidine]-1'-yl- 1,5-naphthyridine-3-carbonitrile
  • Step 2 Preparation of 5-[(4-chlorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole
  • a solution of tert-butyl 2-[(4-chlorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate 0.1 g, 0.3 mol, 1 eq
  • methanol 5 methanol
  • hydrochloric acid/dioxane 4 M, 10 mL
  • Step 3 Preparation of 6-chloro-4-[2-[(4-chlorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 72)
  • 5-[(4-chlorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole (81 mg, 0.3 mmol, 1.51 eq, hydrochloride) and N,N-diisopropylethylamine (152 mg, 1.18 mmol, 0.2 mL, 6.00 eq) in dichloromethane (10 mL) and DMF (1 mL) was added 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (
  • Step 2 Preparation of tert-butyl 2-[tert-butyl(dimethyl)silyl]oxypyrrole-1-carboxylate
  • tert-butyl 5-oxo-2H-pyrrole-1-carboxylate 25.00 g, 136.46 mmol, 1 eq
  • dichloromethane 500 mL
  • triethylamine 41.42 g, 409.38 mmol, 56.98 mL, 3.0 eq
  • [tert-butyl(dimethyl)silyl] trifluoromethanesulfonate 43.29 g, 163.75 mmol, 1.2 eq
  • Step 3 Preparation of tert-butyl 2-methyl-5-oxo-2H-pyrrole-1-carboxylate
  • tert-butyl 2-[tert-butyl(dimethyl)silyl]oxypyrrole-1-carboxylate 32.00 g, 107.58 mmol, 1 eq
  • dichloromethane 400 mL
  • iodomethane 45.81 g, 322.73 mmol, 20.09 mL, 3.0 eq
  • Step 4 Preparation of tert-butyl (1R,3aS,6aR)-5-benzyl-1-methyl-3- oxohexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
  • tert-butyl 2-methyl-5-oxo-2H-pyrrole-1-carboxylate 5.30 g, 26.87 mmol, 1 eq
  • trifluoroacetic acid (1.53 g, 13.44 mmol, 0.5 eq) in dichloromethane (100 mL) under nitrogen 0 °C
  • N-(methoxymethyl)-1-phenyl-N- (trimethylsilylmethyl)methanamine (10.13 g, 36.28 mmol, 85% purity, 1.35 eq) in dichloromethane (20 mL) dropwise, and the reaction mixture was stirred at 20 °C for 15 h.
  • Step 5 Preparation of tert-butyl (1R,3aS,6aR)-5-benzyl-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
  • tert-butyl (1R,3aS,6aR)-5-benzyl-1-methyl-3-oxohexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate (8.00 g, 24.21 mmol, 1 eq) in tetrahydrofuran (50 mL) under nitrogen at 0°C was added borane dimethyl sulfide complex solution (10 M, 6.0 mL, 2.48 eq) dropwise, and the reaction mixture was stirred at 90 °C for 0.5 h.
  • Step 6 Preparation of tert-butyl (1R,3aS,6aR)-1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate
  • a solution of tert-butyl (1R,3aS,6aR)-5-benzyl-1-methylhexahydropyrrolo[3,4-c]pyrrole- 2(1H)-carboxylate (2.00 g, 6.32 mmol, 1 eq) in methanol (30 mL) was added palladium on activated carbon (0.20 g, 10% purity) under nitrogen, and the suspension was degassed under vacuum and purged with hydrogen several times.
  • Step 7 Preparation of tert-butyl (1S,3aR,6aS)-5-(4-fluorobenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
  • 4-fluorobenzaldehyde 219 mg, 1.77 mmol, 2 eq
  • tert-butyl (1R,3aS,6aR)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate 200 mg, 0.88 mmol, 1 eq
  • triethylamine 268 mg, 2.65 mmol, 0.37 mL, 3 eq
  • 1,2-dichloroethane 20 mL
  • Step 8 Preparation of (1S,3aS,6aS)-5-(4-fluorobenzyl)-1- methyloctahydropyrrolo[3,4-c]pyrrole
  • a mixture of tert-butyl (1S,3aR,6aS)-5-(4-fluorobenzyl)-1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate 120 mg, 0.36 mmol, 1 eq
  • dichloromethane 10 mL
  • trifluoroacetic acid 456 mg, 4.00 mmol, 2 mL, 11.15 eq
  • Step 9 Preparation of 8-[(3aS,4S,6aR)-2-[(4-fluorophenyl)methyl] -4-methyl- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile
  • 6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl trifluoromethanesulfonate
  • (1S,3aS,6aS)-5-(4-fluorobenzyl)-1- methyloctahydropyrrolo[3,4-c]pyrrole 120 mg, 0.34 mmol, 1 eq, trifluoroacetate
  • Step 10 Preparation of 8-[(3aS,4S,6aR)-2-[(4-fluorophenyl)methyl] -4-methyl- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (Compound 74) and 8-[(3aR,4R,6aS)-2-[(4- fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 75) The mixture of enantiomers (100 mg, 0.24 mmol, 1 eq) was separated by supercritical fluid
  • Step 1 Preparation of tert-butyl (1R,3aS,6aR)-5-(6-cyano-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
  • tert-butyl (1R,3aS,6aR) -1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate
  • diisopropylethylamine 343 mg, 2.65 mmol, 3
  • Step 2 Preparation of 5-methyl-8-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4- c]pyrrol-2(1H)-yl)-6-oxo-5,6-dihydro-1,5-naphthyridine-2-carbonitrile racemic
  • Step 3 Preparation of 8-[(3aR,4R,6aR)-5-[(4-fluorophenyl)methyl] -4-methyl- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile
  • 4-fluorobenzaldehyde 106 mg, 0.85 mmol, 2 eq
  • triethylamine (215 mg, 2.13 mmol, 5 eq)
  • 5-methyl-8-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4-c]pyrrol- 2(1H)-yl)-6-oxo-5,6-dihydro-1,5-naphthyridine-2-carbonitrile 180 mg, 0.43 mmol, 1 eq, trifluoroacetate
  • Step 4 Preparation of WC-ARV-JM-004-E, 8-[(3aR,4R,6aR)-5-[(4- fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 76) and 8-[(3aS,4S,6aS)- 5-[(4-fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 70) The mixture of enantiomers (150 mg, 0.
  • Step 2 Preparation of tert-butyl 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole- 5-carboxylate
  • tert-butyl 2-benzyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate 150.00 mg, 0.5 mmol, 1 eq
  • ethanol 10 mL
  • palladium on activated carbon catalyst 0.05 g, 10% purity
  • Step 3 Preparation of tert-butyl 2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • Step 4 Preparation of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5- ylmethyl)-5-chloro-phenol
  • dichloromethane 5 mL
  • hydrochloric acid/methanol 4 M, 10 mL
  • Step 5 Preparation of 4-[(3aR,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile
  • Compound 78 A solution of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-ylmethyl)-5-chloro- phenol (33 mg, 0.11 mmol, 1 eq, hydrochloride), triethylamine (57 mg, 0.57 mmol, 0.08 mL, 5.0 eq), and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (29 mg, 0.11 mmol, 1 eq) in dichloromethane (5
  • Step 2 Preparation of N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine
  • dichloromethane 5 mL
  • trifluoroacetic acid 17 mg, 0.15 mmol, 0.01 mL, 1 eq
  • Step 3 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6-chloro-4-[(3R,4R)- 4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile To a solution of N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine (50 mg, 148.67 ⁇ mol, 1 eq, trifluoro
  • Step 1 Preparation of (3S,4R)-N-(4-fluorophenyl)-3-methyl-piperidin-4-amine
  • dichloromethane 5 mL
  • trifluoroacetic acid 36.97 mg, 324.26 ⁇ mol, 24.01 ⁇ L, 1 eq
  • Step 2 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 81)
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- fluorophenyl)-3-methyl-piperidin-4-amine.
  • Step 2 Preparation of 5-chloro-2-[[methyl(4-piperidyl)amino]methyl]phenol
  • a mixture of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl-methyl-amino]piperidine-1- carboxylate (120 mg, 0.338 mmol, 1.0 eq) in dichloromethane (1 mL) was added HCl/dioxane (4 M, 1 mL, 11.8 eq) in one portion at 25°C. The mixture was stirred at 25°C for 1 hour.
  • Step 3 Preparation of 6-chloro-4-[4-[(4-chloro-2-hydroxy-phenyl)methyl-methyl- amino]-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 83)
  • 5-chloro-2-[[methyl(4-piperidyl)amino]methyl]phenol 95 mg, 0.374 mmol, 1.0 eq
  • 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile 95 mg, 0.374 mmol, 1.0 eq
  • CH 3 CN 4 mL
  • Step 2 Preparation of N-(4-fluorophenyl)-N-methyl-piperidin-4-amine
  • HCl/dioxane 4 M, 1 mL, 12.3 eq
  • Step 3 Preparation of 6-chloro-4-[4-(4-fluoro-N-methyl-anilino)-1-piperidyl]-1- methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile (Compound 84)
  • N-(4-fluorophenyl)-N-methyl-piperidin-4-amine 67 mg, 0.322 mmol, 1.0 eq
  • 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile 82 mg, 0.322 mmol, 1.0 eq
  • CH 3 CN diisopropylamine (1.61 mmol, 280 ⁇ L, 5.0 eq)
  • Step 2 Preparation of 6-chloro-1-methyl-2-oxo-4-spiro[indoline-2,4'-piperidine]-1'- yl-1,5-naphthyridine-3-carbonitrile
  • Compound 85 The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile starting from spiro[indoline-2,4'-piperidine]. (yellow solid). LC/MS (ESI) m/z: 406.3 [M+H] + .
  • Step 2 Preparation of 1-methylspiro[indoline-2,4'-piperidine]-3-one
  • 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine]-3-one 50 mg, 0.163 mmol, 1 eq
  • dichloroethane 2 mL
  • 1-chloroethyl carbonochloridate 122 mg, 0.855 mmol, 5.24 eq
  • Step 3 Preparation of 6-chloro-1-methyl-4-(1-methyl-3-oxo-spiro[indoline-2,4'- piperidine]-1'-yl)-2-oxo-1,5-naphthyridine-3-carbonitrile
  • Compound 86 The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile starting from 1-methylspiro[indoline-2,4'-piperidine]-3-one. (yellow solid).
  • Step 3 Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • Step 4 Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • a mixture of (3S,4R)-N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine (105 mg, 0.31 mmol, 1 eq, trifluoroacetic acid), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (79 mg, 0.31 mmol, 1 eq), and diisopropylethylamine (121 mg, 0.94 mmol, 0.16 mL, 3 eq) in acetonitrile (1 mL) was stirred at 40 °C for 12 h.
  • Step 5 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 87) and 6- chloro-4-[(3R,4S)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (Compound 42)
  • the enantiomers were separated by SFC (column: DAICEL CHIRALPAK AD(250mm*30mm, 10 ⁇ m); mobile phase: ethanol (0.1% NH 4 OH) in CO 2 ).
  • Step 2 Preparation of (3aR,6aS)-5-(4-fluorophenyl)-3a,6a-dimethyl- 2,3,4,6- tetrahydro-1H-pyrrolo[3,4-c]pyrrole
  • tert-butyl (3aS,6aR)-2-(4-fluorophenyl)-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate 100 mg, 0.30 mmol, 1 eq
  • dichloromethane 3 mL
  • trifluoroacetic acid 1 mL
  • Step 3 Preparation of 6-chloro-4-[2-(4-fluorophenyl)-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 89)
  • To a solution of (3aR,6aS)-5-(4-fluorophenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H- pyrrolo[3,4-c]pyrrole 50 mg, 0.14 mmol, 1 eq, trifluoroacetate
  • 4,6-dichloro-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile 36.47 mg, 143.54 ⁇ mol, 1 eq
  • Step 2 Preparation of N-[(4-fluorophenyl)methyl]-N-methyl-piperidin-4-amine
  • a solution of tert-butyl 4-[(4-fluorophenyl)methyl-methyl-amino] piperidine-1-carboxylate (120 mg, 0.372 mmol) in HCl/dioxane (5 mL, 4 M) was stirred at 20°C for 1 hour. The reaction was concentrated to give N-[(4-fluorophenyl)methyl]-N-methyl-piperidin-4-amine (96 mg, crude, HCl) as a white solid.
  • Step 3 Preparation of 6-chloro-4-[4-[(4-fluorophenyl)methyl-methyl-amino]-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 92)
  • N-[(4-fluorophenyl)methyl]-N-methyl-piperidin-4-amine 96 mg, 0.371 mmol, HCl
  • CH 3 CN 3 mL
  • 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile 94 mg, 0.371 mmol
  • diisopropylethylamine 239 mg, 1.85 mmol, 323.0 ⁇ L
  • Step 2 Preparation of 4-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H- pyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • tert-butyl (3aR,6aS)-2-(6-chloro-3-cyano-1-methyl-2-oxo-1,5-naphthyridin- 4-yl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (30 mg, 65.51 ⁇ mol, 1 eq) in dichloromethane (3 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at 25 °C for 1 hour.
  • Step 3 Preparation of 4-[(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 96)
  • 4-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl] -6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (30 mg, 63.58 ⁇ mol, 1 eq, trifluoroacetate) in methanol (3 mL) was added sodium acetate (26.08 mg, 0.32 mmol, 5 eq), and the mixture was stirred at
  • Step 2 Preparation of O1-tert-butyl O3,O4-dimethyl pyrrolidine-1,3,4-tricarboxylate
  • dimethyl 1-benzylpyrrolidine-3,4-dicarboxylate (15.0 g, 54.09 mmol, 1.0 eq) and di-tert-butyl dicarbonate (17.71 g, 81.14 mmol, 18.64 mL, 1.5 eq) in ethanol (150 mL) was added palladium on activated carbon catalyst (1.5 g, 10% purity) under nitrogen.
  • the reaction mixture was degassed with hydrogen (3X) and stirred at 20 °C for 15 h under hydrogen (15 psi).
  • Step 3 Preparation of tert-butyl 3,4-bis(hydroxymethyl)pyrrolidine-1-carboxylate
  • tetrahydrofuran 300 mL
  • lithium borohydride 11.91 g, 546.74 mmol, 12.08 eq
  • To the reaction mixture was added dropwise water (100 mL) and methanol (200 mL) over 30 minutes, then stirred at 20 °C for 1 h.
  • Step 4 Preparation of tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1- carboxylate
  • a solution of tert-butyl 3,4-bis(hydroxymethyl)pyrrolidine-1-carboxylate (10.0 g, 43.24 mmol, 1 eq) and TEA (26.25 g, 259.42 mmol, 36.11 mL, 6.0 eq) in dichloromethane (300 mL) was added a solution of methanesulfonyl chloride (17.79 g, 155.30 mmol, 12.02 mL, 3.59 eq) in dichloromethane (10 mL) at 0 °C for 10 minutes.
  • reaction mixture was then stirred at 20 °C for 15 hours.
  • the reaction mixture was treated with saturated aqueous ammonium chloride solution (100 mL) and extracted with ethyl acetate (200 mL x 2).
  • the organic extract was washed with saturated aqueous NaHCO 3 solution (100 mL), brine (100 mL x 3), dried over sodium sulfate, and concentrated under reduced pressure to afford tert- butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (16.0 g, 41.29 mmol, 96% yield) as a yellow oil.
  • Step 5 Preparation of tert-butyl 2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate
  • tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (0.33 g, 0.85 mmol, 1.0 eq) and potassium carbonate (1.18 g, 8.52 mmol, 0.64 mL, 10.0 eq) in acetonitrile (10 mL) was added (4-fluorophenyl)methanamine (319 mg, 2.56 mmol, 0.29 mL, 3.0 eq), and the reaction mixture was stirred at 100 °C for 15 h.
  • Step 7 Preparation of 8-[(3aR,6aR)-2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (Compound 97)
  • 5-[(4-fluorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole 48 mg, 0.19 ⁇ mol, 1 eq, hydrochloride
  • N,N-diisopropylethylamine 120 mg, 0.93 mmol, 0.16 mL, 5.0 eq
  • Step 2 Preparation of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl- piperidine-1-carboxylate
  • a solution of tert-butyl 4-(4-chloro-2-hydroxy-anilino)-3-methyl-piperidine-1-carboxylate (3 g, 8.80 mmol, 1 eq) in methanol (10 mL) were added paraformaldehyde (2.64 g, 88.02 mmol, 2.42 mL, 10 eq) and sodium cyanoborohydride (1.11 g, 17.60 mmol, 2 eq), and the reaction mixture was stirred at 25 °C for 12 hours.
  • Step 3 Preparation of 5-chloro-2-[methyl-(3-methyl-4-piperidyl)amino]phenol
  • dichloromethane 5 mL
  • HCl in dioxane 4 M, 5 mL
  • Step 2 8-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile
  • tert-butyl (3aR,6aS)-2-(6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl)- 3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (30 mg, 0.07 mmol, 1 eq) in dichloromethane (5 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 190.66 eq), and the reaction mixture was stirred at 20 °C for 0.5 hours.
  • Step 3 Preparation of 8-[(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (Compound 101)
  • 8-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (30 mg, 0.07 mmol, 1 eq, trifluoroacetic acid) and 4-chloro-2-hydroxy-benzaldehyde (16.11 mg, 0.1 mmol, 1.5 eq) in CH 3 OH (5 mL) were added sodium acetate (16.88
  • reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC [column: Phenomenex Synergi C18150*25mm* 10um; mobile phase: (7-37% CH 3 CN in water (0.225% formic acid))] to afford 8-[(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]- 3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (10.3 mg, 22.14 ⁇ mol, 32 % yield) as a white solid.
  • reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC [column: Phenomenex Synergi C18150*25mm* 10 ⁇ m; mobile phase: (4-37% CH 3 CN in water (0.225% formic acid))] to afford 8-[(3aS,6aR)-2-[(4-fluorophenyl)methyl]-3a,6a-dimethyl- 1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (29.9 mg, 68.30 ⁇ mol, 50 % yield) as a white solid.
  • Step 2 Preparation of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)piperidine- 1-carboxylate
  • tert-butyl 4-(4-chloro-2-hydroxy-anilino)piperidine-1- carboxylate 300 mg, 0.92 mmol, 1 eq
  • paraformaldehyde 138 mg, 4.59 mmol, 5 eq
  • the mixture was stirred at 20 °C for 11 hour.
  • Sodium triacetoxyborohydride (389 mg, 1.84 mmol, 2 eq) was then added, and the reaction mixture was stirred at 20 °C for 1 hour.
  • Step 3 Preparation of 5-chloro-2-[methyl(4-piperidyl)amino]phenol
  • a solution of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)piperidine-1-carboxylate (100 mg, 0.29 mmol, 1 eq) in dichloromethane (2 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at 20 °C for 0.5 hour. The mixture was concentrated under reduced pressure to give 5-chloro-2-[methyl(4-piperidyl)amino]phenol (70 mg, crude) as a brown oil.
  • Step 4 Preparation of 6-chloro-4-[4-(4-chloro-2-hydroxy-N-methyl-anilino)-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • 5-chloro-2-[methyl(4-piperidyl)amino]phenol 70 mg, 0.29 mmol, 1.23 eq
  • triethylamine 96 mg, 0.94 mmol, 4 eq
  • 4,6-dichloro-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile 60 mg, 0.24 mmol, 1 eq
  • reaction mixture was diluted with water (10 mL) and extracted with dichloromethane (5 mL x 3). The combined organic extracts were washed with brine (5 mL x2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
  • Step 2 Preparation of 5-chloro-N-methyl-N-(4-piperidyl)pyridin-2-amine
  • dichloromethane 2 mL
  • trifluoroacetic acid 1.54 g, 13.51 mmol, 36.67 eq
  • reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (5 mL x 3). The combined organic extract was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated.
  • Step 2 Preparation of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo [3,4-c]pyrrol-5- ylmethyl)-5-chloro-phenol
  • dichloromethane 5 mL
  • trifluoroacetic acid 323 mg, 2.83 mmol, 0.21 mL
  • Step 3 Preparation of 6-chloro-4-[2-[(4-chloro-2-hydroxy-phenyl)methyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 105)
  • 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-ylmethyl)-5-chloro- phenol 150 mg, 0.41 mmol, 1.04 eq, TFA salt
  • triethylamine 159 mg, 1.57 mmol, 0.22 mL, 4 eq
  • 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile 100 mg, 0.39 mmol, 1 eq
  • reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (5 mL x 3). The combined organic extract was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
  • the aqueous phase was extracted with ethyl acetate (300 mL ⁇ 4).
  • the combined organic extract was washed with brine (150 mL ⁇ 2), dried with anhydrous sodium sulfate, filtered, and concentrated in vacuum.
  • the resulting residue was purified by semi-preparative reverse phase HPLC [(column: Phenomenex luna C18 (250*70 mm, 10 um); mobile phase: (5-35% CH 3 CN in water (0.225% formic acid))] to afford ethyl 3-[acetyl(methyl)amino]pyridine-2- carboxylate (9.3 g, 41.85 mmol, 87% yield) as a yellow solid.
  • Step 2 Preparation of ethyl 3-[acetyl(methyl)amino]-1-oxido-pyridin-1- ium-2- carboxylate To a mixture of ethyl 3-[acetyl(methyl)amino]pyridine-2-carboxylate (9.2 g, 41.40 mmol, 1 eq) and urea hydrogen peroxide (5.84 g, 62.10 mmol, 1.5 eq) in dichloromethane (92 mL) at 0 °C was added (2,2,2-trifluoroacetyl) 2,2,2-trifluoroacetate (13.04 g, 62.10 mmol, 8.6 mL, 1.5 eq) dropwise, and the reaction mixture was stirred for 2 h at 20 °C.
  • Step 3 Preparation of ethyl 3-[acetyl(methyl)amino]-6-cyano-pyridine-2- carboxylate
  • ethyl 3-[acetyl(methyl)amino]-1-oxido-pyridin-1-ium-2-carboxylate 9.4 g, 39.46 mmol, 1 eq
  • trimethylsilyl cyanide 4.70 g, 47.35 mmol, 5.9 mL, 1.2 eq
  • dichloromethane 100 mL
  • benzoyl chloride 7.76 g, 55.24 mmol, 6.4 mL, 1.4 eq
  • Triethylamine (5.99 g, 59.18 mmol, 8.2 mL, 1.5 eq) was then added dropwise, and the resulting mixture was stirred for 3 h. The mixture was poured into ice water (100 mL) and stirred for 5 minutes. The aqueous phase was extracted with dichloromethane (100 mL ⁇ 2), and the combined organic extract was washed with brine (50 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered, and concentrated.
  • Step 4 Preparation of 8-hydroxy-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile
  • ethyl 3-[acetyl(methyl)amino]-6-cyano-pyridine-2-carboxylate 7.86 g, 31.79 mmol, 1 eq
  • tetrahydrofuran 80 mL
  • potassium bis(trimethylsilyl)amide 1 M, 39.7 mL, 1.25 eq
  • Step 5 Preparation of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate
  • dichloromethane 20 mL
  • triethylamine 2.01 g, 19.88 mmol, 2 eq
  • dimethylaminopyridine 121 mg, 1.0 mmol, 0.1 eq
  • trifluoromethanesulfonic anhydride 3.37 g, 11.93 mmol, 1.2 eq
  • Step 6 Preparation of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate
  • Step 7 Preparation of 8-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl)-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile.
  • Step 8 Preparation of 8-[2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (Compound 106)
  • 8-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl)-5-methyl-6-oxo- 1,5-naphthyridine-2-carbonitrile 70 mg, 171.00 ⁇ mol, 1 eq, TFA
  • 4-fluorobenzaldehyde 25.47 mg, 205.20 ⁇ mol, 21.58 ⁇ L, 1.2 eq in dichloroethane (5 mL) was added TFA (86.52 mg, 854.99 ⁇ mol, 119.00 ⁇ L, 5 eq) followed by sodium cyano
  • Step 2 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-4-(4- fluoroanilino)-3-methyl-piperidine-1-carboxylate.
  • Step 2 Preparation of 4-[(3aS,6aS)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate.
  • the title compound was prepared in an analogous manner to 6-chloro-4-((3S,4R)-4-((4- fluorophenyl)amino)-3-methylpiperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile stating from 6-chloro-4-[(3R,4S)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid).
  • Step 2 Preparation of ethyl 3-amino-6-methyl-pyridine-2-carboxylate A mixture of ethyl 3-amino-6-bromo-pyridine-2-carboxylate (4.15 g, 16.93 mmol, 1 eq), cyclopentyl(diphenyl)phosphane;dichloropalladium;iron (1.24 g, 1.69 mmol, 0.1 eq), 2,4,6- trimethyl-1,3,5,2,4,6-trioxatriborinane (12.75 g, 50.80 mmol, 14.20 mL, 50% purity, 3 eq), and sodium bicarbonate (3.59 g, 33.87 mmol, 2 eq) in dioxane (40 mL) and water (1.5 mL) was degassed and purged with nitrogen (3X).
  • Step 3 Preparation of ethyl 3-[(2-cyanoacetyl)amino]-6-methyl-pyridine-2-carboxylate 2-Cyanoacetic acid (1.72 g, 20.20 mmol, 1.3 eq) was added to acetic anhydride (20 mL), and the reaction mixture was stirred at 80 °C for 1 hr. Ethyl 3-amino-6-methyl-pyridine-2- carboxylate (2.8 g, 15.54 mmol, 1 eq) was then added, and the reaction mixture was stirred at 80 °C for 1 h, then concentrated under reduced pressure.
  • Step 4 Preparation of 4-hydroxy-6-methyl-2-oxo-1H-1,5-naphthyridine-3-carbonitrile
  • Step 5 Preparation of 4-hydroxy-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • DMF dimethyl sulfoxide
  • Step 6 Preparation of 4-chloro-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a mixture of 4-hydroxy-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (300 mg, 1.39 mmol, 1 eq) and N-ethyl-N-isopropyl-propan-2-amine (1.08 g, 8.36 mmol, 1.46 mL, 6 eq) in acetonitrile (5 mL) were added phosphorus oxychloride (1.07 g, 6.97 mmol, 0.65 mL, 5 eq) and benzyltriethylammonium chloride (635 mg, 2.79 mmol, 2 eq) in one portion at 20 °C under nitrogen, and the reaction mixture was stirred at 20 °C for 12 h.
  • Step 7 Preparation of 4-(5-fluorospiro[3H-benzofuran-2,4'-piperidine]-1'-yl)-1,6- dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile
  • 5-fluorospiro[3H-benzofuran-2,4'-piperidine] 92 mg, 0.3 mmol, 1.34 eq, trifluoroacetic acid
  • diisopropylethylamine 371 mg, 2.87 mmol, 0.5 mL, 13.41 eq
  • 4-chloro-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile 50 mg, 0.2 mmol, 1 eq
  • Step 2 Preparation of 2,4-dimethyl-7-[(3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]-1-piperidyl]-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile
  • the title compounds was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7- spiro[3H-benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate.
  • reaction was diluted with saturated aqueous ammonium chloride (20 mL) and water (20 ml), and the resulting mixture was extracted with ethyl acetate (4 x 20 mL). The combined organic extracts were washed with brine (80 mL), dried over anhydrous sodium sulfate, filtered, and concentrated.
  • Step 2 Preparation of 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1- one
  • a mixture of methyl 1-benzyl-4-[[4-(trifluoromethoxy)phenyl]methyl]piperidine-4- carboxylate (4.3 g, 9.48 mmol, 1 eq, formic acid) in trifluoromethanesulfonic acid (40 mL) was degassed and purged with nitrogen (3X), then stirred at 80 °C for 3 h under nitrogen atmosphere.
  • the reaction mixture was poured into water (200 mL), and the resulting mixture was treated with sodium bicarbonate to adjust the pH to 7 ⁇ 8.
  • Step 3 Preparation of 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-ol
  • 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-one 2.3 g, 6.13 mmol, 1 eq
  • lithium aluminum hydride 465 mg, 12.25 mmol, 2 eq
  • the reaction mixture was then stirred at 0°C for 1 h under nitrogen atmosphere.
  • Step 4 Preparation of 1'-benzyl-5-(trifluoromethoxy)spiro[indane-2,4'-piperidine] To 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-ol (2.8 g, 7.42 mmol, 1 eq) were added triethylsilane (4.31 g, 37.10 mmol, 5.93 mL, 5 eq) and trifluoroacetic acid (5.08 g, 44.52 mmol, 3.30 mL, 6 eq), and the reaction mixture was stirred at 80 °C for 12 h.
  • Step 5 Preparation of 5-(trifluoromethoxy)spiro[indane-2,4'-piperidine]
  • methanol 8 mL
  • 10% palladium on carbon 200 mg, 2.21 mmol, 1 eq
  • the mixture was stirred under hydrogen (50 psi) at 25°C for 12 hours.
  • Step 6 Preparation of 6-chloro-1-methyl-2-oxo-4-(5-(trifluoromethoxy)-1,3- dihydrospiro[indene-2,4'-piperidin]-1'-yl)-1,2-dihydro-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from 5- (trifluoromethoxy)spiro[indane-2,4'-piperidine]. (yellow solid).
  • Step 2 Preparation of (3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4- amine
  • tert-butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1- carboxylate 70 mg, 0.20 mmol, 1 eq
  • dichloromethane 1 mL
  • trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 66.15 eq
  • Step 3 Preparation of 7-[(3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-1-piperidyl]- 2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile
  • 3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4-amine 73 mg, 0.21 mmol, 1 eq, trifluoroacetic acid
  • acetonitrile 2 mL
  • N,N- diisopropylethylamine 132 mg, 1.02 mmol, 0.178 mL, 5 eq
  • (6-cyano- 2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate 72 mg, 0.205 mmol, 1 eq
  • Step 2 Preparation of N-(4-chloro-2-fluoro-phenyl)piperidin-4-amine
  • a mixture of tert-butyl 4-(4-chloro-2-fluoro-anilino)piperidine-1-carboxylate (100 mg, 0.3 mmol, 1 eq) and trifluoroacetic acid (1.08 g, 9.45 mmol, 0.7 mL, 31.09 eq) in dichloromethane (0.7 mL) was degassed and purged with nitrogen atmosphere (3X), then stirred at 25 °C for 0.5 h under nitrogen atmosphere.
  • Step 2 Preparation of N-methyl-1-[4-(trifluoromethoxy)phenyl]piperidin-4-amine H N N F 3 CO
  • a mixture of tert-butyl N-methyl-N-[1-[4-(trifluoromethoxy)phenyl]-4-piperidyl]carbamate (100 mg, 0.28 mmol, 1 eq) and triethylamine (1.1 g, 9.5 mmol, 0.7 mL, 35.40 eq) in dichloromethane (0.7 mL) was degassed and purged with nitrogen (3X), then stirred at 25°C for 0.5 h under nitrogen atmosphere.
  • Step 3 Preparation of 1,6-dimethyl-4-[methyl-[1-[4-(trifluoromethoxy)phenyl]-4- piperidyl]amino]-2-oxo-1,5-naphthyridine-3-carbonitrile
  • 4-chloro-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile 62 mg, 0.27 mmol, 1 eq
  • diisopropylethylamine(102 mg, 0.80 mmol, 0.14 mL, 3 eq) in acetonitrile (2 mL) was degassed and purged with nitrogen (3X), then stirred at 40 °C for 3 h under nitrogen atmosphere.
  • step 2 preparation of 6-chloro-1-methyl-4-((3R,4S)-3-methyl-4-(methyl(4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3S,4R)-3- methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3R,4S)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate.
  • Step 2 Preparation of (3S,4R)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4- amine and (3R,4S)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine Racemic 3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (1.5 g, 5.47 mmol) was purified by SFC (column: DAICEL CHIRALPAK IG(250mm*50mm,10um); mobile phase: [0.1 % ammonium hydroxide ethyl alcohol]; B%: 35%-35%,15;180 min), then further purified by prep-HPLC (column: Phenomenex luna C18150*25 mm* 10 um; mobile phase: [water formic acid-acetonitrile]; B%: 15%-45 %,10 min), and then SFC (column: DAICEL CHIRAL
  • Step 3 Preparation of 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-3- methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (white solid).
  • tert-Butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (240 mg, 48%) was obtained as colorless oil.
  • tert-Butyl (3R,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate 230 mg, 46%) was obtained as a colorless oil.
  • Step 2 Preparation of (3S,4R)-N-(4-chlorophenyl)-3-methyl-piperidin-4-amine
  • dichloromethane 1 mL
  • trifluoroacetic acid 770 mg, 6.75 mmol, 0.5 mL, 21.94 eq
  • Step 3 Preparation of 7-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile
  • 3S,4R)-N-(4-chlorophenyl)-3-methyl-piperidin-4-amine trifluoroacetic acid 100 mg, 0.30 mmol, 1 eq
  • diisopropylethylamine (191 mg, 1.48 mmol, 0.26 mL, 5 eq)
  • (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate 104 mg, 0.30 mmol, 1 eq
  • the reaction mixture was stirred at 40 °C for 12 h.
  • the title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3S,4R)-3- methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,4S)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (yellow solid).
  • Step 2 Preparation of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2- c]pyridine-5-carboxylate
  • O5-benzyl O1-tert-butyl (3aR,7aS)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-1,5-dicarboxylate 400.00 mg, 1.11 mmol, 1 eq
  • dichloromethane 4 mL
  • trifluoroacetic acid 6.16 g, 54.03 mmol, 4.00 mL, 48.68 eq
  • Step 3 Preparation of benzyl (3aR,7aS)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate and benzyl (3aS,7aR)-1-[4- (trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate
  • 1-bromo-4-(trifluoromethoxy)benzene (1.85 g, 7.68 mmol, 1.14 mL, 2.5 eq) in dioxane (8 mL)
  • benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate 800.00 mg, 3.07 mmol, 1
  • the reaction mixture was then stirred at 100 °C for 3 h under nitrogen atmosphere.
  • the reaction was diluted with water (200 mL) and extracted with ethyl acetate (200 mL ⁇ 3).
  • the combined organic layers were washed by brine and dried over sodium sulfate, filtered, and concentrated under reduced pressure.
  • the residue was purified by semi-preparative reverse phase HPLC ([water(ammonium bicarbonate)-acetonitrile];B%: 57% - 87%,8 min).
  • the selected fraction was concentrated to remove most of the acetonitrile, then extracted with ethyl acetate (100 mL ⁇ 2).
  • the product was further purified by SFC (column: DAICEL CHIRALPAK AD (250mm ⁇ 30mm,10um); mobile phase: [0.1% ammonium hydroxide isopropanol]; B%: 30%-30%,6.10; 128 min).
  • Benzyl (3aR,7aS)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (0.35 g, 27 %) was obtained as a colorless oil.
  • Step 4 Preparation of (3aS,7aR)-1-(4-(trifluoromethoxy)phenyl)octahydro-1H- pyrrolo[3,2-c]pyridine
  • benzyl (3aS,7aR)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-5-carboxylate (0.32 g, 0.76 mmol, 1 eq) in tetrahydrofuran (5 mL) was added 10% palladium on carbon (0.3 g, 0.76 mmol, 1 eq) at 25 °C, and the resulting mixture was degassed and purged with hydrogen (3X).
  • Step 5 Preparation of 4-[(3aS,7aR)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from (3aS,7aR)- 1-(4-(trifluoromethoxy)phenyl)octahydro-1H-pyrrolo[3,2-c]pyridine.
  • tert- Butyl 4-oxopiperidine-1-carboxylate (156 mg, 0.78 mmol, 1.0 eq) in tetrahydrofuran (1 mL) was then added dropwise at -78°C, and the reaction mixture was stirred at -78°C for 0.5 h.
  • the reaction was diluted with saturated aqueous ammonium chloride solution (15 mL) and water (15 mL), and the resulting mixture was extracted with ethyl acetate (10 mL ⁇ 3). The combined organic extracts were washed by brine and dried over sodium sulfate, filtered, and concentrated under reduced pressure.
  • Step 2 Preparation of 4-((5-chloropyridin-2-yl)methyl)piperidin-4-ol
  • tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4-hydroxy-piperidine-1- carboxylate 84 mg, 0.26 mmol, 1.0 eq
  • dichloromethane 3 mL
  • trifluoroacetic acid 1.54 g, 13.51 mmol, 1 mL, 52.5 eq
  • Step 3 Preparation of 6-chloro-4-(4-((5-chloropyridin-2-yl)methyl)-4-hydroxypiperidin- 1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile
  • the title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid).
  • Step 2 Preparation of (3aR,7aS)-1-(4-fluorophenyl)octahydro-1H-pyrrolo[3,2- c]pyridine
  • benzyl (3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate 80 mg, 0.226 mmol, 1 eq
  • Step 3 Preparation of 7-[(3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile
  • the title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloroanilino)- 3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aR,7aS)-1-(4-fluorophenyl)octahydro-1H-pyrrolo[3,2-c]pyridine.
  • Step 2 Preparation of ( 3S,4R)-N-(4-fluorophenyl)-3-methyl-piperidin-4-amine
  • a mixture of methyl 5-chloro-2-[(2-cyanoacetyl)amino]benzoate (4.7 g, 18.60 mmol, 1 eq) and 1,8-diazabicyclo[5.4.0]undec-7-ene (4.25 g, 27.90 mmol, 4.21 mL, 1.5 eq) in ethanol (40 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 2 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure.
  • Step 3 Preparation of 6-chloro-4-hydroxy-1-methyl-2-oxo-quinoline-3-carbonitrile To a mixture of 6-chloro-4-hydroxy-2-oxo-1H-quinoline-3-carbonitrile (2.95 g, 13.37 mmol, 1 eq) in DMF (15 mL) was added 60% NaH (1.34 g, 33.43 mmol, 2.5 eq), and the resulting mixture was stirred at 0 °C for 0.5 h. Iodomethane (5.69 g, 40.12 mmol, 2.50 mL, 3 eq) was then added, and the reaction mixture was stirred at 0 °C for 0.5 h under nitrogen.
  • Step 4 Preparation of 4,6-dichloro-1-methyl-2-oxo-quinoline-3-carbonitrile To a mixture of 6-chloro-4-hydroxy-1-methyl-2-oxo-quinoline-3-carbonitrile (2.2 g, 9.38 mmol, 1 eq) and benzyl(triethyl)ammonium;chloride (4.27 g, 18.75 mmol, 2 eq) in acetonitrile (30 mL) were added diisopropylethylamine (7.27 g, 56.26 mmol, 9.80 mL, 6 eq) and phosphoryl chloride (7.19 g, 46.88 mmol, 4.36 mL, 5 eq) in one portion at 20 °C under nitrogen, and the reaction mixture was stirred at 25 °C for 4 h.
  • Step 5 Preparation of 6-chloro-1-methyl-4-((3S,4R)-3-methyl-4-((4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydroquinoline-3- carbonitrile
  • a mixture of (3S,4R)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine 60 mg, 0.22 mmol, 1 eq
  • 4,6-dichloro-1-methyl-2-oxo-quinoline-3-carbonitrile 55 mg, 0.22 mmol, 1 eq
  • diisopropylethylamine 85 mg, 0.66 mmol, 114.31 uL, 3 eq
  • reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex C18150*25mm*10um; mobile phase: [water(ammonium hydrogen carbonate)-acetonitrile]; B%: 55%-85%, 8min) to afford 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo- quinoline-3-carbonitrile (65 mg, 60%)as a white solid.
  • tert-Butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate 200 mg, 49%) was obtained as a colorless oil.
  • tert-Butyl (3S,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (170 mg, 42%) was obtained as a colorless oil.
  • Step 2 Preparation of 7-[(3R,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile
  • the title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloroanilino)- 3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate. (white solid).

Abstract

Disclosed herein are compounds of Formula (A), or a pharmaceutically acceptable salt thereof, which inhibit the activity of one or both of diacylglycerol kinase alpha (DGKα). These compounds are useful in the treatment of proliferative diseases (e.g., cancer) and viral infections.

Description

DGK TARGETING COMPOUNDS AND USES THEREOF RELATED APPLICATIONS This application claims the benefit of priority to U.S. Provisional Application No. 63/305,668, filed February 1, 2022, the entire contents of which are incorporated herein by reference. BACKGROUND Human cancers harbor numerous genetic and epigenetic alterations, generating neoantigens potentially recognizable by the immune system (Sjoblom et al. (2006) Science 314:268–74). The adaptive immune system, comprised of T and B lymphocytes, has powerful anti-cancer potential, with a broad capacity and exquisite specificity to respond to diverse tumor antigens. Further, the immune system demonstrates considerable plasticity and a memory component. The successful harnessing of all these attributes of the adaptive immune system would make immunotherapy unique among all cancer treatment modalities. However, although an endogenous immune response to cancer is observed in preclinical models and patients, this response is ineffective, and established cancers are viewed as “self” and tolerated by the immune system. Contributing to this state of tolerance, tumors may exploit several distinct mechanisms to actively subvert anti-tumor immunity. These mechanisms include dysfunctional T-cell signaling (Mizoguchi et al. (1992) Science 258:1795–98), suppressive regulatory cells (Facciabene et al. (2012) Cancer Res.72:2162–71), and the co- opting of endogenous “immune checkpoints,” which serve to down-modulate the intensity of adaptive immune responses and protect normal tissues from collateral damage, by tumors to evade immune destruction (Topalian et al. (2012) Curr. Opin. Immunol.24:1–6; Mellman et al. (2011) Nature 480:480–89). Accordingly, new and novel anticancer agents that are safe and effective in restoring T-cell activation, lowering antigen threshold, enhancing antitumor functionality, and/or overcoming the suppressive effects of one or more endogenous immune checkpoints are needed in the art for the treatment of cancer. SUMMARY Disclosed herein are compounds that have activity as inhibitors of diacylglycerol kinase alpha (DGKα). Additionally, in embodiments, the disclosed compounds cause the degradation of DGKα. In embodiments, compounds described herein have desirable efficacy, stability, bioavailability, therapeutic index, and toxicity values that are important to their use as pharmaceuticals. In aspects, provided herein are compounds of having the structure of Formula (A): (A) or a pharmaceutically acceptable salt thereof, wherein: Ring A is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaromatic ring fused to ring B; Ring C is a heterocyclic ring, which may be further substituted in addition to R3 and R4; R1 is C1–C4 alkyl; R2 is H or an electron withdrawing functional group; each R3 is independently NR5R6; each R4 is independently C1–C4 alkyl or C1–C4 alkoxy, or one R3 and one R4, together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring; R5 and R6 are each independently H, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or R5 and R6, together with the nitrogen to which they are attached, form a substituted or unsubstituted ring; m and n are each independently 0, 1, 2, 3, or 4. In embodiments, disclosed herein are compounds having the structure of Formula (A): (A) or a pharmaceutically acceptable salt thereof, wherein Ring A is a substituted or unsubstituted heteroaromatic ring fused to ring B. In embodiments of the compound of Formula (I), the compound has the structure of Formula (II): (II) or a pharmaceutically acceptable salt thereof, wherein R9 is H or C1–C4 alkyl. In embodiments of Formula II, R9 is straight or branched C1–C4 alkyl. In embodiments of Formula II, R9 is methyl. In embodiments of the compound of Formula (I), the compound has the structure of Formula (III): (III) or a pharmaceutically acceptable salt thereof, wherein R10 is C1–C4 alkyl or an electron withdrawing functional group. In embodiments of Formula (III), R10 is straight or branched C1–C4 alkyl. In embodiments of the compound of Formula (III), the compound has the structure of Formula (IIIa): (IIIa) or a pharmaceutically acceptable salt thereof. In embodiments of the compound of Formula (III) or Formula (IIIa), R10 is selected from halogen, -CH3, -SO2CF3, -N(R11)2, -NO2, -SO2R12, -C≡N, -C(R13)3, -COR14, -CO2R16, - CON(R17)2, and -N=O, wherein: each of R11, R12, R14, R15, R16, and R17 is independently H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl); and each R13 is halogen. In embodiments of the compound of Formulae (III) and (IIIa), R10 is -Cl. In embodiments of the compound of Formula (III) and (IIIa), R10 is -C≡N. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R1 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R1 is methyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R2 is selected from H, halogen, -SO2CF3, -N(R32)2, -NO2, -SO2R26, -C≡N, -C(R27)3, -COR28, - CO2R30, -CON(R31)2, and -N=O, wherein: each of R32, R26, R28, R30, and R31 is independently H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl); and each R33 is halogen. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R2 is H. In embodiments of the compounds of Formula (I), (II), (III), and (IIIa), R2 is -C≡N. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R4 is C1– C4 alkyl (e.g., straight or branched C1–C4 alkyl). In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R4 is C1– C4 alkoxy. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is further substituted by one or more substituents selected from R15, R18, R19, R20, R21, and R22. In embodiments any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from ; ; ; and , wherein: R18 and R19 are each, independently H, OH, C1–C4 alkoxy, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R20 is H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring; R21 is H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, R23 and R24 are each, independently H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R15 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and indicates the point of attachment of ring C to ring B. In embodiments of Ring C, R21 is straight or branched C1–C4 alkyl. In embodiments, R22 is straight or branched C1–C4 alkyl. In embodiments of Ring C, R23 and R24 are each, independently H, straight or branched C1–C4 alkyl, aryl, or heteroaryl. In embodiments of Ring C, R15 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is , wherein R18 and R19, together with the carbon atom to which they are attached, form a 5- or 6-membered substituted or unsubstituted carbocyclic ring or a 5- or 6- membered substituted or unsubstituted heterocyclic ring. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is
Figure imgf000008_0001
, wherein R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached form a substituted or unsubstituted 5-, 6-, or 7-membered carbocyclic ring or a substituted or unsubstituted 5-, 6-, or 7-membered heterocyclic ring. In embodiments of Ring C, R20 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000008_0002
; ; ; ; ; ; ; ; and
Figure imgf000008_0004
Figure imgf000008_0003
In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is wherein R21 is
Figure imgf000008_0006
, wherein:
Figure imgf000008_0005
R25 is selected from OH, halogen, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), C1–C4 alkoxy, C1–C4 haloalkyl, C1–C4 haloalkoxy, aryl, and heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R21 to N. In embodiments of any one of the compounds of Formula (I), (II), (III), and (IIIa), ring C is
Figure imgf000009_0001
, wherein R is
Figure imgf000009_0002
, wherein: R25 is selected from OH, halogen, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), C1–C4 alkoxy, C1–C4 haloalkyl, C1–C4 haloalkoxy, aryl, and heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R21 to N. In embodiments of Ring C, each R25 is independently selected from OH, halogen, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), C1–C4 alkoxy, C1–C4 haloalkyl, C1–C4 haloalkoxy, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is
Figure imgf000009_0003
, wherein R21 is selected from
Figure imgf000009_0004
Figure imgf000009_0005
In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is w 21
Figure imgf000010_0001
herein R is selected from
Figure imgf000010_0002
; ; ; ; ; ; and
Figure imgf000010_0003
In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000010_0004
; ; ; ; ; ; ; and
Figure imgf000010_0005
In embodiments of Ring C, R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. In embodiments of Ring C, R22 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is , wherein R18 and R19 are each independently selected from -H, -OH, C1
Figure imgf000011_0001
– C4 alkoxy, C1–C4 alkyl (straight or branched C1–C4 alkyl), and
Figure imgf000011_0002
, wherein: R26 is H, C1–C4 alkyl (straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, C1–C4 alkyl (straight or branched C1–C4 alkyl), halogen, C1–C4 alkoxy, or C1–C4 haloalkoxy, or two adjacent R27 groups, and the carbon atoms two which they are attached, form a substituted or unsubstituted 5- or 6-membered ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R18 and R19 to ring C. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000011_0003
Figure imgf000011_0004
and
Figure imgf000011_0005
, wherein R18 and R19 are each independently selected from -H, -OH, C1–C4 alkyl, and
Figure imgf000011_0006
, wherein: R26 is H, C1–C4 alkyl (straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, C1–C4 alkyl (straight or branched C1–C4 alkyl), halogen, C1–C4 alkoxy, or C1–C4 haloalkoxy, or two adjacent R27 groups, and the carbon atoms two which they are attached, form a substituted or unsubstituted 5- or 6-membered ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R18 and R19 to ring C. In embodiments of Ring C, R26 is straight or branched C1–C4 alkyl. In embodiments of Ring C, each R27 is -OH, straight or branched C1–C4 alkyl, C1–C4 alkoxy, or C1–C4 haloalkoxy. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from ; ; ; ; ; ; ; ; ;
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000017_0001
. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is
Figure imgf000017_0002
; ; ; ; , wherein 18 19
Figure imgf000017_0003
R and R , together with the carbon atom to which they are attached, for
Figure imgf000017_0004
m , wherein: X and Y are each, independently CH2, NR29, O, or C=O; R28 is -OH, halogen, C1–C4 alkyl (straight or branched C1–C4 alkyl), C1–C4 haloalkyl, C1–C4 alkoxy, or C1–C4 haloalkoxy; R29 is H or C1–C4 alkyl (straight or branched C1–C4 alkyl); and w is 0, 1, or 2. In embodiments of Ring C, each R28 is independently selected from -OH, halogen, straight or branched C1–C4 alkyl, C1–C4 haloalkyl, C1–C4 alkoxy, and C1–C4 haloalkoxy. In embodiments of Ring C, R29 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formula (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000018_0001
; ; ; ; ; ; ; and
Figure imgf000018_0002
In embodiments of any one of the compounds of Formula (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000018_0003
; and
Figure imgf000018_0004
wherein R34 is C1–C4 alkyl. In embodiments of Ring C, R34 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is
Figure imgf000019_0001
, wherein R32 is
Figure imgf000019_0002
In embodiments of the compounds of Formula (I), (II), (III), and (IIIa), ring C is ;
Figure imgf000019_0003
; ; ; ; ; o
Figure imgf000019_0004
, wherein R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. In embodiments of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is selected from
Figure imgf000020_0001
Figure imgf000021_0001
; ; ; ; and
Figure imgf000021_0002
. In embodiments, ring C is
Figure imgf000021_0003
or
Figure imgf000021_0004
. In embodiments, disclosed herein are compounds selected from:
Figure imgf000022_0001
 
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
;
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
;
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
or a pharmaceutically acceptable salt thereof. In embodiments, disclosed herein are compounds selected from:
Figure imgf000045_0001
Figure imgf000046_0001
;
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
or a pharmaceutically acceptable salt thereof, wherein: X is CH2, O, or NR29; and R29 is H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl). In embodiments, disclosed herein are compounds selected from:
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
or a pharmaceutically acceptable salt thereof. Also disclosed herein are pharmaceutical compositions comprising one or more compounds disclosed herein and a pharmaceutically acceptable excipient. In embodiments, the pharmaceutical compositions further comprise one or more additional anti-cancer agents. In embodiments, at least one of the additional anti- cancer agents is an immune checkpoint inhibitor. In aspects, disclosed herein are methods treating cancer, comprising administering a therapeutically effective amount of one or more compounds disclosed herein, or a pharmaceutical composition thereof, to a subject in need thereof. In embodiments, the cancer is selected from colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma. In aspects, disclosed herein are methods of inhibiting the activity of diacylglycerol kinase alpha (DGKα), comprising administering a therapeutically effective amount one or more compounds disclosed herein, or a pharmaceutical composition thereof, to a subject in need thereof. DETAILED DESCRIPTION Diacylglycerol kinases (DGKs) are lipid kinases that mediate the conversion of diacylglycerol to phosphatidic acid thereby terminating T-cell functions propagated through the TCR signaling pathway. Thus, DGKs serve as intracellular checkpoints, and inhibition of DGKs is expected to enhance T-cell signaling pathways and T-cell activation. Knock-out mouse models of DGKα have shown a hyper-responsive T-cell phenotype and improved anti- tumor immune activity (Zha Y et al. Nature Immunology, (2006) 12:1343; Olenchock B. A. et al., Nature (2006) 11: 1174–81). Furthermore, tumor infiltrating lymphocytes isolated from human renal cell carcinoma patients were observed to overexpress DGKα that resulted in inhibited T-cell function (Prinz, P. U. et al. J Immunology (2012) 12:5990–6000). Thus, DGKα may be viewed as a target for cancer immunotherapy (Riese M. J. et al. Front Cell Dev Biol. (2016) 4: 108; Chen, S. S. et al. Front Cell Dev Biol. (2016) 4: 130; Avila-Flores, A. et al. Immunology and Cell Biology (2017) 95: 549-563; Noessner, E. Front Cell Dev Biol. (2017) 5: 16; Krishna, S., et al. Front Immunology (2013) 4:178; Jing, W. et al. Cancer Research (2017) 77: 5676–86). There remains a need for compounds useful as inhibitors of DGKα. Accordingly, disclosed herein are compounds that have activity as inhibitors of DGKα. In embodiments, the disclosed compounds selectively inhibit DGKα. In embodiments, the disclosed compounds are selective inhibitors for DGKα over other diacylglycerol kinases (e.g., DGKζ). In embodiments, the disclosed compounds cause the degradation of DGKα. In embodiments, the disclosed compounds cause the selective degradation of DGKα. In embodiments, the disclosed compounds cause the selective degradation of DGKα over other diacylglycerol kinases (e.g., DGKζ). The disclosed compounds can be used to treat certain diseases or disorders. In embodiments, the disclosed compounds can be used for the treatment of certain cancers (e.g., colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma). Definitions Unless otherwise defined herein, scientific, and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, chemistry, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurochemistry, virology, immunology, microbiology, pharmacology, genetics and protein and nucleic acid chemistry, described herein, are those well-known and commonly used in the art. The methods and techniques of the present disclosure are generally performed, unless otherwise indicated, according to conventional methods well-known in the art and as described in various general and more specific references that are cited and discussed throughout this specification. See, e.g., “Principles of Neural Science,” McGraw-Hill Medical, New York, N.Y. (2000); Motulsky, “Intuitive Biostatistics”, Oxford University Press, Inc. (1995); Lodish et al. “Molecular Cell Biology, 4th ed.,” W. H. Freeman & Co., New York (2000); Griffiths et al. “Introduction to Genetic Analysis, 7th ed. ,” W. H. Freeman & Co., N.Y. (1999); and Gilbert et al. “Developmental Biology, 6th ed.,” Sinauer Associates, Inc., Sunderland, M A (2000). Chemistry terms used herein are used according to conventional usage in the art, as exemplified by “The McGraw-Hill Dictionary of Chemical Terms,” Parker S., Ed., McGraw- Hill, San Francisco, Calif. (1985). All the above, and all other publications, patents, and published patent applications referred to in this application are specifically incorporated by reference herein. In case of conflict, the present specification, including its specific definitions, will control. As used herein, the term “cell” is meant to refer to a cell that is in vitro, ex vivo, or in vivo. In embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In embodiments, an in vitro cell can be a cell in a cell culture. In embodiments, an in vivo cell is a cell living in an organism such as a mammal. As used herein, the term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, “contacting” the DGKα enzyme with a compound of the invention includes the administration of a compound of the present invention to a subject, such as a human, having DGKα, as well as, for example, introducing a of compound the invention into a sample containing a cellular or purified preparation containing the DGKα enzyme. The term “DGKα degrader” refers to an agent that targets the DGKα enzyme resulting in its degradation. The term “DGKα inhibitor” refers to an agent capable of inhibiting the enzymatic activity of diacylglycerol kinase alpha (DGKα) in T-cells resulting in enhanced T-cell stimulation. The DGKα inhibitor can be a reversible DGKα inhibitor. “A reversible DGKα inhibitor” is a compound that reversibly inhibits DGKα enzyme activity either at the catalytic site or at a non-catalytic site. The terms “patient,” “subject,” and “individual” are used interchangeably herein and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (including bovines, porcines, etc.), companion animals (e.g., canines, felines, etc.) and rodents (e.g., mice and rats). In embodiments, the subject is a human. As used herein, the term “selective inhibitors of DGKα” refers to a compound’s to selectively inhibit the activity of DGKα as compared to other diacylglycerol kinases (e.g., DGKζ). As used herein, the term “selective degraders of DGKα” refers to a compound’s to selectively degrade DGKα as compared to other diacylglycerol kinases (e.g., DGKζ). “Treating” a condition or patient, and “treatment” refer to taking steps to obtain beneficial or desired results, including clinical results, via administration of a compound or composition of the present invention. Beneficial or desired results include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total. “Treatment” also can mean prolonging survival as compared to expected survival if not receiving treatment. “Administering” or “administration of” a compound or a composition of the invention to a subject can be carried out using any of a variety of routes and methods known to those skilled in the art based on the directions of an attending healthcare provider. For example, a compound or pharmaceutical composition of the invention can be administered to a subject in need of such treatment by any of the following routes: intravenously, arterially, intradermally, intramuscularly, intraperitoneally, subcutaneously, ocularly, sublingually, buccally, orally (by ingestion), intranasally (by inhalation), intravaginally, intraspinally, intrathecally, intracerebrally, and transdermally (by absorption through the skin). A compound or composition of the present invention can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, delayed, or controlled release of the compound or composition. Administering also can be performed, for example, once, a plurality of times, and/or over one or more extended periods. Appropriate methods of administering a compound or pharmaceutical composition of the invention to the subject will also depend on a variety of factors such as, for example, age, weight, gender, and physical condition of the subject, as well as the chemical and biological properties of the compound or pharmaceutical composition (e.g., solubility, absorption, bioavailability, metabolism, stability, and toxicity). In embodiments, a compound or pharmaceutical composition of present invention is administered orally, e.g., to a subject by ingestion. In embodiments, the orally administered compound or pharmaceutical composition is in a controlled release (e.g., a delayed release, extended release, or slow release) formulation. As used herein, the phrase “conjoint administration” refers to any form of administration of two or more different therapeutic agents such that the second agent is administered while the previously administered therapeutic agent is still effective in the body (e.g., the two agents are simultaneously effective in the patient, which may include synergistic effects of the two agents). For example, the different therapeutic compounds can be administered either in the same formulation or in separate formulations, either simultaneously or sequentially. Thus, an individual who receives such treatment can benefit from a combined effect of different therapeutic agents. A “therapeutically effective amount” or a “therapeutically effective dose” of a compound or pharmaceutical composition of the invention is an amount of the drug or composition that, when administered to a subject, will have the intended therapeutic effect. The full therapeutic effect does not necessarily occur by administration of one dose and may occur only after administration of a series of doses. Thus, a therapeutically effective amount may be administered in one or more administrations. The effective amount needed for a subject will depend, for example, upon the subject’s age, weight, health, gender, and the nature and extent of the condition (e.g., cancer) being treated. The attending healthcare provider will generally determine the effective amount for a given situation according to these and other factors. The term “alkyl” refers to saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl-substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups. In preferred embodiments, a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g., C1–C6 for straight chains, C3–C6 for branched chains), and more preferably four or less carbons in its backbone. The term “alkoxy” refers to an alkyl group having an oxygen attached thereto, represented by -O-alkyl. For example, “C1–C4 alkoxy” includes methoxy, ethoxy, propoxy, isopropoxy, and butoxy. The term “Cx–Cy,” when used in conjunction with a chemical moiety (e.g, alkyl, alkenyl, alkoxy) is meant to include groups that contain from x to y carbons in the chain. A C1–C6 alkyl group, for example, contains from one to six carbon atoms in the chain; a C1–C4-alkyl contains from one to four carbon atoms in the chain. The term “aryl,” as used herein, includes substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon. Preferably the ring is a 5- to 7-membered ring. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings and wherein at least one of the rings is aromatic, i.e., the other cyclic rings can be selected from cycloalkyls, cycloalkenyls, aryls, heteroaryls, heterocyclyls, and combinations thereof. Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like. The term “carbocyclic ring,” as used herein, refers to a non-aromatic saturated or unsaturated ring in which each atom of the ring is carbon that is substituted or unsubstituted. Preferably, a carbocycle ring contains from 3 to 10 atoms, more preferably from 3 to 8 atoms, including 5 to 7 atoms, such as, for example, 5 or 6 atoms. The term “electron withdrawing group,” as used herein, means an atom or functional group that draws electron density from neighboring atoms towards itself, typically by resonance or inductive effects. Exemplary electron withdrawing groups include, but are not limited to, trifluoromethylsulfonyl groups (-SO2CF3), substituted or unsubstituted ammonium groups (-NR3+, wherein each R is H or alkyl), nitro groups (-NO2), sulfonic acid groups (-SO3H), sulfonyl groups (SO2R, wherein R is alkyl), cyano groups (-C≡N), haloformyl groups (-COX, wherein X is Cl, Br, or I), formyl groups (-CHO), acyl groups (-COR, wherein R is alkyl), carboxyl groups (-CO2H), alkoxycarboxyl groups (-CO2R, wherein R is alkyl), substituted or unsubstituted amino carbonyl groups (-CONH2, -CONHR, -CONR2, wherein R is alkyl), halogen groups, and nitroso groups (-N=O). The terms “halo,” “halogen,” and “halogen groups,” as used herein, refer to a substituent group from Group 17 of the periodic table of the elements and includes fluoro (-F), chloro (- Cl), bromo(-Br), and iodo (-I) substituent groups. The terms “haloalkyl” and “haloalkoxy” mean alkyl or alkoxy, respectively, substituted with one or more halogen atoms. The term “heteroaryl” or “heteroaromatic” includes substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The term “heteroaryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be selected from cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and heterocyclyls. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like. The term “heteroatom” as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur. The term “heterocyclic ring” refers to substituted or unsubstituted non-aromatic ring structure, preferably 3- to 10-membered rings, more preferably 3- to 7-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The term “heterocyclic ring” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be selected from cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and heterocyclyls. Heterocyclyl groups include, for example, diazinane, imidazolidine, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, octahydropyrrolo[3,4-c]pyrrole, and the like. The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched, and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. The term “aromatic ring” includes aryl and heteroaryl rings. The term “ring” includes aryl, cycloalkyl, heterocyclic, carbocyclic, and heteroaryl, ring systems. Certain compounds described herein may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. When a disclosed compound is named or depicted by structure with or without indicating stereochemistry, it is understood that the name or structure encompasses all possible stereoisomers, geometric isomers, including essentially pure stereo or geometric isomers, as well as combination thereof. When the stereochemical configuration at a chiral center in a compound having one or more chiral centers is depicted by its chemical name (e.g., where the configuration is indicated in the chemical name by “R” or “S”) or structure (e.g., the configuration is indicated by “wedge” bonds), the enrichment of the indicated configuration relative to the opposite configuration is greater than 50%, 60%, 70%, 80%, 90%, 99%, or 99.9%. “Enrichment of the indicated configuration relative to the opposite configuration” is a mole percent and is determined by dividing the number of compounds with the indicated stereochemical configuration at the chiral center(s) by the total number of all the compounds with the same or opposite stereochemical configuration in a mixture. When the stereochemical configuration at a chiral center in a compound having two or more chiral centers is depicted by its structure (e.g., the configuration is indicated by “wedge” bonds), and the configuration about the two or more chiral centers is labelled as “or1”, this means that compound comprises a mixture of stereoisomers and the stereochemistry is relative in the depicted configuration between the two or more stereocenters. For example, means that the compound comprises a mixture of and . When the stereochemical configuration at a chiral center in a compound having two or more chiral centers is depicted by its structure (e.g., the configuration is indicated by “wedge” bonds), and the configuration about the two or more chiral centers is labelled as “abs”, this means that the compound is stereochemically enriched as the drawn stereochemistry or as the opposite, i.e., is absolute of one or the other stereoisomers. For example,
Figure imgf000091_0001
means that the compound is
Figure imgf000091_0002
or
Figure imgf000091_0003
at an enrichment of the indicated configuration relative to the opposite configuration of greater than 50%, 60%, 70%, 80%, 90%, 99% or 99.9%. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. “Enantiomer” means one of a pair of molecules that are mirror images of each other and are not superimposable. Diastereomers are stereoisomers that contain two or more asymmetrically substituted carbon atoms. “Geometric isomers” are stereoisomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a carbocyclyl ring, or to a bridged bicyclic system. Enantiomeric and diastereomeric mixtures can be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral- phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and diastereomers can also be obtained from diastereomerically- or enantiomerically-pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods. Compounds of the Invention Disclosed herein are compounds that have activity as inhibitors of DGKα. In preferred embodiments, the disclosed compounds selectively inhibit DGKα. In embodiments, the disclosed compounds are selective inhibitors of DGKα over other diacylglycerol kinases (e.g., DGKζ). In embodiments, the disclosed compounds cause the degradation of DGKα. In embodiments, the disclosed compounds cause the selective degradation of DGKα. In embodiments, the disclosed compounds cause the selective degradation of DGKα over other diacylglycerol kinases (e.g., DGKζ). In embodiments, the disclosed compounds have desirable efficacy, stability, bioavailability, therapeutic index, and toxicity values that are important to their use as pharmaceuticals. In embodiments, disclosed herein is a compound of having a structure according to Formula (A):
Figure imgf000092_0001
(A) or a pharmaceutically acceptable salt thereof, wherein: Ring A is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaromatic ring fused to ring B; Ring C is a substituted or unsubstituted carbocyclic ring or a substituted or unsubstituted heterocyclic ring; R1 is alkyl (e.g., straight or branched C1–C6 alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, isobutyl, neopentyl, pentyl, hexyl)); R2 is H or an electron withdrawing functional group; each R3 is independently NR5R6; each R4 is independently alkyl (e.g., straight or branched C1–C6 alkyl (e.g., methyl, ethyl, propyl, isopropyl, butyl, isobutyl, neopentyl, pentyl, hexyl)) or alkoxy (e.g., C1–C6 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, neopentoxy, pentoxyl, hexoxy)), or one R3 and one R4, together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring; R5 and R6 are each, independently H, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or R5 and R6, together with the nitrogen to which they are attached, form a substituted or unsubstituted ring; and m and n are each, independently 0 or an integer greater than 0 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10). In embodiments of Formula (A), disclosed herein are compounds having the structure according to Formula (I): (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is a substituted or unsubstituted heteroaromatic ring fused to ring B Ring C is a substituted or unsubstituted heterocyclic ring; R1 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl); R2 is H or an electron withdrawing functional group; each R3 is independently NR5R6; each R4 is independently C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl) or C1–C4 alkoxy, or one R3 and one R4, together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring; R5 and R6 are each independently H, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or R5 and R6, together with the nitrogen to which they are attached, form a substituted or unsubstituted ring; m and n are each independently 0, 1, 2, 3, or 4. In embodiments, disclosed herein is a compound of having the structure according to Formula (I): (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is a 5- or 6-membered substituted or unsubstituted heteroaromatic ring fused to ring B; Ring C is a 5- to 8-membered substituted or unsubstituted heterocyclic ring; R1 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl); R2 is H or an electron withdrawing functional group; each R3 is independently NR5R6; each R4 is independently C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl) or C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy), or one R3 and one R4, together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring; R5 and R6 are each independently H, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or R5 and R6, together with the nitrogen to which they are attached, form a substituted or unsubstituted ring; and m and n are each independently 0, 1, 2, 3, or 4. In embodiments, disclosed herein is a compound of Formula (A) or Formula (I), wherein Ring A is selected from pyridine, imidazole, thiazole, oxazole, pyrazole, and isoxazole. In embodiments, disclosed herein is a compound of Formula (A) or Formula (I), wherein Ring A is selected from piperidine, diazinane, octahydropyrrolo[3,4-c]pyrrole, imidazolidine, and pyrrolidine. In embodiments, disclosed herein is a compound of Formula (A) or Formula (I), having a structure according to Formula (IIa): (IIa) or a pharmaceutically acceptable salt thereof, wherein Y is S or CH or CR33; R9 is H, halogen, cyano (-CN), C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), C1–C4 haloalkyl (e.g., fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, chloroethyl, dichloroethyl, trichloroethyl, fluoropropyl, difluoropropyl, trifluoropropyl, chloropropyl, dichloropropyl, trichloropropyl), C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy), or NR9a, wherein R9a is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl); and R33 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), C1–C4 haloalkyl (e.g., fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, chloroethyl, dichloroethyl, trichloroethyl, fluoropropyl, difluoropropyl, trifluoropropyl, chloropropyl, dichloropropyl, trichloropropyl), or C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy),. In certain embodiments, R9 is straight or branched C1–C4 alkyl. For example, in particular embodiments, R9 is methyl, ethyl, propyl, or butyl. In specific embodiments, R9 is methyl. In embodiments, disclosed herein is a compound of Formula (A) or Formula (I), having a structure according to Formula (II): (II) or a pharmaceutically acceptable salt thereof, wherein R9 is H, halogen, cyano (-CN), C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl such as methyl, ethyl, propyl, isopropyl, butyl), C1–C4 haloalkyl (e.g., fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, fluoroethyl, difluoroethyl, trifluoroethyl, chloroethyl, dichloroethyl, trichloroethyl, fluoropropyl, difluoropropyl, trifluoropropyl, chloropropyl, dichloropropyl, trichloropropyl), C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy), or NR9a, wherein R9a is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl). In embodiments, R9 is straight or branched C1–C4 alkyl. For example, in particular embodiments, R9 is methyl, ethyl, propyl, or butyl. In embodiments of Formula (II), R9 is methyl. In embodiments, disclosed herein is a compound of Formula (A) or Formula (I), having the structure according to Formula (III): (III) or a pharmaceutically acceptable salt thereof, wherein R10 is H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), carbocycle (e.g., C3– C7-cycloalkyl), or an electron withdrawing functional group; and q is 1 or 2. In embodiments of Formula (III), R10 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl) or an electron withdrawing functional group. In embodiments, R10 is methyl, ethyl, propyl, isopropyl, butyl. In embodiments of Formula III), R10 is methyl. In embodiments, disclosed herein is a compound of Formula (III), having the structure of Formula (IIIa): (IIIa) or a pharmaceutically acceptable salt thereof. In embodiments of Formula (IIIa), R10 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, or butyl). In embodiments of Formula (IIIa), R10 is straight or branched C1–C4 alkyl. In embodiments of Formula (IIIa), R10 is methyl (-CH3). In embodiments, R10 is selected from halogen (e.g., -F, -Cl), -CH3, -SO2CF3, -N(R11)2, -NO2, -SO2R12, -C≡N, -C(R13)3, -COR14, -CSR15, -CO2R16, -CON(R17)2, and -N=O, wherein: each of R11, R12, R14, R15, R16, and R17 is independently H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl); and each R13 is halogen (e.g., -F, -Cl). In embodiments, R10 is -Cl. Alternatively, R10 is -F. In embodiments, R10 is -C≡N. In embodiments, R10 is -N(R11)2, -SO2R12, or -C(R13)3, -CSR15, wherein each of R11, R12, and R15 is independently H or methyl. In embodiments, R10 is -COR14, wherein R14 is H or methyl. In embodiments, R10 is -CHO. In embodiments, R10 is -CO(CH3). In embodiments, R10 is -CO2(R16), wherein R16 is H or methyl. In embodiments, R10 is CO2(CH3) In embodiments, R10 is -CON(R17)2, wherein each R17 is independently H or methyl. In embodiments, R10 is -CONH(CH3). In embodiments, R10 is -CON(CH3)2. In embodiments, R10 is -N=O. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R1 is straight or branched C1–C4 alkyl. In embodiments of any one of Formulae (A), (I), (II), (III), or (IIIa), R1 is methyl, ethyl, propyl, or butyl. In embodiments of Formulae (A), (I), (II), (III), or (IIIa), wherein R1 is methyl. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R2 is selected from H, halogen, -SO2CF3, - N(R32)2, -NO2, -SO2R26, -C≡N, -C(R27)3, -COR28, -CSR29, -CO2R30, -CON(R31)2, and -N=O, wherein: each of R32, R26, R28, R29, R30, and R31 is independently H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl); and each R33 is halogen (e.g., -F, -Cl). In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R2 is H. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R2 is -C≡N. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein R2 is -N(R32)2, -SO2R26, -C(R33)3, -COR28, - CSR29, -CO2R30, or -CON(R31)2, wherein each of R32, R26, R28, R29, R30, and R31 is independently H or methyl; and each R33 is halogen. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is further substituted by one or more substituents selected from R15, R18, R19, R20, R21, and R22. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), ring C is further substituted by one or more substituents selected from R15, R18, R19, R20, R21, and R22 wherein: R15 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R18 and R19 are each, independently C1–C4 alkoxy, C1–C4 alkyl, or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R20 is straight or branched C1–C4 alkyl, or an electron withdrawing group, or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring; R21 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl; R22 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and R23 and R24 are each, independently straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R4 is straight or branched C1–C4 alkyl. In embodiments of any one of the compounds of Formulae (I), (II), (III), and (IIIa), R4 is C1– C4 alkoxy. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from
Figure imgf000100_0001
and
Figure imgf000100_0002
, wherein: R15 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R18 and R19 are each, independently H, OH, C1–C4 alkoxy, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring (e.g., a 5- or 6-membered substituted or unsubstituted carbocyclic ring or a 5- or 6-membered substituted or unsubstituted heterocyclic ring); R20 is H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy), or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a ring (e.g., 5-, 6-, or 7-membered substituted or unsubstituted carbocyclic ring or a 5-, 6-, or 7-membered substituted or unsubstituted heterocyclic ring); R21 is H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R23 and R24 are each, independently H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and indicates the point of attachment of ring C to ring B. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from
Figure imgf000101_0001
; ; ; and
Figure imgf000101_0002
, wherein: R15 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R18 and R19 are each, independently OH, C1–C4 alkoxy, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring (e.g., a 5- or 6-membered substituted or unsubstituted carbocyclic ring or a 5- or 6-membered substituted or unsubstituted heterocyclic ring); R20 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, isopropoxy, butoxy), or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a ring (e.g., 5-, 6-, or 7-membered substituted or unsubstituted carbocyclic ring or a 5-, 6-, or 7-membered substituted or unsubstituted heterocyclic ring); R21 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R23 and R24 are each, independently H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and indicates the point of attachment of ring C to ring B. In embodiments of Ring C, any one of R32, R26, R28, R29, R30, or R31 is straight or branched C1–C4 alkyl. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from ; and
Figure imgf000102_0002
,
Figure imgf000102_0001
wherein: R18 and R19 are each, independently H, OH, C1–C4 alkoxy, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring (e.g., a 5- or 6-membered substituted or unsubstituted carbocyclic ring or a 5- or 6-membered substituted or unsubstituted heterocyclic ring); R20 is H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), or R18 and R20 , or R19 and R20, together with the carbon atoms to which they are attached, form a ring (e.g., 5-, 6-, or 7-membered substituted or unsubstituted carbocyclic ring or a 5-, 6-, or 7-membered substituted or unsubstituted heterocyclic ring); R21 is H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), aryl, or heteroaryl; R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R23 and R24 are each, independently H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R15 is C1–C4 alkyl e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and indicates the point of attachment of ring C to ring B. In embodiments of Ring C, R21 is straight or branched C1–C4 alkyl. In embodiments of Ring C, R22 is straight or branched C1–C4 alkyl. In embodiments of Ring C, R15 is unsubstituted C1–C4 alkyl. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is 18
Figure imgf000103_0001
, wherein R and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted 5- or 6-membered carbocyclic ring or a substituted or unsubstituted 5- or 6- membered heterocyclic ring. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is 18
Figure imgf000104_0001
, wherein R and R20, or R19 and R20, together with the carbon atoms to which they are attached form a substituted or unsubstituted 5-, 6-, or 7-membered carbocyclic ring or a substituted or unsubstituted 5-, 6-, or 7-membered heterocyclic ring. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from
Figure imgf000104_0002
; ; ; ; ; ; ;
Figure imgf000104_0003
; and
Figure imgf000104_0004
. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is , wherein R21 is , wherein R25 is selected from OH, halogen (-Cl, -F), C1–C4 alkyl (e.g., straight or branched C1– C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), C1–C4 haloalkyl (e.g., -CH2F, -CHF2, -CF3, -CH2CH2F, - CH2CHF2, -CH2CF3, -(CH2)2CH2F, -(CH2)2CHF2, -(CH2)2CF3, -(CH2)3CH2F, -(CH2)3CHF2, -(CH2)3CF3), C1–C4 haloalkoxy (e.g., -OCH2F, -OCHF2, -OCF3, -OCH2CH2F, -OCH2CHF2, -OCH2CF3, -O(CH2)2CH2F, -O(CH2)2CHF2, -O(CH2)2CF3, -O(CH2)3CH2F, -O(CH2)3CHF2, -O(CH2)3CF3), substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R21 to N. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is
Figure imgf000105_0001
R , wherein R21 is
Figure imgf000105_0002
, wherein Each R25 is independently selected from OH, halogen (-Cl, -F), C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), C1–C4 haloalkyl (e.g., -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -(CH2)2CH2F, -(CH2)2CHF2, -(CH2)2CF3, -(CH2)3CH2F, -(CH2)3CHF2, -(CH2)3CF3), C1–C4 haloalkoxy (e.g., -OCH2F, -OCHF2, -OCF3, -OCH2CH2F, -OCH2CHF2, -OCH2CF3, -O(CH2)2CH2F, -O(CH2)2CHF2, -O(CH2)2CF3, -O(CH2)3CH2F, -O(CH2)3CHF2, -O(CH2)3CF3), substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R21 to N. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is
Figure imgf000106_0001
, wherein R21 is selected from
Figure imgf000106_0002
; ; ; ; ; ; and
Figure imgf000106_0003
In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is 21
Figure imgf000106_0004
, wherein R is selected from
Figure imgf000106_0005
; ; ; ; ; ; and
Figure imgf000106_0006
.
In embodiments, disclosed herein is a compound having the structure according to any one of Formula (A), (I), (II), (III), or (IIIa), wherein ring C is selected from
Figure imgf000107_0001
; ; ; ; ;
Figure imgf000107_0002
; ; ; ; ; ;
Figure imgf000107_0003
; ; ; ; ; and
Figure imgf000107_0004
. In embodiments of Ring C, R22 is straight or branched C1–C4 alkyl. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is , wherein R18 and R19 are each independently selected from -H, -OH, alkyl (e.g., straight or branched C1–C4 alkyl) and , wherein: R26 is H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such methyl, ethyl, propyl, isopropyl, butyl), halogen (-Cl, -F), C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), or C1–C4 haloalkyl (e.g., -CH2F, -CHF2, -CF3, -CH2CH2F, - CH2CHF2, -CH2CF3, -(CH2)2CH2F, -(CH2)2CHF2, -(CH2)2CF3, -(CH2)3CH2F, - (CH2)3CHF2, -(CH2)3CF3), C1–C4 haloalkoxy (e.g., -OCH2F, -OCHF2, -OCF3, - OCH2CH2F, -OCH2CHF2, -OCH2CF3, -O(CH2)2CH2F, -O(CH2)2CHF2, -O(CH2)2CF3, -O(CH2)3CH2F, -O(CH2)3CHF2, -O(CH2)3CF3), or two adjacent R27 groups, and the carbon atoms two which they are attached, form a 5- or 6-membered substituted or unsubstituted ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R18 and R19 to ring C. In embodiments, disclosed herein is a compound having the structure according to any one of N Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is R18 R19 ; ; ; ; ; ; ; ; ; ; ; ; or , wherein R18 and R19 are each independently selected from -H, -OH, alkyl (e.g., straight or branched C1–C4 alkyl), and , wherein: R26 is H, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), halogen (-Cl, -F), C1–C4 alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy), or C1–C4 haloalkyl (e.g., -CH2F, -CHF2, -CF3, -CH2CH2F, - CH2CHF2, -CH2CF3, -(CH2)2CH2F, -(CH2)2CHF2, -(CH2)2CF3, -(CH2)3CH2F, - (CH2)3CHF2, -(CH2)3CF3), C1–C4 haloalkoxy (e.g., -OCH2F, -OCHF2, -OCF3, - OCH2CH2F, -OCH2CHF2, -OCH2CF3, -O(CH2)2CH2F, -O(CH2)2CHF2, -O(CH2)2CF3, -O(CH2)3CH2F, -O(CH2)3CHF2, -O(CH2)3CF3), or two adjacent R27 groups, and the carbon atoms two which they are attached, form a 5- or 6-membered substituted or unsubstituted ring; and q is 0, 1, or 2. In embodiments of Ring C, R26 is straight or branched C1–C4 alkyl. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ;
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from ;
Figure imgf000114_0001
; ; In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is
Figure imgf000114_0002
; ; or
Figure imgf000114_0003
Figure imgf000115_0001
, wherein R18 and R19, together with the carbon atom to which they are attached, form
Figure imgf000115_0002
, wherein: X and Y are each, independently CH2, NR29, O, or C=O; R28 is -OH, halogen (-Cl, -F), C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), C1–C4 haloalkyl (e.g., -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -(CH2)2CH2F, -(CH2)2CHF2, -(CH2)2CF3, - (CH2)3CH2F, -(CH2)3CHF2, -(CH2)3CF3), C1–C4 alkyoxy (e.g., methoxy, ethoxy, propoxy, butoxy), or C1–C4 haloalkoxy (e.g., -OCH2F, -OCHF2, -OCF3, -OCH2CH2F, -OCH2CHF2, -OCH2CF3, -O(CH2)2CH2F, -O(CH2)2CHF2, -O(CH2)2CF3, - O(CH2)3CH2F, -O(CH2)3CHF2, -O(CH2)3CF3); or R29 is H or C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl); and w is 0, 1, or 2. In embodiments of Ring C, R29 is straight or branched C1–C4 alkyl.
In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from
Figure imgf000116_0001
Figure imgf000116_0002
; and
Figure imgf000116_0003
. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is selected from
Figure imgf000116_0004
; ; ; ; and
Figure imgf000116_0005
, wherein R34 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl). In embodiments of Ring C, R34 is straight or branched C1–C4 alkyl. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is , 32
Figure imgf000116_0006
wherein R is
Figure imgf000116_0007
; ; ; or
Figure imgf000116_0008
. In embodiments, the disclosed herein is a compound having the structure of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is
Figure imgf000117_0001
; ; ;
Figure imgf000117_0002
; ; ; ; ; ;
Figure imgf000117_0003
; ; ; or
Figure imgf000117_0004
, wherein R22 is C1–C4 alkyl (e.g., straight or branched C1–C4 alkyl, such as methyl, ethyl, propyl, isopropyl, butyl), substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. In embodiments of Ring C, R22 is straight or branched C1–C4 alkyl. In embodiments, the disclosed herein is a compound having the structure of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C selected from
Figure imgf000117_0005
; ; ; ; ; ;
Figure imgf000117_0006
; ; ; ; ; ;
Figure imgf000118_0001
; ; ; ; ; and
Figure imgf000118_0002
. In embodiments, disclosed herein is a compound having the structure according to any one of Formulae (A), (I), (II), (III), or (IIIa), wherein ring C is or
Figure imgf000118_0003
Figure imgf000118_0004
In embodiments, disclosed herein is a compound having a chemical formula according to any one of Compounds 1–234, as shown in Table 1. Table 1.
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
176 177 178
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
or a pharmaceutically acceptable salt thereof. In embodiments, disclosed herein is a compound having a chemical formula according to any one of Compounds 235–311, as shown in Table 2. Table 2.
Figure imgf000184_0002
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
or a pharmaceutical salt thereof.
Example 312: Exemplary Synthesis of 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl 5-[5-(trifluoromethoxy)- 2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate
Figure imgf000208_0001
To a solution of 2-bromo-5-(trifluoromethoxy)pyridine (2.67 g, 11.05 mmol, 1 eq) and tert- butyl 2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine-1-carboxylate (2.50 g, 11.05 mmol, 1 eq) in dioxane (25 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H- imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (1.07 g, 1.10 mmol, 0.1 eq) and cesium carbonate (10.80 g, 33.14 mmol, 3 eq). The mixture was stirred at 100 °C for 3 h. To the reaction mixture was added water (30 mL) and extracted with ethyl acetate 60 mL (30 mL x 2). The combined organic layers were washed with saturated sodium chloride (30 mL x 2), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=100/1 to 5/1) to give tert-butyl 5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (3.74 g, 9.65 mmol, 87% yield) as a yellow oil. LC/MS: (ESI) m/z: 388.1 [M+1] +.
Step 2: Preparation of tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate and tert-butyl (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-1-carboxylate Racemic tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-1-carboxylate (1.9 g, 4.90 mmol, 1 eq) was purified by SFC (column: REGIS(S,S)WHELK-O1(250mm x 25mm,10um); mobile phase: [neutral-ethyl alcohol]; 15%-15%, 200min) to afford tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2- pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (870 mg, 2.25 mmol, 45% yield) as a green oil (SFC:Rt=1.061 min) and tert-butyl (3aS,7aR)-5-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1- carboxylate (830 mg, 2.14 mmol, 43% yield)) as a green oil (SFC: Rt = 1.209 min). The absolute stereochemical configuration was tentatively assigned. Step 3: Preparation of WC-ARV-JM-047-A-2a, (3aR,7aS)-5-[5-(trifluoromethoxy)-2- pyridyl]-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine To a solution of tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (500 mg, 1.29 mmol, 1 eq) in dichloromethane (5 mL) was added trifluoroacetic acid (7.70 g, 67.53 mmol, 5.00 mL, 52.32 eq). The mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a- octahydropyrrolo[3,2-c]pyridine (517 mg, crude, trifluoroacetic acid) as a yellow solid. LC/MS: MS (ESI) m/z: 288.3 [M+1] +. Step 4: Preparation of 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile To a solution of (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a- octahydropyrrolo[3,2-c]pyridine (517 mg, 1.29 mmol, 1 eq, trifluoroacetic acid) and 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (327 mg, 1.29 mmol, 1 eq) in acetonitrile (5 mL) was added diisopropylethylamine (833 mg, 6.44 mmol, 1.12 mL, 5 eq). The mixture was stirred at 25 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 x 25mm x 10um; mobile phase: [water (formic acid)- acetonitrile]; 58%-88%, 58min) to give 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (465.7 mg, 0.89 mmol, 69% yield, 96% purity) as a yellow solid. LC/MS: (ESI) m/z: 505.2507.3, 505.3 [M+1] +.1H NMR: (400 MHz, DMSO-d6) δ: 8.13 (d, J = 2.4 Hz, 1H), 8.02 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.58 (dd, J = 2.4, 9.2 Hz, 1H), 6.88 (d, J = 9.6 Hz, 1H), 5.54 - 5.42 (m, 1H), 4.46 - 4.34 (m, 1H), 3.99 - 3.89 (m, 2H), 3.84 - 3.75 (m, 1H), 3.63 (dd, J = 4.4, 13.6 Hz, 1H), 3.50 (s, 3H), 3.21 - 3.13 (m, 1H), 2.57 (d, J = 6.4 Hz, 1H), 2.24 - 2.15 (m, 1H), 2.08 - 1.99 (m, 1H), 1.96 - 1.86 (m, 1H), 1.81 - 1.71 (m, 1H) In embodiments, disclosed herein is a compound having a chemical formula according to any one of Compounds 312–390, as shown in Table 3. These compounds were made using a similar synthesis as is described for Compound 312, which is discussed in more detail below.
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
In embodiments, disclosed herein is a compound having a chemical formula according to any one of Compounds 391-449, as shown in Table 4. These compounds were made using a similar synthesis as is desribed for Compounds 397, 416, and 429, which is discussed in more detail below.
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Pharmaceutical Compositions Disclosed herein are pharmaceutical composition comprising a compound of the invention as defined hereinbefore, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable excipients. The compositions of the invention may be in a form suitable for oral use (for example, as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups, or elixirs), for topical use (for example, as creams, ointments, gels, or aqueous or oily solutions, suspensions, or transdermal patches), for administration by inhalation (for example, as a finely divided powder or a liquid aerosol), for administration by insufflation (for example, as a finely divided powder), or for parenteral administration (for example, as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular, intraperitoneal, or as a suppository for rectal or intravaginal dosing). The pharmaceutical compositions of the invention can be obtained by conventional procedures in view of this disclosure using conventional pharmaceutical excipients well- known in the art. Thus, compositions intended for oral use may contain, for example, one or more coloring, sweetening, flavoring, and/or preservative agents. Pharmaceutically acceptable excipients that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) (e.g., d-alpha-tocopherol, polyethyleneglycol 1000 succinate), surfactants used in pharmaceutical dosage forms (e.g., tweens), polyethoxylated castor oil (e.g., CREMOPHOR surfactant (BASF)) or other similar polymeric delivery matrices, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts), colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins (e.g., alpha-, beta-, and gamma-cyclodextrin), chemically modified derivatives thereof (e.g., hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-cyclodextrins), or other solubilized derivatives also may be advantageously used to enhance delivery of compounds of the compounds described herein. The amount of the compound(s) that can be administered and the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention depends on a variety of factors, including the age, weight, sex, the medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. Thus, the dosage regimen can vary widely, but may be routinely determined using standard methods. The daily dose can be administered in one to four doses divided per day. Other dosing schedules include one dose per week and one dose per two-day cycle. Methods of Treatment The compounds of the invention as defined hereinbefore, or a pharmaceutically acceptable salt thereof, are useful for the treatment of cancer. In embodiments, disclosed herein the compounds of the invention, or a pharmaceutically acceptable salt thereof, can be used in the treatment of diseases or disorders associated with DGK target inhibition in T-cells. In some such embodiments, the compound of the invention is prepared in combination with one or more additional therapeutic agents for conjoint administration for treating diseases or disorders associated with DGK target inhibition in T- cells. Also provided herein are methods for treating a subject suffering from or susceptible to a medical condition that is associated with DGK target inhibition in T-cells. For example, the compounds described herein may be used to treat or prevent viral infections and proliferative diseases such as cancer. More specifically, disease or conditions that are associated with DGK target inhibition in T cells include viral and other infections (e.g., skin infections, GI infection, urinary tract infections, genito-urinary infections, systemic infections), and proliferative diseases (e.g., cancer). In embodiments, the subject is being treated for cancer. Types of cancers that may be treated with a compound of the invention include, but are not limited to, brain cancers, skin cancers, bladder cancers, ovarian cancers, breast cancers, gastric cancers, pancreatic cancers, prostate cancers, colon cancers, blood cancers, lung cancers and bone cancers. Examples of such cancer types include neuroblastoma, intestine carcinoma such as rectum carcinoma, colon carcinoma, familiar adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, esophageal carcinoma, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, renal carcinoma, kidney parenchymal carcinoma, ovarian carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, pancreatic carcinoma, prostate carcinoma, testis carcinoma, breast carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, Hodgkin lymphoma, non-Hodgkin lymphoma, Burkitt lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gall bladder carcinoma, bronchial carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, choroid melanoma, seminoma, rhabdomyosarcoma, craniopharyngioma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma and plasmocytoma. In preferred embodiments, the compounds of the invention treat colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma. In embodiments, the compounds of the invention can inhibit activity of the diacylglycerol kinase alpha (DGKα). For example, the compounds of the invention can be used to inhibit activity of DGKα in a cell or in a subject in need of modulation of DGKα by administering a therapeutically effective amount of a compound of the invention or a salt thereof. In embodiments, the compounds of the invention can cause the degradation of DGKα. For example, the compounds of the invention can be used to degrade DGKα in a cell or in a subject in need of said treatment by administering a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof. Also disclosed herein are methods of treating diseases or conditions associated with activity or expression, including abnormal activity and/or overexpression, of DGKαin a subject by administering to the subject in need of such treatment a therapeutically effective amount or dose of any one or more compounds of the invention or a pharmaceutical composition thereof. Exemplary diseases can include any disease, disorder, or condition that is directly or indirectly linked to expression or activity of the DGKαenzyme, such as over expression or abnormal activity. A DGKα-associated disease can also include any disease, disorder, or condition that can be prevented, ameliorated, or cured by modulating the DGKα enzyme activity. Examples of DGKα associated diseases include cancer and viral infections such as HIV infection, hepatitis B, and hepatitis C. Combination Therapies In embodiments, the compounds of the invention may be conjointly administered with an additional anticancer, such as an immuno-oncology agent. In certain embodiments, the compounds of the invention are administered concurrently with the additional anticancer agent. Alternatively, the compounds of the invention may be co-formulated with the anticancer agent. Exemplary anticancer agents include, but are not limited to, a small molecule drugs, antibodies, or other biologics or small molecules. Exemplary biologics include, but are not limited to, cancer vaccines, antibodies, and cytokines. In embodiments, the antibody is a monoclonal antibody. In some such embodiments, the monoclonal antibody is humanized or human. In embodiments, the anticancer agent is an immuno-oncology agent. In some such embodiments, the immuno-oncology agent is (i) an agonist of a stimulatory (including a co- stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co-inhibitory) signal on T cells, both of which result in amplifying antigen-specific T-cell responses (often referred to as immune checkpoint regulators). Certain stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7- DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD4OL, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4- IBB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DRS, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTOR, LIGHT, DcR3, HVEM, VEGUTL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin α/TNFβ, TNFR2, TNFα, LTβR, Lymphotoxin a 1β2, FAS, FASL, RELT, DR6, TROY, NGFR. In embodiments, T-cell responses can be stimulated by a combination of a compound of the invention and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-IBB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX4OL, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H. Other agents that can be combined with the compounds of the invention for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, the compounds of the invention can be combined with antagonists of KIR, such as lirilumab. Yet other agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (e.g., International Patent Publication Nos. WO11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044) or FPA-008 (e.g., International Patent Publication Nos. WO11/140249; WO13169264; WO14/036357). In embodiments, the compounds of the invention can be used with one or more of agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD-1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites. In embodiments, the additional anticancer agent is a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody. Suitable CTLA-4 antibodies include, for example, YERVOY (ipilimumab), or tremelimumab. In embodiments, the additional anticancer agent is a PD-1 antagonist, such as an antagonistic PD-1 antibody. Suitable PD-1 antibodies include, for example, OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; e.g., International Patent Publication No. WO2012/145493). The immuno-oncology agent may also include pidilizumab (CT-011), though its specificity for PD-1 binding has been questioned. Another approach to target the PD-1 receptor is the recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgG1, called AMP-224. In embodiments, the additional anticancer agent is a PD-L1 antagonist, such as an antagonistic PD-L1 antibody. Suitable PD-L1 antibodies include, for example, MPDL3280A (RG7446; e.g., International Patent Publication No. WO2010/077634), durvalumab (MEDI4736), BMS-936559 (e.g., International Patent Publication No. WO2007/005874), and MSB0010718C (e.g., International Patent Publication No. WO2013/79174). In embodiments, the additional anticancer agent is a LAG-3 antagonist, such as an antagonistic LAG-3 antibody. Suitable LAG3 antibodies include, for example, BMS-986016 (e.g., International Patent Publication Nos. WO10/19570, WO14/08218), or IMP-731 or IMP-321 (e.g., International Patent Publication Nos. WO08/132601, WO09/44273). In embodiments, the additional anticancer agent is a CD137 (4-1BB) agonist, such as an agonistic CD137 antibody. Suitable CD137 antibodies include, for example, urelumab, and PF-05082566 (e.g., International Patent Publication No. WO12/32433). In embodiments, the additional anticancer agent is a GITR agonist, such as an agonistic GITR antibody. Suitable GITR antibodies include, for example, BMS-986153, BMS-986156, TRX- 518 (e.g., International Patent Publication Nos. WO06/105021, WO09/009116), and MK- 4166 (e.g., International Patent Publication No. WO11/028683). In embodiments, the additional anticancer agent is an IDO antagonist. Suitable IDO antagonists include, for example, INCB-024360 e.g., International Patent Publication Nos. (WO2006/122150, WO07/75598, WO08/36653, WO08/36642), indoximod, BMS-986205, or NLG-919 (e.g., International Patent Publication No. WO09/73620, WO09/1156652, WO11/56652, WO12/142237). In embodiments, the additional anticancer agent is an OX40 agonist, such as an agonistic OX40 antibody. Suitable OX40 antibodies include, for example, MEDI-6383 or MEDI-6469. In embodiments, the additional anticancer agent is an OX4OL antagonist, such as an antagonistic OX40 antibody. Suitable OX4OL antagonists include, for example, RG-7888 (e.g., International Patent Publication No. WO06/029879). In embodiments, the additional anticancer agent is a CD40 agonist, such as an agonistic CD40 antibody. In yet another embodiment, the anticancer agent is a CD40 antagonist, such as an antagonistic CD40 antibody. Suitable CD40 antibodies include, for example, lucatumumab or dacetuzumab. In embodiments, the additional anticancer agent agent is a CD27 agonist, such as an agonistic CD27 antibody. Suitable CD27 antibodies include, for example, varlilumab. In embodiments, the additional anticancer agent is MGA271 (to B7H3) (e.g., International Patent Publication No. WO11/109400). Combination therapies, as disclosed herein, are intended to embrace conjoint administration of these therapeutic agents; for example, administration of said therapeutic agents in a sequential manner, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be affected by any appropriate route including, but not limited to, oral routes, parental routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally. Alternatively, for example, all therapeutic agents may be administered orally, or both therapeutic agents may be administered by parentally, e.g., by intravenous injection. Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g., surgery or radiation treatment). Where the combination therapy further comprises a non-drug treatment, the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks. One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g., IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compounds of the invention for treatment of DGKα associated diseases, disorders, or conditions. The agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms. Suitable additional anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine, cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide. In embodiments, in the treatment of cancer (e.g., melanoma) suitable additional agents for use in combination with the compounds of the invention include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the “Dartmouth regimen,” which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM. The compounds of the invention also can be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF). In embodiments, the compounds of the invention also can be used in combination with vaccine therapy in the treatment of cancer (e.g., melanoma). Antimelanoma vaccines are, in some ways, similar to the anti-virus vaccines that are used to prevent diseases caused by viruses such as polio, measles, and mumps. Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body’s immune system to destroy melanoma cells. Suitable additional anti-cancer agents also include, for example, anti-metabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6- mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine. Suitable additional anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxyco-formycin, mitomycin-C, L-asparaginase, interferons (especially IFN- a), etoposide, and teniposide. Other cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, and droloxafine. Also suitable are cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors. Other additional anti-cancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-10 or TGF-β). Other additional anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4. Other additional anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T-cell transfer. Additional anti-cancer agents also include anti-cancer vaccines, such as, for example, dendritic cells, synthetic peptides, DNA vaccines, and recombinant viruses. The treatment methods of the invention may optionally include conjointly administering at least one signal transduction inhibitor (STI). A “signal transduction inhibitor” is an agent that selectively inhibits one or more vital steps in signaling pathways, in the normal function of cancer cells, thereby leading to apoptosis. Suitable STIs include, but are not limited to: (i) bcr/abl kinase inhibitors such as, for example, STI 571 (GLEEVEC®); (ii) epidermal growth factor (EGF) receptor inhibitors such as, for example, kinase inhibitors ORES SA®, SSI-774) and antibodies (Imclone: C225 [Goldstein et al. Clin. Cancer Res., 1:1311–18 (1995)], and Abgenix: ABX-EGF); (iii) her-2/neu receptor inhibitors such as farnesyl transferase inhibitors (FTI) such as, for example, L-744,832 (Kohl et al. Nat. Med., 1(8):792–97 (1995)); (iv) inhibitors of Akt family kinases or the Akt pathway, such as, for example, rapamycin (see, for example, Sekulic et al. Cancer Res., 60:3504–13 (2000)); (v) cell cycle kinase inhibitors such as, for example, flavopiridol and UCN-01 (see, for example, Sausville Curr. Med. Chem. Anti-Canc. Agents, 3:47–56 (2003)); and (vi) phosphatidyl inositol kinase inhibitors such as, for example, LY294002 (see, for example, Vlahos et al. J. Biol. Chem., 269:5241–48 (1994)). Alternatively, at least one STI and at least one compound of Formula (I) may be in separate pharmaceutical compositions. In specific embodiments of the present invention, at least one compound of the invention and at least one STI may be administered to the patient conjointly. In other words, at least one compound of the invention may be administered first or at least one STI may be administered first and the other is administered next; or at least one compound of the invention and at least one STI may be administered at the same time. Additionally, when more than one compound of invention and/or STI is used, the compounds may be administered in any order. Also disclosed herein are pharmaceutical compositions for the treatment of a chronic viral infections in a subject comprising administering a therapeutically effective amount of at least one compound of the invention, optionally, at least one chemotherapeutic drug, and, optionally, at least one antiviral agent, in a pharmaceutically acceptable carrier. In embodiments, one or more compounds of the invention, one or more chemotherapeutic drugs, and/or one or more antiviral agents are administered conjointly. For example, in embodiments, at least one compound of the invention may be administered first or at least one chemotherapeutic agent may be administered first. Alternatively, at least one compound of the invention and the at least one STI may be administered at the same time. Additionally, when more than one compound of the invention and/or chemotherapeutic agent is used, the compounds may be administered in any order. Similarly, any antiviral agent or STI may also be administered at any point in relation to the administration of the compound of the invention. Chronic viral infections that may be treated using the present combinatorial treatment include, but are not limited to, diseases caused by hepatitis C virus (HCV), human papilloma virus (HPV), cytomegalovirus (CMV), herpes simplex virus (HSV), Epstein-Barr virus (EBV), varicella zoster virus, coxsackie virus, human immunodeficiency virus (HIV). Notably, parasitic infections (e.g., malaria) may also be treated by the above methods wherein compounds known to treat the parasitic conditions are optionally added in place of the antiviral agents. Suitable antiviral agents contemplated for use in combination with the compound of Formula (I) can comprise nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), non- nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors and other antiviral drugs. Examples of suitable NRTIs include zidovudine (AZT); didanosine (ddl); zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(P0M)- PMEA]; lobucavir; BCH-I0652; emitricitabine [(-)-FTC]; beta-L-FD4 (also called beta-L- D4C and named beta-L-2′,3′-dicleoxy-5-fluoro-cytidene); DAPD, ((−)-beta-D-2,6-diamino- purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI- RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721; AG-1549; MKC-442 (1-(ethoxy-methyl)-5-(1-methylethyl)-6-(phenylmethyl)-(2,4(1H,3H)- pyrimidinedione); and (+)-calanolide A (NSC-675451) and B. Typical suitable protease inhibitors include saquinavir (Ro 31-8959); ritonavir (ABT-538); indinavir (MK-639); nelfnavir (AG-1343); amprenavir (141W94); lasinavir; DMP-450; BMS-2322623; ABT-378; and AG-1549. Other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and Yissum Project No.11607. Also disclosed herein are pharmaceutical kits useful, for example, in the treatment or prevention of DGKα-associated diseases or disorders referred to herein, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit. Methods of Preparation The compounds of the present invention may be synthesized by many methods available to those skilled in the art in view of the present disclosure. General synthetic schemes for preparing compounds of the present invention are described below. These schemes are illustrative and are not meant to limit the possible techniques one skilled in the art may use to prepare the compounds disclosed herein. Different methods to prepare the compounds of the present invention will be evident to those skilled in the art. Examples of compounds of the present invention prepared by methods described in the general schemes are given in the Examples section set out hereinafter. Preparation of homochiral examples may be carried out by techniques known to one skilled in the art. For example, homochiral compounds may be prepared by separation of racemic products or diastereomers by chiral phase preparative HPLC. Alternatively, the example compounds may be prepared by methods known to give enantiomerically or diastereomerically enriched products. The reactions and techniques described in this section are performed in solvents appropriate to the reagents and materials employed and are suitable for the transformations being effected. Also, in the description of the synthetic methods given below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and work-up procedures, are chosen to be the conditions standard for that reaction, which should be readily recognized by one skilled in the art. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule must be compatible with the reagents and reactions proposed. Such restrictions to the substituents that are compatible with the reaction conditions will be readily apparent to one skilled in the art, with alternatives required when incompatible substituents are present. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a compound of the invention. It will also be recognized that another major consideration in the planning of any synthetic route in this field is the judicious choice of a protecting group used for protection of reactive functional groups present in the compounds described in this invention. An authoritative account describing the many alternatives to the trained practitioner is Wuts and Greene, Greene’s Protective Groups in Organic Synthesis, Fourth Edition, Wiley and Sons (2007).
EXEMPLIFICATION Synthetic Preparation of Compound Embodiments Synthesis of Compound 1 Preparation of 7-[(3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6- carbonitrile (Compound 1)
Figure imgf000258_0001
The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aS,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (off-white solid). LC/MS (ESI) m/z: 476.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.16 (d, J = 8.4 Hz, 2H), 6.56 (d, J = 8.8 Hz, 2H), 4.22 - 4.14 (m, 2H), 4.11 - 4.02 (m, 2H), 3.54 - 3.49 (m, 2H), 3.47 (s, 3H), 3.30 - 3.29 (m, 2H), 3.19 (t, J = 9.2 Hz, 2H), 2.69 (s, 3H).
Synthesis of Compound 2 Preparation of 7-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 2) The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3S,4R)-N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 426.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.03 (t, J = 8.8 Hz, 2H), 6.87 - 6.81 (m, 2H), 4.50 (d, J = 12.4 Hz, 1H), 4.32 (d, J = 13.6 Hz, 1H), 3.96 (td, J = 4.0, 12.4 Hz, 1H), 3.73 (dd, J = 2.4, 13.2 Hz, 1H), 3.48 (s, 3H), 3.42 - 3.38 (m, 1H), 2.79 (s, 3H), 2.71 (s, 3H), 2.41 (dd, J = 4.0, 12.4 Hz, 1H), 2.35 - 2.27 (m, 1H), 1.76 - 1.68 (m, 1H), 0.82 (d, J = 6.8 Hz, 3H). Synthesis of Compound 3
Step 1: Preparation of tert-butyl 4-[(2,5-difluorophenyl)methyl]-4-hydroxy- piperidine-1-carboxylate To isopropyl magnesium chloride (2 M, 6.72 mL, 1.43 eq) in tetrahydrofuran was added a solution of 2-bromo-1,4-difluoro-benzene (1.81 g, 9.38 mmol, 1 eq) in tetrahydrofuran, and the resulting mixture was stirred at 25°C for 6 h. Bromocopper;methylsulfanylmethane (193 mg, 0.9 mmol, 0.1 eq) was added then added, and the resulting mixture was stirred for 10 minutes at 25°C. A solution of tert-butyl 1-oxa-6-azaspiro[2.5]octane-6-carboxylate (2 g, 9.38 mmol, 1 eq) in tetrahydrofuran was then added over a period of 5 min, maintaining the temperature at 25°C, and the reaction mixture was stirred at 25°C for 12 h. Saturated aqueous NH4Cl (20 ml) was added, and the resulting mixture was extracted with ethyl acetate (20 mL*2). The combined organic extracts were washed with brine (40 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give the tert-butyl 4-[(2,5-difluorophenyl)methyl]-4-hydroxy-piperidine-1-carboxylate (2.23 g, 6.80 mmol, 73% yield) as a yellow oil. LC/MS (ESI) m/z: 254.0 [M -73]+. Step 2: Preparation of 5-fluorospiro[3H-benzofuran-2,4'-piperidine] A mixture of tert-butyl 4-[(2,5-difluorophenyl)methyl]-4-hydroxy-piperidine-1-carboxylate (1 g, 3.05 mmol, 1 eq) and potassium tert-butoxide (1 g, 9.16 mmol, 3 eq) in 1,2- dimethoxyethane (15 mL) was stirred at 60 °C for 12 h. The material was collected by filtration and then treated with concentrated hydrochloric acid. The resulting mixture was concentrated, and the resulting residue was purified by prep-HPLC (column: YMC Triart C18250*50 mm*7 µm; mobile phase: [1-35% CH3CN in water (HCl)]) to afford 5- fluorospiro[3H-benzofuran-2,4'-piperidine] (50 mg, 0.2 mmol, 7% yield, HCl salt) as a white solid. LC/MS (ESI) m/z: 208.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ 9.84 - 9.44 (m, 2H), 6.92 - 6.80 (m, 2H), 6.68 (dd, J = 4.0, 8.8 Hz, 1H), 3.48 - 3.36 (m, 4H), 3.08 (s, 2H), 2.32 - 2.2 (m, 2H), 2.16 - 2.12 (m, 2H). Step 3: Preparation of 7-(5-fluorospiro[3H-benzofuran-2,4'-piperidine]-1'-yl)-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 3) The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 5- fluorospiro[3H-benzofuran-2,4'-piperidine]. (off-white solid). LC/MS (ESI) m/z: 411.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.08 (dd, J = 2.8, 8.4 Hz, 1H), 6.92 (dt, J = 2.8, 9.2 Hz, 1H), 6.76 (dd, J = 4.4, 8.8 Hz, 1H), 4.12 - 4.04 (m, 2H), 3.96 - 3.88 (m, 2H), 3.48 (s, 3H), 3.12 (s, 2H), 2.72 (s, 3H), 2.04 - 1.96 (m, 4H).
Synthesis of Compound 4 Preparation of 7-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-5-yl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 4)
Figure imgf000262_0001
Compound 4 racemic The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 5-(4- fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (white solid). LC/MS (ESI) m/z: 410.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.01 (t, J = 8.8 Hz, 2H), 6.49 (dd, J = 4.4, 9.2 Hz, 2H), 4.18 (dd, J = 5.6, 9.2 Hz, 2H), 4.11 - 4.01 (m, 2H), 3.50 - 3.44 (m, 5H), 3.20 - 3.12 (m, 2H), 2.69 (s, 3H), 2.48 - 2.39 (m, 2H).
Synthesis of Compound 6
Figure imgf000263_0001
Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl- piperidine-1-carboxylate To a mixture of tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (120 mg, 0.37 mmol, 1 eq), formaldehyde (149.89 mg, 1.85 mmol, 0.14 mL, 37% purity, 5 eq), and sodium cyanoborohydride (70 mg, 1.11 mmol, 3 eq) in methanol (1 mL) and tetrahydrofuran (1 mL) was added acetic acid (53 mg, 0.88 mmol, 0.05 mL, 2.37 eq), and the reaction mixture was stirred at 40 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3S,4R)-4-(4-chloro- N-methyl-anilino)-3-methyl-piperidine-1-carboxylate (100 mg, 0.30 mmol, 80% yield) as colorless oil. LC/MS (ESI) m/z: 339.0 [M+H]+.1H NMR (400MHz, DMSO-d6) δ 7.22 - 7.13 (m, 2H), 6.80 - 6.69 (m, 2H), 4.10 (d, J = 2.4 Hz, 1H), 3.89 - 3.81 (m, 1H), 3.81 - 3.65 (m, 1H), 3.17 - 2.96 (m, 1H), 2.95 - 2.78 (m, 1H), 2.77 (s, 3H), 2.12 (s, 1H), 2.04 - 1.96 (m, 1H), 1.49 (dd, J = 2.4, 12.4 Hz, 1H), 1.40 (s, 9H), 0.80 (d, J = 7.2 Hz, 3H). Step 2: Preparation of (3S,4R)-N-(4-chlorophenyl)-N,3-dimethyl-piperidin-4-amine
Figure imgf000264_0001
A mixture of tert-butyl (3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.30 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 45.77 eq) in dichloromethane (1 mL) was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated under reduced pressure to give (3S,4R)-N-(4-chlorophenyl)-N,3-dimethyl- piperidin-4-amine (100 mg, crude, TFA salt) as a colorless oil. LC/MS (ESI) m/z: 239.0 [M+H]+. Step 3: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 6)
Figure imgf000264_0002
The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- chlorophenyl)-N,3-dimethyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 456.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 8.8 Hz, 1H), 7.82 (d, J = 8.8 Hz, 1H), 7.20 (d, J = 8.8 Hz, 2H), 6.85 (d, J = 9.2 Hz, 2H), 4.34 (d, J = 12.4 Hz, 1H), 4.14 (td, J = 3.2, 12.4 Hz, 1H), 4.04 - 3.95 (m, 1H), 3.91 - 3.83 (m, 1H), 3.53 (s, 3H), 3.45 (t, J = 11.6 Hz, 1H), 2.85 (s, 3H), 2.78 - 2.69 (m, 1H), 2.33 (d, J = 3.6 Hz, 1H), 1.82 - 1.72 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H). Synthesis of Compound 7 Step 1: Preparation of tert-butyl (3aR,4R,6aS)-4-methyl-2-[4- (trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate To a mixture of tert-butyl (3aR,4R,6aS)-4-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole-5-carboxylate (257 mg, 1.14 mmol, 1 eq) ,1-bromo-4-(trifluoromethoxy)benzene (274 mg, 1.14 mmol, 0.2 mL, 1 eq), and Cs2CO3 (1.11 g, 3.41 mmol, 3 eq) in N,N- dimethylacetamide (2.5 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro- 2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (55 mg, 0.06 mmol, 0.05 eq) under nitrogen atmosphere, and the suspension was degassed and purged with nitrogen (3X). The reaction mixture was stirred at 80°C for 3 h under N2. The mixture was diluted with water (6 mL) and extracted with ethyl acetate (6 mL*2). The combined organic extracts were washed with brine (12 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography(gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3aR,4R,6aS)-4-methyl-2-[4- (trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (181 mg, 0.55 mmol, 41% yield) as a yellow oil. LC/MS (ESI) m/z: 387.2 [M+H]+. Step 2: Preparation of (3R,3aR,6aR)-3-methyl-5-[4-(trifluoromethoxy)phenyl]- 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aR,4R,6aS)-4-methyl-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (90 mg, 0.23 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 57.99 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes. The mixture was concentrated under vacuum to give (3R,3aR,6aR)-3-methyl-5- [4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (100 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 287.3 [M+H]+.
Step 3: Preparation of 4-[(3aR,4R,6aS)-4-methyl-2-[[4- (trifluoromethoxy)phenyl]methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]- 6-chloro-1-methyl-1,5-naphthyridin-2-one The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,3aR,6aR)-3- methyl-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole and (6-chloro-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate. (off-white solid). LC/MS (ESI) m/z: 479.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.95 (d, J = 9.2 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 7.13 (d, J = 8.4 Hz, 2H), 6.61 - 6.53 (m, 2H), 5.63 (s, 1H), 5.06 - 4.90 (m, 1H), 4.00 (dd, J = 8.4, 11.2 Hz, 1H), 3.56 (dd, J = 8.8, 9.6 Hz, 1H), 3.50 - 3.40 (m, 5H), 3.27 - 3.23 (m, 1H), 3.21 (d, J = 4.0 Hz, 1H), 3.13 (dd, J = 6.0, 10.0 Hz, 1H), 2.90 - 2.80 (m, 1H), 1.21 (d, J = 6.4 Hz, 3H). Synthesis of Compound 8
Step 1: Preparation of tert-butyl (3aS,6aR)-2-[4-(trifluoromethoxy)phenyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl (3aS,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (150 mg, 0.71 mmol, 1 eq) and 1-bromo-4-(trifluoromethoxy)benzene (255 mg, 1.06 mmol, 0.16 mL, 1.5 eq) in dioxane (3 mL) were added Cs2CO3 (690 mg, 2.12 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (34 mg, 0.035 mmol, 0.05 eq), and the reaction mixture was stirred at 100 °C for 4 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to afford tert-butyl (3aS,6aR)-2-[4-(trifluoromethoxy)phenyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (270 mg, crude) as a green oil. LC/MS (ESI) m/z: 372.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.09 (d, J = 8.8 Hz, 2H), 6.51 - 6.46 (m, 2H), 3.65 (dd, J = 7.2, 11.2 Hz, 2H), 3.51 (d, J = 2.8 Hz, 2H), 3.37 (d, J = 10.4 Hz, 1H), 3.31 - 3.23 (m, 1H), 3.20 (dd, J = 3.2, 9.2 Hz, 2H), 3.00 (s, 2H), 1.46 (s, 9H). Step 2: Preparation of (3aR,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole
To a solution of tert-butyl (3aS,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (120 mg, 0.32 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 21 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under reduced pressure to give (3aR,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole (120 mg, crude, TFA salt) as a brown oil. LC/MS (ESI) m/z: 273.3 [M+H]+. Step 3: 4-[(3aS,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 8) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-[4- (trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (yellow solid). LC/MS (ESI) m/z: 490.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.00 (d, J = 9.2 Hz, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.61 - 6.54 (m, 2H), 4.51 (dd, J = 6.8, 12.4 Hz, 2H), 4.09 (s, 2H), 3.55 - 3.50 (m, 2H), 3.48 (s, 3H), 3.29 - 3.24 (m, 2H), 3.20 - 3.13 (m, 2H). Synthesis of Compound 9 Step 1: Preparation of WC-ARV-JM-027-C-1, tert-butyl (3aS,6aR)-3a,6a-dimethyl-2- [4-(trifluoromethoxy)phenyl]-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl (3aR,6aS)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4- c]pyrrole-5-carboxylate (150 mg, 0.62 mmol, 1 eq) and 1-bromo-4- (trifluoromethoxy)benzene (225 mg, 0.93 mmol, 0.14 mL, 1.5 eq) in dioxane (3 mL) were added Cs2CO3 (610 mg, 1.87 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (30 mg, 0.03 mmol, 0.05 eq), and the reaction mixture was stirred at 100 °C for 4 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to afford tert-butyl (3aS,6aR)-3a,6a-dimethyl-2-[4-(trifluoromethoxy)phenyl]-1,3,4,6-tetrahydropyrrolo[3,4- c]pyrrole-5-carboxylate (85 mg, 0.21 mmol, 34% yield) as a brown oil. LC/MS (ESI) m/z: 401.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.07 (d, J = 8.8 Hz, 2H), 6.45 - 6.39 (m, 2H), 3.43 - 3.34 (m, 4H), 3.31 - 3.26 (m, 2H), 3.22 (d, J = 9.6 Hz, 2H), 1.44 (s, 9H), 1.13 (s, 6H). Step 2: Preparation of (3aR,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole To a solution of tert-butyl (3aS,6aR)-3a,6a-dimethyl-2-[4-(trifluoromethoxy)phenyl]-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (85 mg, 0.21 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (77 mg, 6.8 mmol, 0.5 mL, 31.8 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under reduced pressure to give (3aR,6aS)-3a,6a-dimethyl-5-[4-(trifluoromethoxy)phenyl]-2,3,4,6- tetrahydro-1H-pyrrolo[3,4-c]pyrrole (85 mg, crude, TFA salt) as a brown oil. LC/MS (ESI) m/z: 301.2 [M+H]+. Step 3: Preparation of 4-[(3aS,6aR)-3a,6a-dimethyl-2-[4-(trifluoromethoxy)phenyl]- 1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 9) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-3a,6a- dimethyl-5-[4-(trifluoromethoxy)phenyl]-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrole. (yellow solid). LC/MS (ESI) m/z: 518.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.00 (d, J = 9.2 Hz, 1H), 7.77 (d, J = 9.2 Hz, 1H), 7.15 (d, J = 8.4 Hz, 2H), 6.55 - 6.49 (m, 2H), 4.35 - 4.30 (m, 2H), 4.24 - 4.17 (m, 2H), 3.50 (s, 1H), 3.49 - 3.46 (m, 4H), 3.26 (d, J = 10.4 Hz, 2H), 1.17 (s, 6H). Synthesis of Compound 10
Figure imgf000272_0001
Step 1: Preparation of ethyl 2-methyl-5-(methylamino)thiazole-4-carboxylate To a mixture of ethyl 5-amino-2-methyl-thiazole-4-carboxylate (15 g, 80.55 mmol, 1 eq) in tetrahydrofuran (150 mL) was added sodium hydride (4.19 g, 104.71 mmol, 60% purity, 1.3 eq) in portions, and the mixture was stirred at 0°C for 0.5 h. Iodomethane (19.44 g, 136.93 mmol, 8.52 mL, 1.7 eq) was then added, and the reaction mixture was stirred at 20°C for 12 h under nitrogen. The mixture was diluted with saturated aqueous NH4Cl (300 mL), then extracted with ethyl acetate (3 × 300 mL). The combined organic extract was washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 3:1 petroleum ether/ethyl acetate) to give ethyl 2-methyl-5-(methylamino)thiazole-4-carboxylate (18 g, 89.88 mmol, 56% yield) as a yellow solid. LC/MS (ESI) m/z: 200.9 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.31 - 7.15 (m, 1H), 4.48 - 4.33 (m, 2H), 3.03 - 3.00 (m, 3H), 2.63 - 2.50 (m, 3H), 1.46 - 1.34 (m, 3H). Step 2: Preparation of ethyl 5-[(2-cyanoacetyl)-methyl-amino]-2-methyl-thiazole-4- carboxylate A mixture of 2-cyanoacetic acid (3.06 g, 35.95 mmol, 1.2 eq) in acetic anhydride (32.70 g, 320.31 mmol, 30.00 mL, 10.69 eq) was stirred at 80 °C for 0.5 h. Ethyl 2-methyl-5- (methylamino)thiazole-4-carboxylate (6 g, 29.96 mmol, 1 eq) was then added, and the reaction mixture was stirred at 80 °C for 0.5 h under nitrogen. The mixture was concentrated, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give ethyl 5-[(2-cyanoacetyl)-methyl-amino]-2-methyl- thiazole-4-carboxylate (2.5 g, 9.35 mmol, 31% yield) as a yellow solid. LC/MS (ESI) m/z: 268.1 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 4.47 - 4.40 (m, 2H), 3.43 (d, J = 15.2 Hz, 2H), 3.28 (s, 3H), 2.81 - 2.74 (m, 3H), 1.42 - 1.37 (m, 3H). Step 3: Preparation of 7-hydroxy-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6- carbonitrile A mixture of ethyl 5-[(2-cyanoacetyl)-methyl-amino]-2-methyl-thiazole-4-carboxylate (2.5 g, 9.35 mmol, 1 eq) and 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine (1.71 g, 11.22 mmol, 1.69 mL, 1.2 eq) in ethanol (20 mL) was stirred at 20 °C for 0.5 h under nitrogen. The mixture was concentrated, and the resulting residue was purified by prep-HPLC (column: Welch Xtimate C18250*70mm#10µm; mobile phase: [0-30% CH3CN in water (0.225% formic acid)]) to give 7-hydroxy-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (1.1 g, 4.97 mmol, 53% yield) as a white solid. LC/MS (ESI) m/z: 221.9 [M+1]+.1H NMR (400 MHz, DMSO-d6) δ 3.50 (s, 3H), 2.74 (s, 3H). Step 4: Preparation of (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate To a mixture of 7-hydroxy-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (1.1 g, 4.97 mmol, 1 eq), triethylamine (1.51 g, 14.92 mmol, 2.08 mL, 3 eq), and 4- dimethylaminopyridine (61 mg, 0.50 mmol, 0.1 eq) in dichloromethane (10 mL) at 0°C was added trifluoromethylsulfonyl trifluoromethanesulfonate (2.81 g, 9.94 mmol, 1.64 mL, 2 eq), and the reaction mixture was stirred at 0°C for 1 h under nitrogen. The mixture was diluted with water (30 mL) and extracted with dichloromethane (30 mL*2). The combined organic extracts were washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 10/1 to 1:1 etroleum ether/ethyl acetate) to give (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4- b]pyridin-7-yl) trifluoromethanesulfonate (1.2 g, 3.40 mmol, 68% yield) as a yellow solid. LC/MS(ESI) m/z: 354.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 3.50 (s, 3H), 2.74 (s, 3H). Step 5: Preparation of 2,4-dimethyl-5-oxo-7-spiro[3H-benzofuran-2,4'-piperidine]-1'- yl-thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 10) A mixture of (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate (80 mg, 0.23 mmol, 1 eq), spiro[3H-benzofuran-2,4'-piperidine] (51 mg, 0.23mmol, 1 eq, hydrogen chloride), and diisopropylethylamine (88 mg, 0.68 mmol, 0.12 mL, 3 eq) in acetonitrile (2 mL) was degassed and purged with nitrogen (3X), then stirred at 40 °C for 4 hours under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure, and the resulting residue was triturated with DMF (0.1 mL) and acetonitrile (0.2 mL) at 25°C, then collected by filtration to afford 2,4-dimethyl-5- oxo-7-spiro[3H-benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile (33 mg, 0.082 mmol, 36% yield) as a white solid. LC/MS (ESI) m/z: 393.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.21 (d, J = 7.2 Hz, 1H), 7.10 (t, J = 7.2 Hz, 1H), 6.86 - 6.77 (m, 2H), 4.13 - 4.04 (m, 2H), 3.97 - 3.88 (m, 2H), 3.49 (s, 3H), 3.11 (s, 2H), 2.70 (s, 3H), 2.03 - 1.95 (m, 4H). Synthesis of Compound 11
Figure imgf000276_0002
Compound 11 Step 1: Preparation of tert-butyl (3R,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3- methyl-piperidine-1-carboxylate
Figure imgf000276_0001
To a mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.07 mmol, 1 eq) and 4-chloro-2-methyl-aniline (2.39 g, 16.88 mmol, 1.2 eq) in methanol (15 mL) and acetic acid (15 mL) was added borane 2-methylpyridine (3.01 g, 28.13 mmol, 2 eq), and the reaction mixture was stirred at 25 °C for 12 h under nitrogen. The mixture was concentrated under vacuum, and the resulting residue was purified by prep-HPLC (column: Welch Xtimate C18250*70mm#10um; mobile phase: [53-88% CH3CN in water (NH4HCO3)]) followed by prep-SFC (column: DAICEL CHIRALPAK AD-H(250mm*30mm,5um); mobile phase: 20% isopropanol (0.1% NH4OH) in CO2) to afford the two diastereomers. Compound tert-butyl (3R,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-piperidine-1- carboxylate (600 mg, 1.77 mmol, 21% yield) was obtained as a white solid.1H NMR (400 MHz, DMSO-d6) δ 7.02 - 6.97 (m, 2H), 6.61 (d, J = 8.4 Hz, 1H), 4.35 - 4.31 (m, 1H), 3.94 - 3.78 (m, 1H), 3.70 - 3.56 (m, 2H), 3.13 - 2.83 (m, 2H), 2.09 (s, 3H), 2.06 (d, J = 3.6 Hz, 1H), 1.74 - 1.62 (m, 1H), 1.54 (d, J = 3.6 Hz, 1H), 1.39 (s, 9H), 0.76 (d, J = 7.2 Hz, 3H). tert-Butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-piperidine-1-carboxylate (1.2 g, 3.54 mmol, 43% yield) was obtained as a white solid.1H NMR (400 MHz, DMSO-d6) δ 7.01 - 6.91 (m, 2H), 6.58 (d, J = 9.2 Hz, 1H), 4.54 (d, J = 9.2 Hz, 1H), 3.91 (d, J = 11.6 Hz, 2H), 3.12 – 3.08 (m, 1H), 2.85 (d, J = 1.6 Hz, 1H), 2.06 (s, 3H), 1.88 (dd, J = 3.2, 13.2 Hz, 1H), 1.75 - 1.59 (m, 1H), 1.40 (s, 9H), 1.26 - 1.15 (m, 1H), 0.88 (d, J = 6.4 Hz, 3H). Step 2: Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3R,4S)-4-(4-chloro-2-methyl-anilino)-3- methyl-piperidine-1-carboxylate The enantiomers were separated by prep-SFC(column: DAICEL CHIRALCEL OD(250mm*30mm,10um); mobile phase: 20% isopropanol (0.1% NH4OH) in CO2). Absolute configuration was arbitrarily assigned. tert-Butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-piperidine-1-carboxylate (780 mg) was obtained as a white solid. LC/MS (ESI) m/z: 339.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.03 - 6.97 (m, 2H), 6.61 (d, J = 8.4 Hz, 1H), 4.38 - 4.32 (m, 1H), 3.92 - 3.75 (m, 1H), 3.71 - 3.56 (m, 2H), 3.34 (s, 3H), 3.17 (d, J = 5.2 Hz, 1H), 3.10 (d, J = 12.0 Hz, 1H), 3.04 - 2.79 (m, 1H), 1.76 - 1.63 (m, 1H), 1.57 - 1.50 (m, 1H), 1.42 - 1.38 (m, 9H), 0.76 (d, J = 7.2 Hz, 3H). tert-Butyl (3R,4S)-4-(4-chloro-2-methyl-anilino)-3-methyl-piperidine-1-carboxylate (630 mg) was obtained as a white solid. LC/MS (ESI) m/z: 339.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.03 - 6.97 (m, 2H), 6.61 (d, J = 8.4 Hz, 1H), 4.34 (d, J = 7.6 Hz, 1H), 3.93 - 3.73 (m, 1H), 3.70 - 3.57 (m, 2H), 3.34 (s, 3H), 3.17 (d, J = 4.8 Hz, 1H), 3.10 (d, J = 12.0 Hz, 1H), 2.89 (s, 1H), 1.74 - 1.62 (m, 1H), 1.57 - 1.49 (m, 1H), 1.39 (s, 9H), 0.76 (d, J = 7.2 Hz, 3H). Step 3: Preparation of (3S,4R)-N-(4-chloro-2-methyl-phenyl)-3-methyl-piperidin-4- amine To a solution of tert-butyl (3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.29 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 23 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under vacuum to afford (3S,4R)-N-(4-chloro-2- methyl-phenyl)-3-methyl-piperidin-4-amine (100 mg, crude, TFA salt) as a colorless oil. LC/MS (ESI) m/z: 239.0 [M+H]+. Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 11)
The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS (ESI) m/z: 456.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.04 - 6.99 (m, 2H), 6.72 (d, J = 9.2 Hz, 1H), 4.55 (d, J = 8.0 Hz, 1H), 4.18 (d, J = 13.8 Hz, 1H), 4.00 - 3.93 (m, 1H), 3.89 - 3.81 (m, 2H), 3.63 - 3.54 (m, 1H), 3.53 (s, 3H), 2.28 - 2.18 (m, 1H), 2.15 (s, 3H), 2.07 (s, 1H), 1.89 - 1.79 (m, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 12 Synthesis of 6-chloro-4-[(3S,4S)-4-(4-chloro-2-methyl-anilino)-3-methyl-1-piperidyl]- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 12) The title compound was prepared in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- chloro-2-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3S,4S)-4-(4-chloro-2-methyl-anilino)-3-methyl- piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 456.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 7.05 - 6.93 (m, 2H), 6.75 - 6.65 (m, 1H), 4.75 (d, J = 9.2 Hz, 1H), 4.37 - 4.20 (m, 2H), 3.66 - 3.55 (m, 1H), 3.53 (s, 3H), 3.19 - 3.11 (m, 1H), 2.26 - 2.16 (m, 1H), 2.13 - 2.06 (m, 4H), 1.69 - 1.55 (m, 1H), 0.97 (d, J = 6.4 Hz, 3H). Synthesis of Compound 13 Preparation of 6-chloro-4-[(3S,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 13) The title compound was prepared in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- chloro-2-methoxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3S,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 472.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.8 (d, J = 9.2 Hz, 1H), 6.84 (d, J = 2.4 Hz, 1H), 6.80 - 6.76 (m, 1H), 6.68 (d, J = 8.4 Hz, 1H), 4.76 (d, J = 9.6 Hz, 1H), 4.32 - 4.2 (m, 2H), 3.80 (s, 3H), 3.64 - 3.56 (m, 1H), 3.52 (s, 3H), 3.4 - 3.32 (m, 1H), 3.16 (dd, J = 11.2, 12.8 Hz, 1H), 2.16 - 2.04 (m, 2H), 1.64 - 1.52 (m, 1H), 0.96 (d, J = 6.4 Hz, 3H). Synthesis of Compound 14 Preparation of 6-chloro-4-[(3R,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 14)
The title compound was prepared in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- chloro-2-methoxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3R,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 472.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 8.8 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 6.84 (d, J = 2.0 Hz, 1H), 6.80 - 6.76 (m, 1H), 6.68 (d, J = 8.4 Hz, 1H), 4.76 (d, J = 9.6 Hz, 1H), 4.24 (t, J = 13.6 Hz, 2H), 3.80 (s, 3H), 3.6 - 3.56 (m, 1H), 3.52 (s, 3H), 3.4 - 3.36 (m, 1H), 3.16 (t, J = 12.0 Hz, 1H), 2.16 - 2.04 (m, 2H), 1.64 - 1.52 (m, 1H), 0.96 (d, J = 6.4 Hz, 3H). Synthesis of Compound 15 Preparation of 2,4-dimethyl-5-oxo-7-spiro[indane-2,4'-piperidine]-1'-yl-thiazolo[5,4- b]pyridine-6-carbonitrile (Compound 15) The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 1,3- dihydrospiro[indene-2,4'-piperidine] and (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4- b]pyridin-7-yl) trifluoromethanesulfonate. (off-white solid). LC/MS (ESI) m/z: 391.1 [M+H] +.1H NMR (400 MHz, DMSO-d6) δ 7.24 - 7.17 (m, 2H), 7.15 - 7.09 (m, 2H), 3.94 (s, 4H), 3.48 (s, 3H), 2.88 (s, 4H), 2.69 (s, 3H), 1.78 - 1.72 (m, 4H). Synthesis of Compound 16 Preparation of 6-chloro-4-[(3R,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 16) The title compound was prepared in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- chloro-2-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3R,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl- piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 456.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 7.06 - 6.92 (m, 2H), 6.73 - 6.67 (m, 1H), 4.75 (d, J = 9.2 Hz, 1H), 4.35 - 4.21 (m, 2H), 3.64 - 3.56 (m, 1H), 3.53 (s, 3H), 3.15 (dd, J = 11.2, 12.8 Hz, 1H), 2.28 - 2.17 (m, 1H), 2.16 - 2.10 (m, 1H), 2.09 (s, 3H), 1.67 - 1.55 (m, 1H), 0.97 (d, J = 6.8 Hz, 3H). Synthesis of Compound 17 Preparation of 6-chloro-4-[(3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 17)
The title compound was prepared in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- chloro-2-methoxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate. (off-white solid). LC/MS (ESI) m/z: 472.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 2.4 Hz, 1H), 6.84 - 6.76 (m, 1H), 6.76 - 6.68 (m, 1H), 4.6 (d, J = 8.8 Hz, 1H), 4.16 - 4.04 (m, 1H), 3.92 - 3.8 (m, 6H), 3.6 (t, J = 10 Hz, 1H), 3.52 (s, 3H), 2.32 (dd, J = 1.6, 3.6 Hz, 1H), 2.08 (d, J = 2.8 Hz, 1H), 1.88 (td, J = 4.4, 8.8 Hz, 1H), 0.76 (s, 3H). Synthesis of Compound 18 Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 18)
Figure imgf000283_0001
C Compound 18 The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS (ESI) m/z: 456.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.04 - 6.99 (m, 2H), 6.72 (d, J = 9.2 Hz, 1H), 4.55 (d, J = 8.0 Hz, 1H), 4.18 (d, J = 13.8 Hz, 1H), 4.00 - 3.93 (m, 1H), 3.89 - 3.81 (m, 2H), 3.63 - 3.54 (m, 1H), 3.53 (s, 3H), 2.28 - 2.18 (m, 1H), 2.15 (s, 3H), 2.07 (s, 1H), 1.89 - 1.79 (m, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 19
Figure imgf000284_0001
Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3R,4R)-4-(4-chloro-2-methoxy-anilino)-3- methyl-piperidine-1-carboxylate A mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.07 mmol, 1 eq), 4- chloro-2-methoxy-aniline (2.22 g, 14.07 mmol, 1 eq), and borane 2-methylpyridine (3 g, 28.13 mmol, 2 eq) in methanol (15 mL) and acetic acid (15 mL) and stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the crude product was diluted with water (40 mL) and extracted with ethyl acetate (40 mL*2). The combined organic extracts were washed with brine (80 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC (column: YMC Triart C18 70*250 mm*7 µm; mobile phase: [51-81% CH3CN in water (NH4HCO3)]) followed by SFC (column: DAICEL CHIRALPAK AD-H (250 mm*30 mm,5 um); mobile phase: 20% isopropanol (0.1% NH4OH) in CO2) to afford the diastereomers. tert-Butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1-carboxylate (1.57 g, 4.42 mmol, 31% yield) was obtained as an off-white oil.1H NMR (400 MHz, DMSO-d6) δ 6.88 (d, J = 2.4 Hz, 1H), 6.80 (dd, J = 2.4, 8.4 Hz, 1H), 6.64 (d, J = 8.8 Hz, 1H), 4.48 (d, J = 8.8 Hz, 1H), 3.8 (s, 3H), 3.68 - 3.52 (m, 2H), 3.32 (s, 1H), 3.16 - 3.08 (m, 1H), 3 - 2.84 (m, 1H), 2.12 - 2.0 (m, 1H), 1.68 - 1.48 (m, 2H), 1.40 (s, 9H), 0.76 (d, J = 7.2 Hz, 3H). tert-Butyl (3R,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1-carboxylate (624 mg, 1.76 mmol, 13% yield) was obtained as an off-white oil. 1H NMR (400 MHz, DMSO- d6) δ 6.84 (d, J = 2.4 Hz, 1H), 6.76 (dd, J = 2.4, 8.4 Hz, 1H), 6.60 (d, J = 8.8 Hz, 1H), 4.68 (d, J = 9.6 Hz, 1H), 3.88 (d, J = 11.6 Hz, 2H), 3.80 (s, 3H), 3.12 - 3.00 (m, 1H), 2.88 - 2.76 (m, 1H), 1.88 - 1.8 (m, 1H), 1.68 - 1.52 (m, 1H), 1.40 (s, 9H), 1.24 - 1.08 (m, 2H), 0.84 (d, J = 6.4 Hz, 3H). Step 2: Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate tert-butyl (3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl- piperidine-1-carboxylate The enantiomers were separated by SFC (column: DAICEL CHIRALCEL OD (250 mm*30 mm,10 um); mobile phase: 20% isopropanol in CO2). Absolute configuration was arbitrarily assigned. tert-Butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1-carboxylate (731 mg, 2.06 mmol, 46% yield) was obtained as an off-white oil. LC/MS (ESI) m/z: 299.0 [M- 55]+. tert-Butyl (3R,4S)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1-carboxylate (758 mg, 2.14 mmol, 48% yield) was obtained as an off-white oil. LC/MS (ESI) m/z: 299.0 [M- 55]+. Step 3: Preparation of (3S,4R)-N-(4-chloro-2-methoxy-phenyl)-3-methyl-piperidin-4- amine A mixture of tert-butyl (3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.28 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 47.93 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes. The mixture was concentrated under vacuum to give (3S,4R)-N-(4-chloro-2-methoxy-phenyl)-3-methyl- piperidin-4-amine (120 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 255.1 [M+H]+ Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-methoxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 19)
Figure imgf000287_0001
The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- chloro-2-methoxy-phenyl)-3-methyl-piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 472.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.8 (d, J = 9.2 Hz, 1H), 6.88 (d, J = 2.4 Hz, 1H), 6.84 - 6.76 (m, 1H), 6.76 - 6.72 (m, 1H), 4.6 (d, J = 8.8 Hz, 1H), 4.08 (d, J = 12.4 Hz, 1H), 3.88 - 3.8 (m, 6H), 3.64 - 3.56 (m, 1H), 3.52 (s, 3H), 2.32 (s, 1H), 2.2 - 2.08 (m, 1H), 1.92 - 1.80 (m, 1H), 0.84 (d, J = 7.2 Hz, 3H). Synthesis of Compounds 21 and 20
Figure imgf000287_0002
Step 1: Preparation of tert-butyl (3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3- methyl-piperidine-1-carboxylate A mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.1 mmol, 1 eq) , 2- amino-5-fluoro-phenol (2.68 g, 21 mmol, 1.5 eq), and acetic acid (84.47 mg, 1.41 mmol, 0.08 mL, 0.1 eq) in CH3OH (30 mL) was stirred at 50 °C for 0.5 h. Sodium cyanoborohydride (2.65 g, 42 mmol, 3 eq) was then added, and the reaction mixture was stirred at 50 °C for 12 h. The mixture was diluted with water (150 mL) and extracted with ethyl acetate (150 mL*4). The combined organic extracts were washed with brine (300 mL*2), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC (column: YMC Triart C18250*50mm*7um; mobile phase: [37-67% CH3CN in water (NH4HCO3)]) to afford the desired products. tert-Butyl (3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-piperidine-1-carboxylate (1 g, 3 mmol, 22% yield) was obtained as a brown oil.1H NMR (400 MHz, DMSO-d6) δ 9.84 (d, J = 2.0 Hz, 1H), 6.56 - 6.49 (m, 2H), 6.47 - 6.40 (m, 1H), 4.12 - 4.01 (m, 2H), 3.63 - 3.50 (m, 2H), 3.09 (d, J = 12.0 Hz, 1H), 2.08 - 2.00 (m, 1H), 1.60 - 1.49 (m, 2H), 1.39 (s, 9H), 0.75 (d, J = 6.8 Hz, 3H). tert-Butyl (3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-piperidine-1-carboxylate (480 mg, 1.48 mmol, 11% yield) was obtained as a brown oil. LC/MS (ESI) m/z: 269.0 (M-55)+. 1H NMR (400 MHz, DMSO-d6) δ 9.74 (s, 1H), 6.52 - 6.46 (m, 2H), 6.45 - 6.39 (m, 1H), 4.11 (d, J = 9.2 Hz, 1H), 3.88 (d, J = 12.4 Hz, 2H), 3.04 - 2.92 (m, 1H), 2.90 - 2.73 (m, 1H), 1.88 (dd, J = 3.2, 13.2 Hz, 1H), 1.59 - 1.52 (m, 1H), 1.40 (s, 9H), 1.14 - 1.06 (m, 1H), 0.89 (d, J = 6.4 Hz, 3H). Step 2: Preparation of 5-fluoro-2-[[(3S,4R)-3-methyl-4-piperidyl]amino]phenol To a solution of tert-butyl (3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-piperidine-1- carboxylate (400 mg, 1.23 mmol, 1 eq) in dichloromethane (4 mL) was added trifluoroacetic acid (3.08 g, 27 mmol, 2 mL, 22 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under reduced pressure to give 5-fluoro-2-[[(3S,4R)-3-methyl- 4-piperidyl]amino]phenol (400 mg, crude, TFA salt) as a brown oil. LC/MS (ESI) m/z: 225.1 [M+H]+.
Step 3: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS MS (ESI) m/z: 442.0 [M+H]+. Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 21) and 6- chloro-4-[(3R,4S)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (Compound 20) The enantiomers were separated by prep-HPLC (column: DAICEL CHIRALPAK AD(250mm*30mm, 10 µm); mobile phase: [0.1% NH4OH in ethanol]; B%: 50%-50%, 3.2; 30 min). The enantiomers were further purified by prep-HPLC(column: Phenomenex luna C18150*25mm* 10um; mobile phase: [31-61% CH3CN in water (0.225% formic acid)]). Absolute configuration was arbitrarily assigned. 6-Chloro-4-[(3S,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (30 mg, 0.07 mmol, 29% yield) was obtained as a yellow solid. LC/MS (ESI) m/z: 442.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 10.04 - 9.64 (m, 1H), 8.07 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 6.64 (d, J = 6.0, 8.8 Hz, 1H), 6.57 - 6.51 (m, 1H), 6.47 (t, J = 2.8, 8.8 Hz, 1H), 4.18 (d, J = 9.2 Hz, 1H), 4.11 - 4.00 (m, 1H), 3.92 - 3.74 (m, 3H), 3.63 - 3.55 (m, 1H), 3.52 (s, 3H), 2.30 (s, 1H), 2.12 - 2.02 (m, 1H), 1.90 - 1.82 (m, 1H), 0.84 (d, J = 7.2 Hz, 3H). 6-Chloro-4-[(3R,4S)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (40 mg, 0.09 umol, 39% yield) was obtained as a yellow solid. LC/MS (ESI) m/z: 442.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.2 (dd, J = 2.4, 12.0 Hz, 1H), 7.0 (dd, J = 1.2, 8.8 Hz, 1H), 6.8 (t, J = 9.2 Hz, 1H), 5.2 (d, J = 7.2 Hz, 1H), 3.72 - 3.56 (m, 2H), 3.2 - 3.00 (m, 1H), 2.96 - 2.8 (m, 1H), 2.08 - 2.00 (m, 1H), 1.76 - 1.60 (m, 1H), 1.56 - 1.48 (m, 2H), 1.40 (s, 9H), 0.76 (d, J = 6.8 Hz, 3H). Synthesis of Compound 22 Preparation of 6-chloro-4-[(3R,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 22) The title compound was made in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4-chloro-2- fluoro-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1- carboxylate. (off-white solid). LC/MS (ESI) m/z: 460 [M+H]+.1H NMR (400 MHz, DMSO- d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 7.20 (dd, J = 2.4, 11.6 Hz, 1H), 7.00 (dd, J = 1.6, 8.8 Hz, 1H), 6.96 - 6.88 (m, 1H), 5.52 - 5.44 (m, 1H), 4.20 - 4.08 (m, 1H), 3.96 (dd, J = 5.2, 13.2 Hz, 1H), 3.92 - 3.76 (m, 2H), 3.60 (t, J = 10.4 Hz, 1H), 3.52 (s, 3H), 2.32 (d, J = 2.0 Hz, 1H), 2.28 - 2.16 (m, 1H), 1.92 - 1.76 (m, 1H), 0.84 (d, J = 6.8 Hz, 3H). Synthesis of Compound 23
Figure imgf000292_0001
Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate, tert-butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate, tert-butyl (3R,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate, tert-butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate A mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.07 mmol, 1 eq), 4- chloro-2-fluoro-aniline (2 g, 14.07 mmol, 1 eq), and borane 2-methylpyridine (3 g, 28.13 mmol, 2 eq) in acetic acid (15 mL) and methanol (15 mL) was stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the crude product was diluted with water (40 mL) and extracted with ethyl acetate (40 mL*2). The combined organic extracts were washed with brine (80 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC (column: YMC Triart C1870*250mm*7µm; mobile phase: [51-81% CH3CN in water (NH4HCO3)]) followed by SFC (column: DAICEL CHIRALPAK AD-H (250mm*30mm, 5µm); mobile phase: 20% methanol (0.1% NH4OH) in CO2) to afford the desired four isomers. tert-Butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3- methyl-piperidine-1-carboxylate was further purified by SFC (column: DAICEL CHIRALPAK AD-H (250mm*30mm, 5µm); mobile phase: 20% methanol (0.1% NH4OH) in CO2). tert-Butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate was purified by SFC (column: DAICEL CHIRALPAK AD-H (250 mm*30 mm, 5 µm); mobile phase: 25% methanol (0.1% NH4OH) in CO2). Absolute configuration was arbitrarily assigned. tert-Butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (333 mg, 0.97 mmol, 7% yield) was obtained as an off-white oil.1H NMR (400 MHz, DMSO-d6) δ 7.2 (dd, J = 2.4, 11.6 Hz, 1H), 7.20 (td, J = 1.2, 8.8 Hz, 1H), 6.80 (t, J = 9.2 Hz, 1H), 5.20 (d, J = 7.2 Hz, 1H), 3.92 - 3.76 (m, 1H), 3.68 (dd, J = 2.8, 13.2 Hz, 1H), 3.64 - 3.56 (m, 1H), 3.08 (d, J = 12.0 Hz, 1H), 2.96 - 2.76 (m, 1H), 2.04 (dd, J = 3.6, 6.8 Hz, 1H), 1.76 - 1.6 (m, 1H), 1.52 - 1.48 (m, 1H), 1.40 (s, 9H), 0.76 (d, J = 7.2 Hz, 3H). tert-Butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (260 mg, 0.76 mmol, 5% yield) was obtained as an off-white oil.1H NMR (400 MHz, DMSO-d6) δ 7.2 (dd, J = 2.4, 11.8 Hz, 1H), 7.0 (dd, J = 1.2, 8.8 Hz, 1H), 6.8 (t, J = 9.2 Hz, 1H), 5.2 (d, J = 7.2 Hz, 1H), 3.72 - 3.56 (m, 2H), 3.2 - 3.00 (m, 1H), 2.96 - 2.8 (m, 1H), 2.08 - 2.00 (m, 1H), 1.76 - 1.60 (m, 1H), 1.56 - 1.48 (m, 2H), 1.40 (s, 9H), 0.76 (d, J = 6.8 Hz, 3H). tert-Butyl (3R,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (107 mg, 0.31 mmol, 2% yield) was obtained as an off-white oil.1H NMR (400 MHz, DMSO-d6) δ 7.2 (dd, J = 2.4, 12.0 Hz, 1H), 6.96 (d, J = 8.4 Hz, 1H), 6.8 (t, J = 9.2 Hz, 1H), 5.4 (dd, J = 2.2, 9.2 Hz, 1H), 4.0 - 3.84 (m, 2H), 3.12 (dq, J = 3.6, 9.9 Hz, 1H), 2.92 - 2.72 (m, 1H), 1.84 (dd, J = 3.2, 13.2 Hz, 1H), 1.68 - 1.56 (m, 1H), 1.40 (s, 9H), 1.32 - 1.2 (m, 1H), 0.88 (d, J = 6.8 Hz, 3H). tert-Butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (88 mg, 0.26 mmol, 2% yield) was obtained as an off-white oil.1H NMR (400 MHz, DMSO-d6) δ 7.2 (dd, J = 2.4, 12.0 Hz, 1H), 6.96 (td, J = 1.2, 8.8 Hz, 1H), 6.80 - 6.72 (m, 1H), 5.36 (dd, J = 2.0, 9.2 Hz, 1H), 3.96 - 3.80 (m, 2H), 3.12 (dq, J = 4.0, 9.92 Hz, 1H), 2.92 - 2.72 (m, 1H), 1.84 (dd, J = 3.2, 12.8 Hz, 1H), 1.72 - 1.6 (m, 1H), 1.40 (s, 9H), 1.28 - 1.20 (m, 1H), 0.84 (d, J = 6.4 Hz, 3H) Step 2: Preparation of (3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4- amine A mixture of tert-butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.29 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 46.30 eq) in dichloromethane (1 mL) and stirred at 25 °C for 15 minutes. The mixture was concentrated under vacuum to give (3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl- piperidin-4-amine (120 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 243.1 [M+H]+. Step 3: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 23)
A mixture of (3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4-amine (120 mg, 0.34 mmol, 1 eq, trifluoroacetic acid), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (70 mg, 0.28 mmol, 8.19e-1 eq), and diisopropylethylamine (371.00 mg, 2.87 mmol, 0.5 mL, 8.53 eq) in acetonitrile (1.5 mL) was stirred at 40 °C for 3 h. The mixture was concentrated, and the crude product was triturated in DMF, (1 mL) washed with acetonitrile (1 mL*2), and collected by filtration to afford 6-chloro-4-[(3S,4R)-4-(4-chloro-2-fluoro- anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (90.1 mg, 0.19 mmol, 57% yield) as a white solid. LC/MS (ESI) m/z: 460.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.60 (d, J = 9.2 Hz, 1H), 7.20 (dd, J = 2.4, 11.6 Hz, 1H), 7.2 (dd, J = 2.0, 8.8 Hz, 1H), 6.96 - 6.88 (m, 1H), 5.48 (dd, J = 2.0, 8.4 Hz, 1H), 4.20 - 4.08 (m, 1H), 4.00 - 3.92 (m, 1H), 3.88 - 3.76 (m, 2H), 3.60 - 3.56 (m, 1H), 3.52 (s, 3H), 2.32 - 2.28 (m, 1H), 2.28 - 2.16 (m, 1H), 1.88 - 1.76 (m, 1H), 0.84 (d, J = 6.8 Hz, 3H). Synthesis of Compound 24 Preparation of 1,6-dimethyl-2-oxo-4-spiro[3H-benzofuran-2,4'-piperidine]-1'-yl-1,5- naphthyridine-3-carbonitrile (Compound 24) The title compound was made in an analogous manner to 1,6-dimethyl-2-oxo-4-spiro[indane- 2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 6-chloro-1-methyl-2-oxo- 4-spiro[3H-benzofuran-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile. (brown solid). LC/MS (ESI) m/z: 387.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J = 8.8 Hz, 1H), 7.60 (d, J = 8.8 Hz, 1H), 7.20 (d, J = 7.6 Hz, 1H), 7.12 (t, J = 7.6 Hz, 1H), 6.88 - 6.76 (m, 2H), 4.04 - 3.88 (m, 4H), 3.52 (s, 3H), 3.16 (s, 2H), 2.56 (s, 3H), 2.08 (t, J = 5.6 Hz, 4H). Synthesis of Compound 25 Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methoxy-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 25) Step 1: Preparation of (3S,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine A mixture of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1-carboxylate (100 mg, 0.31 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 43.81 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes. The mixture was concentrated under vacuum to give (3S,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine (125 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 255.1 [M+H]+.
Step 2: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 25) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- fluorophenyl)-3-methoxy-piperidin-4-amine. (off-white solid). LC/MS (ESI) m/z: 442.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 6.92 (t, J = 8.8 Hz, 2H), 6.80 - 6.64 (m, 2H), 5.44 (d, J = 8.8 Hz, 1H), 4.32 - 4.12 (m, 2H), 3.84 (d, J = 12.4 Hz, 2H), 3.64 (s, 1H), 3.52 (s, 3H), 3.52 - 3.48 (m, 1H), 3.20 (s, 3H), 2.28 - 2.16 (m, 1H), 1.88 - 1.76 (m, 1H). Synthesis of Compound 26 Preparation of 1,6-dimethyl-2-oxo-4-spiro[indane-2,4'-piperidine]-1'-yl-1,5- naphthyridine-3-carbonitrile (Compound 26)
To a mixture of 6-chloro-1-methyl-2-oxo-4-spiro[indane-2,4'-piperidine]-1'-yl-1,5- naphthyridine-3-carbonitrile (200 mg, 0.49 mmol, 1 eq), 2,4,6-trimethyl-1,3,5,2,4,6- trioxatriborinane (372 mg, 1.48 mmol, 0.41 mL, 50% purity, 3 eq) and potassium carbonate (137 mg, 0.99 mmol, 2 eq) in dioxane (2 mL) and water (0.4 mL) was added cyclopentyl(diphenyl)phosphane;dichloropalladium;iron (36 mg, 0.05 mmol, 0.1 eq) under nitrogen. The suspension was degassed under vacuum and purged with nitrogen 3 times. The mixture was then stirred under nitrogen at 90 °C for 12 h. The mixture was collected by filtration, and the residue was purified by prep-HPLC (column: Phenomenex C18 150*25mm*10um; mobile phase: [49-79% CH3CN in water (NH4HCO3)]) to give 1,6- dimethyl-2-oxo-4-spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile (17.9 mg, 0.05 mmol, 9% yield) as an off-white solid. LC/MS (ESI) m/z: 385.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J = 8.8 Hz, 1H), 7.56 (d, J = 8.8 Hz, 1H), 7.24 - 7.20 (m, 2H), 7.16 - 7.12 (m, 2H), 3.92 (s, 4H), 3.52 (s, 3H), 2.92 (s, 4H), 2.54 (s, 3H), 1.84 (t, J = 5.2 Hz, 4H). Synthesis of Compounds 27
Figure imgf000298_0001
Compound 27 Step 1: Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was made in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4-fluoro-2- hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl-piperidine-1- carboxylate. (yellow solid). LC/MS (ESI) m/z: 441.9 [M+H]+. Step 2: Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The enantiomer was separated by SFC (column: DAICEL CHIRALPAK AD(250mm*30mm, 10 um); mobile phase: 50% ethanol (0.1% NH4OH) in CO2) followed by prep-HPLC (column: Welch Ultimate C18150*25mm*5µm; mobile phase: [32-62% CH3CN in water (0.225% formic acid)]) to give 6-chloro-4-[(3R,4R)-4-(4-fluoro-2-hydroxy-anilino)-3- methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (48 mg, 0.11 mmol, 40% yield) as a yellow solid. Absolute configuration was arbitrarily assigned. LC/MS (ESI) m/z: 442.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.77 (s, 1H), 8.06 (d, J = 8.8 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 6.63 - 6.41 (m, 3H), 4.30 - 4.12 (m, 3H), 3.61 - 3.50 (m, 4H), 3.21 - 3.12 (m, 1H), 2.16 - 2.00 (m, 2H), 1.59 - 1.43 (m, 1H), 0.98 (d, J = 6.8 Hz, 3H). Synthesis of Compounds 29 and 28 Preparation of 6-chloro-4-[(3R,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 29) and 6-chloro-4- [(3S,4S)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 28) The title compounds were made in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6- chloro-4-[(3R,4S)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile starting from tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl- piperidine-1-carboxylate. 6-Chloro-4-[(3R,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (53.7 mg, 0.121 mmol, 27% yield) (Compound 29). (yellow solid). LC/MS (ESI) m/z: 442.1[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 7.17 - 6.97 (m, 2H), 6.78 - 6.55 (m, 2H), 5.80 (d, J = 8.8 Hz, 1H), 4.33 - 4.15 (m, 2H), 3.62 - 3.54 (m, 1H), 3.52 (s, 3H), 3.29 - 3.23 (m, 1H), 3.21 - 3.10 (m, 1H), 2.17 - 2.09 (m, 1H), 2.06 - 1.96 (m, 1H), 1.57 - 1.43 (m, 1H), 0.97 (d, J = 6.4 Hz, 3H). 6-Chloro-4-[(3S,4S)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (50.2 mg, 0.112 mmol, 25% yield, 98% purity) (Compound 28). (yellow solid). LC/MS (ESI) m/z: 442.1[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.06 (d, J = 8.8 Hz, 2H), 6.65 (d, J = 8.8 Hz, 2H), 5.80 (d, J = 9.2 Hz, 1H), 4.33 - 4.14 (m, 2H), 3.62 - 3.54 (m, 1H), 3.52 (s, 3H), 3.30 - 3.23 (m, 1H), 3.16 (dd, J = 10.8, 12.8 Hz, 1H), 2.18 - 2.08 (m, 1H), 2.07 - 1.92 (m, 1H), 1.55 - 1.44 (m, 1H), 0.97 (d, J = 6.4 Hz, 3H). Synthesis of Compound 30 Preparation of 6-chloro-4-[(3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 30) The title compound was made in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4-chloro-2- fluoro-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1- carboxylate. (yellow solid). LC/MS (ESI) m/z: 460.0 [M+H]+.1H NMR (400 MHz, DMSO- d6) δ 8.04 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.16 (dd, J = 2.4, 11.6 Hz, 1H), 7.00 (dd, J = 2.0, 8.8 Hz, 1H), 6.92 - 6.84 (m, 1H), 5.56 (dd, J = 2.0, 9.2 Hz, 1H), 4.32 - 4.16 (m, 2H), 3.60 - 3.56 (m, 1H), 3.52 (s, 3H), 3.44 - 3.36 (m, 1H), 3.20 - 3.12 (m, 1H), 2.24 - 2.12 (m, 1H), 2.08 - 2.00 (m, 1H), 1.64 (dq, J = 4.0, 12.0 Hz, 1H), 0.96 (d, J = 6.8 Hz, 3H). Synthesis of Compound 31 Step 1: Preparation of tert-butyl 3,4,4-trimethoxypiperidine-1-carboxylate To tert-butyl 3-hydroxy-4,4-dimethoxy-piperidine-1-carboxylate (500 mg, 1.91 mmol, 1 eq) in tetrahydrofuran (6 mL) at 0 °C was added sodium hydride (115 mg, 2.87 mmol, 60% purity, 1.5 eq), and the resulting mixture was stirred at 0 °C for 0.5 h. Iodomethane (543 mg, 3.83 mmol, 0.24 mL, 2 eq) was then added, and the reaction mixture was stirred at 0°C for 1 h under nitrogen. The mixture was diluted with saturated aqueous ammonium chloride (20 mL) and extracted with ethyl acetate (3 × 20 mL). The combined organic extract was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 3:1 petroleum ether/ethyl acetate) to give tert-butyl 3,4,4-trimethoxypiperidine-1-carboxylate (480 mg, 1.74 mmol, 91% yield) as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 4.11 (d, J = 14.0 Hz, 1H), 3.88 - 3.66 (m, 1H), 3.31 (s, 3H), 3.29 - 3.19 (m, 2H), 3.10 – 3.08 (m, 6H), 2.90 - 2.69 (m, 1H), 1.68 (d, J = 14.0 Hz, 1H), 1.57 - 1.45 (m, 1H), 1.39 (s, 9H). Step 2: Preparation of tert-butyl 3-methoxy-4-oxo-piperidine-1-carboxylate A mixture of tert-butyl 3,4,4-trimethoxypiperidine-1-carboxylate (330 mg, 1.20 mmol, 1 eq) and 4-methylbenzenesulfonic acid (41 mg, 0.24 mmol, 0.2 eq) in acetone (10 mL) and water (1 mL), was stirred at 80 °C for 1 h. The mixture was concentrated under vacuum to give tert- butyl 3-methoxy-4-oxo-piperidine-1-carboxylate (280 mg, crude) as an off-white oil. Step 3: Preparation of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1- carboxylate A mixture of tert-butyl 3-methoxy-4-oxo-piperidine-1-carboxylate (280 mg, 1.22 mmol, 1 eq), 4-fluoroaniline (135.70 mg, 1.22 mmol, 0.12 mL, 1 eq), and borane 2-methylpyridine (261 mg, 2.44 mmol, 2 eq) in methanol (1.5 mL) and acetic acid (1.5 mL) was stirred at 25 °C for 12 h. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep- HPLC (column: Phenomenex luna C18150*25mm* 10µm; mobile phase: [39-69% CH3CN in water (0.225% formic acid)]) to afford tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methoxy- piperidine-1-carboxylate (95 mg, 0.29 mmol, 24% yield) as a gray solid. LC/MS (ESI) m/z: 268.9 (M-55)+.1H NMR (400 MHz, DMSO-d6) δ 6.96 - 6.84 (m, 2H), 6.72 - 6.64 (m, 2H), 5.12 (d, J = 9.2 Hz, 1H), 4.16 (d, J = 13.2 Hz, 1H), 3.96 - 3.76 (m, 1H), 3.56 - 3.44 (m, 1H), 3.40 - 3.36 (m, 1H), 3.28 (s, 3H), 3.00 - 2.72 (m, 2H), 1.60 - 1.48 (m, 2H), 1.40 (s, 9H). Step 4: Preparation of tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy- piperidine-1-carboxylate A mixture of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1-carboxylate (90 mg, 0.28 mmol, 1 eq), formaldehyde (68 mg, 0.83 mmol, 0.06 mL, 37% purity, 3 eq), and sodium cyanoborohydride (52 mg, 0.83 mmol, 3 eq) in methanol (1 mL) was stirred at 25°C for 12 h. The mixture was diluted with water (5 mL) and extracted with ethyl acetate (5 mL*2). The combined organic extracts were washed with brine (10 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy-piperidine-1-carboxylate (89 mg, 0.26 mmol, 95% yield) as a colorless oil. LC/MS (ESI) m/z: 339.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.04 - 6.96 (m, 2H), 6.84 - 6.76 (m, 2H), 4.32 - 4.04 (m, 2H), 3.80 (d, J = 12.0 Hz, 1H), 3.52 - 3.36 (m, 1H), 3.20 (s, 3H), 2.92 (q, J = 13.6 Hz, 1H), 2.80 (s, 3H), 2.04 (d, J = 12.0 Hz, 1H), 1.40 (s, 11H). Step 5: Preparation of (3S,4R)-N-(4-fluorophenyl)-3-methoxy-N-methyl-piperidin-4- amine A mixture of tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy-piperidine-1- carboxylate (89 mg, 0.26 mmol, 1 eq) and trifluoroacetic acid (2 g, 13.51 mmol, 1 mL, 51.36 eq) in dichloromethane (1 mL) and stirred at 25 C for 15 minutes. The mixture was concentrated under vacuum to give (3S,4R)-N-(4-fluorophenyl)-3-methoxy-N-methyl- piperidin-4-amine (100 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 239.1 [M+H]+. Step 6: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 31) A mixture of (3S,4R)-N-(4-fluorophenyl)-3-methoxy-N-methyl-piperidin-4-amine (100 mg, 0.28 mmol, 1 eq, TFA salt), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (50 mg, 196.79 µmol, 6.93e-1 eq), and diisopropylethylamine (371.00 mg, 2.87 mmol, 0.5 mL, 10.11 eq) in acetonitrile (1 mL) was stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the crude product was triturated in DMF (0.5 mL), washed with acetonitrile (1.5 mL), and collected by filtration to afford 6-chloro-4-[(3S,4R)-4-(4- fluoro-N-methyl-anilino)-3-methoxy-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (60.8 mg, 0.13 mmol, 46% yield) as a yellow solid. LC/MS (ESI) m/z: 456.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.84 (d, J = 8.8 Hz, 1H), 7.08 - 7.00 (m, 2H), 6.88 (dd, J = 4.4, 9.2 Hz, 2H), 4.48 (dd, J = 2.4, 13.2 Hz, 1H), 4.36 (d, J = 13.6 Hz, 1H), 4.12 (d, J = 11.6 Hz, 1H), 3.80 (d, J = 13.6 Hz, 1H), 3.64 (s, 1H), 3.52 (s, 3H), 3.48 (t, J = 11.6 Hz, 1H), 3.04 (s, 3H), 2.88 (s, 3H), 2.84 - 2.80 (m, 1H), 1.68 (d, J = 9.6 Hz, 1H). Synthesis of Compound 32 Step 1: Preparation of tert-butyl (3S,4R)-3-methyl-4-[4-(trifluoromethoxy) anilino]piperidine-1-carboxylate. A mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (2 g, 9.38 mmol, 1 eq) ,4- (trifluoromethoxy)aniline (1.66 g, 9.38 mmol, 1 mL, 1 eq), sodium cyanoborohydride (884 mg, 14.07 mmol, 1.5 eq), and acetic acid (56 mg, 0.938 mmol, 0.05 mL, 0.1 eq) in methanol (20 mL) was stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the crude product was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC (column: Welch Xtimate C18250*50mm*10 µm; mobile phase: [35-70% CH3CN in water (TFA)]) to give tert-butyl (3S,4R)-3-methyl-4-[4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (140 mg, 0.374 mmol, 4% yield) as a yellow solid. LC/MS (ESI) m/z: 319.2 (M- 55)+.1H NMR (400 MHz, DMSO-d6) δ 7.04 (d, J = 8.4 Hz, 2H), 6.68 (d, J = 9.2 Hz, 2H), 5.72 (d, J = 8.4 Hz, 1H), 3.52 (dt, J = 4.4, 8.4 Hz, 2H), 3.32 (s, 1H), 3.2 (dd, J = 3.2, 13.2 Hz, 1H), 2.04 - 1.96 (m, 1H), 1.6 - 1.48 (m, 2H), 1.40 (s, 9H), 1.32 - 1.16 (m, 1H), 0.8 (d, J = 6.8 Hz, 3H). Step 2: Preparation of tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy) anilino]piperidine-1-carboxylate. A mixture of tert-butyl (3S,4R)-3-methyl-4-[4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (140 mg, 0.37 mmol, 1 eq), formaldehyde (91 mg, 1.12 mmol, 0.084 mL, 37% purity, 3 eq), and sodium cyanoborohydride (71 mg, 1.12 mmol, 3 eq) in methanol (1.5 mL) was stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate (99 mg, 255 mmol, 68% yield) as a white solid. LC/MS (ESI) m/z: 333.1 (M-55)+.1H NMR (400 MHz, DMSO-d6) δ 7.12 (d, J = 8.4 Hz, 2H), 6.8 (d, J = 7.6 Hz, 2H), 4.16 - 3.96 (m, 1H), 3.92 - 3.68 (m, 2H), 3.16 - 3.00 (m, 1H), 2.80 (s, 3H), 2.16 - 1.96 (m, 2H), 1.52 - 1.48 (m, 1H), 1.40 (s, 9H), 1.28 - 1.12 (m, 1H), 0.80 (d, J = 6.0 Hz, 3H). Step 3: Preparation of (3S,4R)-N,3-dimethyl-N-[4- (trifluoromethoxy)phenyl]piperidin-4-amine. A mixture of tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate (99 mg, 0.26 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 52.99 eq) in dichloromethane (1 mL) was stirred at 25 °C for 15 minutes. The mixture was concentrated under vacuum to give (3S,4R)- N,3-dimethyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (110 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 289.1 [M+H]+. Step 4: Preparation of 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 32) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N,3- dimethyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 506.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.8 (d, J = 8.8 Hz, 1H), 7.2 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 9.2 Hz, 2H), 4.4 - 4.28 (m, 1H), 4.16 (td, J = 3.6, 12.0 Hz, 1H), 4.04 - 3.96 (m, 1H), 3.92 - 3.84 (m, 1H), 3.52 (s, 3H), 3.48 - 3.44 (m, 1H), 2.88 (s, 3H), 2.76 - 2.64 (m, 1H), 2.4 - 2.28 (m, 1H), 1.80 - 1.72 (m, 1H), 0.88 (d, J = 7.2 Hz, 3H). Synthesis of Compounds 33 and 34
Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate and tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate To a mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (2 g, 9.38 mmol, 1 eq) and 4-chloroaniline (1.20 g, 9.38 mmol, 1 eq) in methanol (20 mL) was added sodium cyanoborohydride (1.47 g, 23.44 mmol, 2.5 eq), and the reaction mixture was stirred at 40 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Welch Xtimate C18250*50mm*10 µm; mobile phase: [40- 75% CH3CN in water (0.225% formic acid)]) to give the desired products. tert-Butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (223 mg, 0.69 mmol, 7.3% yield) was obtained as a white solid.1H NMR (400 MHz, DMSO-d6) δ 7.08 - 7.03 (m, 2H), 6.66 - 6.61 (m, 2H), 5.63 (d, J = 8.4 Hz, 1H), 3.79 - 3.60 (m, 1H), 3.57 - 3.47 (m, 2H), 3.30 - 3.27 (m, 1H), 3.16 (dd, J = 3.6, 13.6 Hz, 1H), 2.05 - 1.97 (m, 1H), 1.57 - 1.47 (m, 2H), 1.39 (s, 9H), 0.77 (d, J = 6.8 Hz, 3H). tert-Butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (153 mg, 0.48 mmol, 5.0% yield) was obtained as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 7.09 - 7.00 (m, 2H), 6.61 - 6.53 (m, 2H), 5.80 - 5.45 (m, 1H), 3.87 (d, J = 13.2 Hz, 2H), 3.08 - 2.96 (m, 1H), 2.86 (s, 1H), 2.63 - 2.52 (m, 1H), 1.93 - 1.84 (m, 1H), 1.51 - 1.43 (m, 1H), 1.40 (s, 9H), 1.15 - 1.02 (m, 1H), 0.89 (d, J = 6.4 Hz, 3H). Step 2: Preparation of (3S,4R)-N-(4-chlorophenyl)-3-methyl-piperidin-4-amine To a solution of tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (100 mg, 0.31 mmol, 1 eq) in dichloromethane (1 mL) was added dropwise trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 43.87 eq), and the resulting mixture was stirred at 25 °C for 30 minutes. The reaction mixture was concentrated under reduced pressure to give (3S,4R)- N-(4-chlorophenyl)-3-methyl-piperidin-4-amine (100 mg, crude, TFA salt) as a brown oil. Step 3: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid).1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.10 - 7.05 (m, 2H), 6.75 - 6.70 (m, 2H), 5.90 (d, J = 8.4 Hz, 1H), 4.08 - 3.97 (m, 1H), 3.85 (d, J = 4.4 Hz, 2H), 3.81 - 3.72 (m, 1H), 3.70 - 3.61 (m, 1H), 3.52 (s, 3H), 2.33 - 2.26 (m, 1H), 2.14 - 2.01 (m, 1H), 1.87 - 1.82 (m, 1H), 0.86 (d, J = 7.2 Hz, 3H). Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 33) and 6- chloro-4-[(3R,4S)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 34) The enantiomers were separated by SFC (column: DAICEL CHIRALCEL OJ(250mm*30mm,10µm); mobile phase: 40% methanol (0.1%NH4OH) in CO2) followed by prep-HPLC (column: Phenomenex luna C18150*25mm* 10µm; mobile phase: [50-80% CH3CN in water (0.225% formic acid)]). Absolute configuration was arbitrarily assigned. 6-Chloro-4-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (33.8 mg, 0.076 mmol, 39% yield) (Compound 33). (yellow solid). LC/MS (ESI) m/z: 442.1 [M+1]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 8.8 Hz, 1H), 7.84 - 7.78 (m, 1H), 7.07 (d, J = 8.8 Hz, 2H), 6.80 - 6.63 (m, 2H), 5.94 - 5.86 (m, 1H), 4.06 - 3.96 (m, 1H), 3.85 (d, J = 4.8 Hz, 2H), 3.80 - 3.74 (m, 1H), 3.70 - 3.63 (m, 1H), 3.52 (s, 3H), 2.32 - 2.26 (m, 1H), 2.14 - 2.00 (m, 1H), 1.90 - 1.79 (m, 1H), 0.86 (d, J = 7.2 Hz, 3H). 6-Chloro-4-[(3R,4S)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (31.1 mg, 68.37 umol, 35% yield) (Compound 34). (yellow solid). LC/MS (ESI) m/z: 442.1 [M+1]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.11 - 7.03 (m, 2H), 6.72 (d, J = 8.8 Hz, 2H), 5.94 - 5.86 (m, 1H), 4.11 - 3.96 (m, 1H), 3.85 (d, J = 4.8 Hz, 2H), 3.79 - 3.74 (m, 1H), 3.69 - 3.63 (m, 1H), 3.52 (s, 3H), 2.31 - 2.26 (m, 1H), 2.14 - 2.01 (m, 1H), 1.89 - 1.79 (m, 1H), 0.86 (d, J = 7.2 Hz, 3H). Synthesis of Compound 35 Preparation of 6-chloro-4-[(3R,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 35) The title compound was made in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4-chloro-2- fluoro-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3R,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1- carboxylate. (yellow solid). LC/MS (ESI) m/z: 460.0 [M+H]+.1H NMR (400 MHz, DMSO- d6) δ 8.04 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.2 (dd, J = 2.4, 12.0 Hz, 1H), 7.00 (dd, J = 2.0, 8.8 Hz, 1H), 6.92 - 6.84 (m, 1H), 5.56 (dd, J = 2.0, 9.2 Hz, 1H), 4.32 - 4.20 (m, 2H), 3.60 - 3.56 (m, 1H), 3.52 (s, 3H), 3.44 - 3.36 (m, 1H), 3.16 - 3.08 (m, 1H), 2.20 - 2.12 (m, 1H), 2.04 (dd, J = 3.2, 13.2 Hz, 1H), 1.72 - 1.60 (m, 1H), 0.96 (d, J = 6.8 Hz, 3H).
Synthesis of Compound 36 Step 1: Preparation of tert-butyl 4-(4-chloro-N-methyl-anilino) piperidine-1- carboxylate A mixture of tert-butyl 4-(4-chloroanilino)piperidine-1-carboxylate (200 mg, 0.64 mmol, 1 eq), sodium cyanoborohydride (61 mg, 0.97 mmol, 1.5 eq), and formaldehyde (57 mg, 0.71 mmol, 0.05 mL, 37% purity, 1.1 eq) in methanol (2 mL) was stirred at 25 °C for 12 hours. The mixture was concentrated under vacuum, and the resulting residue was purified by prep- HPLC (column: Waters xbridge 150*25mm 10um; mobile phase: [56-86% CH3CN in water (NH4HCO3)]) to give tert-butyl 4-(4-chloro-N-methyl-anilino) piperidine-1-carboxylate (114 mg, 0.351 mmol, 55% yield) as a white solid. LC/MS (ESI) m/z: 268.9 [M-55]+. Step 2: Preparation of (3R,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine A mixture of tert-butyl 4-(4-chloro-N-methyl-anilino)piperidine-1-carboxylate (114 mg, 0.35 mmol, 1 eq) and trifluoroacetic acid (80 mg, 0.7 mmol, 0.05 mL, 2 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes. The mixture was concentrated under vacuum to give N-(4-chlorophenyl)-N-methyl-piperidin-4-amine (190 mg, crude, trifluoroacetic acid) as a red oil. LC/MS (ESI) m/z: 225.2 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-(4-chloro-N-methyl-anilino)-1-piperidyl]-2-oxo- 1H-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from N-(4- chlorophenyl)-N-methyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 442.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.84 (d, J = 9.2 Hz, 1H), 7.20 (d, J = 9.2 Hz, 2H), 6.8 (d, J = 9.2 Hz, 2H), 4.28 (d, J = 12.4 Hz, 2H), 4.16 - 4.04 (m, 1H), 3.6 - 3.52 (m, 2H), 3.53 (s, 3H), 2.72 (s, 3H), 2.04 – 2.0 (m, 2H), 1.76 (d, J = 12.4 Hz, 2H). Synthesis of Compound 37 Step 1: Preparation of tert-butyl (3S,4R)-4-(4-fluoro-2-methyl-anilino)-3-methyl- piperidine-1-carboxylate To a solution of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (2 g, 9.38 mmol, 1 eq) and 4-fluoro-2-methyl-aniline (1.17 g, 9.38 mmol, 1.0 eq) in methanol (20 mL) was added sodium cyanoborohydride (1.47 g, 23.44 mmol, 2.5 eq), and the reaction mixture was stirred at 40 °C for 12 hr. The mixture concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18 (250*70mm,10 µm); mobile phase: [30-95% CH3CN in water (0.225% formic acid)]) to give tert-butyl (3S,4R)-4- (4-fluoro-2-methyl-anilino)-3-methyl-piperidine-1-carboxylate (250 mg, 0.78 mmol, 8% yield) as a brown solid. LC/MS (ESI) m/z: 267.0 [M-55] +.1H NMR (400 MHz, DMSO-d6) δ 6.90 - 6.74 (m, 2H), 6.64 - 6.52 (m, 1H), 4.13 - 4.02 (m, 1H), 3.93 - 3.77 (m, 1H), 3.70 - 3.62 (m, 1H), 3.57 (td, J = 4.4, 8.4 Hz, 1H), 3.29 (s, 1H), 3.13 - 3.05 (m, 1H), 2.98 - 2.84 (m, 1H), 2.11 - 2.07 (m, 3H), 1.70 - 1.62 (m, 1H), 1.58 - 1.51 (m, 1H), 1.40 (s, 9H), 0.77 (d, J = 6.8 Hz, 3H). Step 2: Preparation of tert-butyl (3S,4R)-4-(4-fluoro-N,2-dimethyl-anilino)-3-methyl- piperidine-1-carboxylate To a mixture of tert-butyl (3S,4R)-4-(4-fluoro-2-methyl-anilino)-3-methyl-piperidine-1- carboxylate (150 mg, 0.47 mmol, 1 eq), sodium cyanoborohydride (88 mg, 1.40 mmol, 3 eq), and formaldehyde (189 mg, 2.33 mmol, 0.17 mL, 37% purity, 5 eq) in methanol (1 mL) and tetrahydrofuran (1 mL) was added acetic acid (105 mg, 1.75 mmol, 0.1 mL, 3.76 eq), and the reaction mixture was stirred at 40 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100:1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3S,4R)-4-(4-fluoro- N,2-dimethyl-anilino)-3-methyl-piperidine-1-carboxylate (87 mg, 0.26 mmol, 56 % yield) as a colorless oil. LC/MS (ESI) m/z: 337.1 [M+H] +.1H NMR (400MHz, DMSO-d6) δ 7.23 (dd, J = 5.6, 8.4 Hz, 1H), 7.08 - 6.92 (m, 2H), 4.17 - 3.67 (m, 2H), 3.56 - 3.39 (m, 1H), 2.98 (td, J = 4.0, 11.6 Hz, 1H), 2.93 - 2.74 (m, 1H), 2.41 (s, 3H), 2.23 (s, 3H), 2.14 - 2.00 (m, 1H), 1.37 (s, 9H), 1.31 - 1.23 (m, 1H), 1.21 - 1.11 (m, 1H), 0.83 - 0.74 (m, 3H). Step 3: Preparation of (3S,4R)-N-(4-fluoro-2-methyl-phenyl)-N,3-dimethyl-piperidin- 4-amine A mixture of tert-butyl (3S,4R)-4-(4-fluoro-N,2-dimethyl-anilino)-3-methyl-piperidine-1- carboxylate (90 mg, 0.27 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1.00 mL, 50.49 eq) in dichloromethane (1 mL) was stirred at 25 °C for 30 min under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give (3S,4R)-N-(4-fluoro- 2-methyl-phenyl)-N,3-dimethyl-piperidin-4-amine (90 mg, crude, TFA salt) as a colorless oil. LC/MS (ESI) m/z: 237.0 [M+H]+. Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N,2-dimethyl-anilino)-3- methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 37) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- fluoro-2-methyl-phenyl)-N,3-dimethyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 454.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 8.8 Hz, 1H), 7.79 (d, J = 8.8 Hz, 1H), 7.32 (dd, J = 5.6, 8.8 Hz, 1H), 7.10 - 6.97 (m, 2H), 4.27 - 4.01 (m, 2H), 3.70 - 3.59 (m, 1H), 3.51 (s, 3H), 3.28 - 3.19 (m, 2H), 2.47 (s, 3H), 2.27 (s, 4H), 1.99 - 1.85 (m, 1H), 1.56 - 1.46 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H). Synthesis of Compound 38
Step 1: Preparation of O2-benzyl O5-tert-butyl (3aR,6aR)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-2,5-dicarboxylate To a mixture of tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (500 mg, 2.36 mmol, 1 eq) and triethylamine (715 mg, 7.07 mmol, 0.98 mL, 3 eq) in dichloromethane (10 mL) was added benzyl chloroformate (482 mg, 2.83 mmol, 0.4 mL, 1.2 eq), and the reaction mixture was stirred at 25°C for 12 h under N2. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (20 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC (column: Phenomenex C18250*50mm*10 um; mobile phase: [40-70% CH3CN in water (NH4HCO3)]) to afford O2-benzyl O5-tert-butyl (3aR,6aR)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-2,5-dicarboxylate (95 mg, 0.27 mmol, 12% yield) as a colorless oil. LC/MS (ESI) m/z: 291.1 [M-55]+. Step 2: Preparation of benzyl (3aS,6aS)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole-5-carboxylate To a solution of O2-benzyl O5-tert-butyl (3aR,6aR)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-2,5-dicarboxylate (95 mg, 0.27 µmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 24.6 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under reduced pressure to give benzyl (3aS,6aS)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate (90 mg, crude, TFA salt) as a brown oil. LC/MS (ESI) m/z: 247.0 [M+H] +. Step 3: Preparation of benzyl (3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate A mixture of benzyl (3aS,6aS)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (250 mg, 0.69 mmol, 1 eq, trifluoroacetic acid) , 4-fluoro-1-iodo-2-methyl- benzene (245 mg, 1.04 mmol, 1.5 eq), Cs2CO3 (1.13 g, 3.47 mmol, 5 eq), and 1,3-bis[2,6- bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (34 mg, 0.03 mmol, 0.05 eq) in dioxane (3 mL) was degassed and purged with nitrogen (3X), and the reaction mixture was stirred at 100 °C for 4 hours under nitrogen atmosphere. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (25 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give benzyl (3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (200 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 355.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.42 - 7.30 (m, 5H), 6.87 - 6.76 (m, 3H), 5.21 - 5.13 (m, 2H), 3.82 - 3.73 (m, 2H), 3.39 - 3.31 (m, 2H), 3.21 - 3.13 (m, 4H), 2.42 - 2.31 (m, 2H), 2.28 (s, 3H). Step 4: Preparation of (3aS,6aS)-5-(4-fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole To a solution of benzyl (3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (200 mg, 564.31 umol, 1 eq) in CH3OH (5 mL) was added palladium on activated carbon catalyst (50 mg, 10% purity) under nitrogen, and the resulting suspension was degassed under vacuum and purged with hydrogen several times. The mixture was then stirred under hydrogen (15 psi) at 30°C for 12 hours. The mixture was concentrated to give (3aS,6aS)-5-(4-fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole (120 mg, crude) as a colorless oil. LC/MS (ESI) m/z: 221.1 [M+H]+. Step 5: Preparation of 4-[(3aR,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 38) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aS,6aS)-5-(4- fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (yellow solid). LC/MS (ESI) m/z: 438.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.01 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 6.94 (d, J = 9.6 Hz, 1H), 6.88 - 6.82 (m, 2H), 4.26 - 4.14 (m, 4H), 3.50 (s, 3H), 3.46 (d, J = 10.0 Hz, 2H), 3.23 - 3.17 (m, 2H), 2.46 - 2.39 (m, 2H), 2.27 (s, 3H). Synthesis of Compound 39
Figure imgf000323_0001
Step 1: Preparation of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl]piperazine-1- carboxylate A mixture of tert-butyl piperazine-1-carboxylate (357 mg, 1.92 mmol, 1 eq), 4-chloro-2- hydroxy-benzaldehyde (300 mg, 1.92 mmol, 1 eq), and sodium cyanoborohydride (241 mg, 3.83 mmol, 2 eq) in methanol (3 mL) was stirred at 25 °C for 12 h. The mixture was concentrated under vacuum, and the resulting crude product was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC (column: Phenomenex C18250*50mm*10 µm; mobile phase: [49-79% CH3CN in water (NH4HCO3)]) to give tert-butyl 4-[(4-chloro-2- hydroxy-phenyl)methyl]piperazine-1-carboxylate (530 mg, 1.62 mmol, 85% yield) as a yellow oil. LC/MS (ESI) m/z: 327.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ 6.88 (d, J = 8.0 Hz, 1H), 6.84 (d, J = 2.0 Hz, 1H), 6.8 - 6.8 (m, 1H), 3.68 (s, 2H), 3.6 - 3.2 (m, 4H), 2.52 (s, 4H), 1.48 (s, 9H). Step 2: Preparation of 5-chloro-2-(piperazin-1-ylmethyl)phenol A mixture of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl]piperazine-1-carboxylate (20 mg, 0.06 mmol, 1 eq) and trifluoroacetic acid (154 mg, 1.35 mmol, 0.1 mL, 22.07 eq) in dichloromethane (0.2 mL) was stirred at 20 °C for 0.5 h. The mixture was concentrated to give 5-chloro-2-(piperazin-1-ylmethyl)phenol (21 mg, crude, TFSA salt) as a yellow oil. LC/MS (ESI) m/z: 227.2 [M+H]+. Step 3: Preparation of 7-{4-[(4-chloro-2-hydroxyphenyl)methyl]piperazin-1-yl}-2,4- dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile (Compound 39) The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 5- chloro-2-(piperazin-1-ylmethyl)phenol. (white solid). LC/MS (ESI) m/z: 430.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.21 (d, J=8.8 Hz, 1H), 6.87 - 6.79 (m, 2H), 3.93 (s, 4H), 3.61 (s, 2H), 3.48 (s, 3H), 2.68 (s, 3H), 2.61 (s, 4H).
Synthesis of Compound 40 Step 1: Preparation of tert-butyl (3aS,6aR)-2-(4-fluoro-2-methyl-phenyl)- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a mixture of tert-butyl (3aS,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (400 mg, 1.88 mmol, 1 eq), 1-bromo-4-fluoro-2-methyl-benzene (392 mg, 2.07 mmol, 1.1 eq), and potassium carbonate (781 mg, 5.65 mmol, 3 eq) in dioxane (4 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (91 mg, 0.94 mmol, 0.05 eq) under nitrogen atmosphere, and the reaction mixture was degassed, purged with nitrogen (3X), and then stirred at 100 °C for 4 h. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3aS,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate (117 mg, 0.37 mmol, 19% yield) as a white solid. LC/MS (ESI) m/z: 265.0 [M- 55]+.1H NMR (400 MHz, CDCl3) δ 6.8 - 6.8 (m, 3H), 3.68 (s, 2H), 3.32 (s, 2H), 3.12 (s, 2H), 2.92 (s, 4H), 2.28 (s, 3H), 1.44 (s, 9H). Step 2: Preparation of (3aR,6aS)-5-(4-fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aS,6aR)-2-(4-fluoro-2-methyl-phenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (110 mg, 0.34 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 1 eq) in dichloromethane (1 mL) was stirred at 25℃ for 15 minutes. The mixture was concentrated under vacuum to give (3aR,6aS)-5-(4-fluoro-2- methyl-phenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (190 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 221.0 (M+H)+. Step 3 Preparation of 6-chloro-4-((3aR,6aS)-5-(4-fluoro-2- methylphenyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile (Compound 40) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-(4- fluoro-2-methyl-phenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (yellow solid). LC/MS (ESI) m/z: 438.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.0 (d, J = 8.4 Hz, 1H), 7.8 (d, J = 8.8 Hz, 1H), 7.2 - 6.8 (m, 3H), 4.48 - 4.36 (m, 2H), 4.28 (d, J = 12.0 Hz, 2H), 3.52 (s, 3H), 3.16 (d, J = 6.0 Hz, 2H), 3.08 (d, J = 9.2 Hz, 4H), 2.16 (s, 3H).
Synthesis of Compound 41 Step 1: Preparation of O5-benzyl O1-tert-butyl 3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-1,5-dicarboxylate To a mixture of tert-butyl 2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine-1-carboxylate (250 mg, 1.10 mmol, 1 eq) and triethylamine (223 mg, 2.21 mmol, 0.31 mL, 2 eq) in dichloromethane (5 mL) was added benzyl chloroformate (226 mg, 1.33 mmol, 0.19 mL, 1.2 eq) dropwise, and the reaction mixture stirred at 20 °C for 12 h under N2. The mixture was concentrated, and the resulting residue was purified by prep-HPLC (column: Waters xbridge 150*25mm 10um; mobile phase: [42-72% CH3CN in water (NaHCO3)]) to give O5-benzyl O1-tert-butyl 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1,5-dicarboxylate (170 mg, 0.47 mmol, 43% yield) as a yellow solid. LC/MS (ESI) m/z: 383.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.38 - 7.29 (m, 5H), 5.14 (d, J=3.2 Hz, 2H), 3.93 (s, 3H), 3.47 - 3.27 (m, 3H), 3.02 - 2.75 (m, 1H), 2.40 - 2.19 (m, 1H), 2.05 - 1.92 (m, 1H), 1.91 - 1.75 (m, 2H), 1.66 - 1.58 (m, 1H), 1.46 (s, 9H). Step 2: Preparation of benzyl 1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate A mixture of O5-benzyl O1-tert-butyl 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine- 1,5-dicarboxylate (170 mg, 0.47 mmol, 1 eq) and trifluoroacetic acid (3.08 g, 27.01 mmol, 2 mL, 57.27 eq) in dichloromethane (2 mL) was stirred at 25 °C for 15 minutes. The mixture was concentrated under vacuum to give benzyl 1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2- c]pyridine-5-carboxylate (200 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 261.0 [M+H]+. Step 3: Preparation of benzyl 1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate
To a mixture of benzyl 1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5-carboxylate (200 mg, 0.53 mmol, 1 eq, trifluoroacetic acid) ,1-fluoro-4-iodo-benzene (296 mg, 1.34 mmol, 0.15 mL, 2.5 eq), and Cs2CO3 (522 mg, 1.60 mmol, 3 eq) in DMF (2 mL) was added 1,3- bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine; dichloropalladium (26 mg, 0.027 mmol, 0.05 eq) was added under nitrogen atmosphere, and the reaction mixture was stirred at 80 °C for 3 h. The mixture was diluted with water (8 mL) and extracted with ethyl acetate (8 mL*2). The combined organic extracts were washed with brine (16 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give benzyl 1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (110 mg, 0.31 mmol, 58% yield) as a white solid. LC/MS (ESI) m/z: 355.2 [M+H]+. Step 4: Preparation of 1-(4-fluorophenyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2- c]pyridine To a solution of benzyl 1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (108 mg, 0.31 mmol, 1 eq) in CH3OH (3 mL) was added palladium on activated carbon catalyst (30 mg, 10% purity) under nitrogen, and the resulting suspension was degassed under vacuum and purged with hydrogen several times. The mixture was stirred under hydrogen (15 psi) at 25°C for 12 hours. The mixture was filtered, and the filtrate was concentrated to give 1-(4-fluorophenyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2- c]pyridine (70 mg, crude) as a colorless oil. LC/MS (ESI) m/z: 221.0 [M+H]+. Step 5: Preparation of 4-[(3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 41) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from 1-(4- fluorophenyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine. (yellow solid). LC/MS (ESI) m/z: 438.1 [M+H]+.1H NMR (400 MHz, DMSO) δ 8.08 (d, J = 9.2 Hz, 1H), 7.8 (d, J = 9.2 Hz, 1H), 7.00 (t, J = 8.8 Hz, 2H), 6.56 (dd, J = 4.4, 8.8 Hz, 2H), 4.24 - 4.08 (m, 2H), 4.04 - 3.88 (m, 2H), 3.52 (s, 3H), 3.49 - 3.4 (m, 1H), 3.24 - 3.16 (m, 1H), 2.6 (d, J = 6.8 Hz, 2H), 2.24 - 2.12 (m, 1H), 2.12 - 2.04 (m, 2H), 1.84 - 1.72 (m, 1H). Synthesis of Compound 43
Step 1: Preparation of tert-butyl 4-(4-chloroanilino)piperidine-1-carboxylate A mixture of tert-butyl 4-oxopiperidine-1-carboxylate (600 mg, 3.01 mmol, 1 eq), 4- chloroaniline (384 mg, 3.01 mmol, 1 eq), sodium cyanoborohydride (473.10 mg, 7.53 mmol, 2.5 eq), and acetic acid (18 mg, 0.30 mmol, 0.017 mL, 0.1 eq) in methanol (6 mL) was stirred at 25°C for 12 h. The mixture was concentrated under vacuum, and the crude product was diluted with water (5 mL) and extracted with ethyl acetate (5 mL*2). The combined organic extracts were washed with brine (10 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100:1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl 4-(4-chloroanilino)piperidine-1- carboxylate (250 mg, 0.80 mmol, 27% yield) as a white solid. LC/MS (ESI) m/z: 255.1 [M- 55]+.1H NMR (400 MHz, CDCl3) δ 7.18 - 7.05 (m, 2H), 6.58 - 6.50 (m, 2H), 4.05 (d, J = 8.0 Hz, 2H), 2.92 (t, J = 12.0 Hz, 2H), 2.05 - 1.98 (m, 2H), 1.47 (s, 11H), 1.32 (d, J = 10.8 Hz, 2H). Step 2: Preparation of N-(4-chlorophenyl)piperidin-4-amine A mixture of tert-butyl 4-(4-chloroanilino)piperidine-1-carboxylate (62 mg, 0.20 mmol, 1 eq) and trifluoroacetic acid (924 mg, 8.10 mmol, 0.6 mL, 83.96 eq) in dichloromethane (0.6 mL) was stirred at 25 °C for 15 minutes. The mixture was concentrated under vacuum to give N- (4-chlorophenyl)piperidin-4-amine (100 mg, crude, trifluoroacetic acid) as a red oil. LC/MS (ESI) m/z: 211.0 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-(4-chloroanilino)-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile A mixture of N-(4-chlorophenyl)piperidin-4-amine (41 mg, 0.13 mmol, 6.49e-1 eq, trifluoroacetic acid), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (50 mg, 0.2 mmol, 1 eq) and diisopropylethylamine (76 mg, 0.59 mmol, 0.10 mL, 3 eq) in acetonitrile (0.6 mL) was stirred at 40 °C for 12 hours. The mixture was concentrated under vacuum, and the resulting precipitate was triturated in dimethylformamide (1.5 mL), washed with acetonitrile (1.5 mL), and collected by filtration to afford 6-chloro-4-[4-(4-chloroanilino)-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (30.4 mg, 0.07 mmol, 35% yield) as a yellow solid. LC/MS (ESI) m/z: 428.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 7.09 (d, J = 8.8 Hz, 2H), 6.66 (d, J = 8.8 Hz, 2H), 5.88 (d, J = 8.0 Hz, 1H), 4.20 (br d, J = 12.8 Hz, 2H), 3.64 - 3.56 (m, 2H), 3.53 (s, 3H), 3.30 (s, 1H), 2.10 (d, J = 10.8 Hz, 2H), 1.77 - 1.55 (m, 2H).
Synthesis of Compound 44
Figure imgf000335_0001
Step 1: Preparation of ethyl 3-acetamido-6-chloro-pyridine-2-carboxylate A mixture of ethyl 3-amino-6-chloro-pyridine-2-carboxylate (4 g, 19.94 mmol, 1 eq), and acetic anhydride (2.04 g, 19.94 mmol, 1.87 mL, 1 eq) in dioxane (40 mL) was stirred at 110°C for 12 h. The mixture was concentrated under vacuum, and the resulting residue was purified by silica gel flash chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give ethyl 3-acetamido-6-chloro-pyridine-2-carboxylate (5 g, crude) as a yellow solid. LC/MS (ESI) m/z: 243.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ 11.00 (s, 1H), 9.12 (d, J = 9.2 Hz, 1H), 7.48 (d, J = 8.8 Hz, 1H), 4.51 (q, J = 7.2 Hz, 2H), 2.28 (s, 3H), 1.47 (t, J = 7.2 Hz, 3H). Step 2: Preparation of ethyl 3-[acetyl(methyl)amino]-6-chloro-pyridine-2-carboxylate A mixture of ethyl 3-acetamido-6-chloro-pyridine-2-carboxylate (200 mg, 0.82 mmol, 1 eq), methyl iodide (234 mg, 1.65 mmol, 0.1 mL, 2 eq), and Cs2CO3 (671 mg, 2.06 mmol, 2.5 eq) in DMF (2 mL) was stirred at 25 °C for 12 h. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic layers were washed with brine (20 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by column chromatograph on silica gel (gradient: 0 to 1:1 petroleum ether/ethyl acetate) followed by prep-HPLC (column: Waters xbridge 150*25mm 10µm; mobile phase: [10-40% CH3CN in water (NH4HCO3)]) to give ethyl 3- [acetyl(methyl)amino]-6-chloro-pyridine-2-carboxylate (110 mg, 0.43 mmol, 52% yield) as a white solid. LC/MS (ESI) m/z: 257.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.12 (s, 1H), 7.6 (s, 1H), 4.48 - 4.4 (m, 2H), 3.08 (s, 3H), 1.92 - 1.84 (m, 3H), 1.40 (t, J = 7.2 Hz, 3H). Step 3: Preparation of 6-chloro-4-hydroxy-1-methyl-1,5-naphthyridin-2-one To a solution of ethyl 3-[acetyl(methyl)amino]-6-chloro-pyridine-2-carboxylate (110 mg, 0.43 mmol, 1 eq) in tetrahydrofuran (1 mL) at 0°C was added dropwise potassium bis(trimethylsilyl)amide (1 M, 0.64 mL, 1.5 eq), and the reaction mixture was stirred at 0 °C for 2 hr. The pH of the mixture was adjusted to 5 by addition of 1 M hydrochloric acid. The mixture was filtered, and the residue was washed with acetonitrile (2 mL) to give 6-chloro-4- hydroxy-1-methyl-1,5-naphthyridin-2-one (32 mg, 0.15 mmol, 35% yield) as a white solid. LC/MS (ESI) m/z: 210.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.01 (d, J = 8.8 Hz, 1H), 7.72 (d, J = 8.8 Hz, 1H), 6.10 (s, 1H), 3.52 (s, 3H) Step 4: Preparation of (6-chloro-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate A mixture of 6-chloro-4-hydroxy-1-methyl-1,5-naphthyridin-2-one (32 mg, 0.15 mmol, 1 eq), trifluoromethanesulfonic anhydride (107 mg, 0.38 mmol, 0.062 mL, 2.5 eq), triethylamine (77 mg, 0.76 mmol, 0.11 mL, 5 eq), and 4-dimethylaminopyridine (2 mg, 0.015 mmol, 0.1 eq) in dichloromethane (1 mL) was stirred at 0 °C for 2 hr under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give (6-chloro-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (33 mg, 0.095 mmol, 63% yield) as a yellow solid. LC/MS (ESI) m/z: 342.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.23 (d, J = 8.8 Hz, 1H), 7.91 (d, J = 8.8 Hz, 1H), 7.31 (s, 1H), 3.63 (s, 3H). Step 5: Preparation of tert-butyl 2-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate A mixture of tert-butyl 4-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (200 mg, 0.88 mmol, 1 eq), 1-bromo-4-fluoro-benzene (185 mg, 1.06 mmol, 0.12 mL, 1.2 eq), Cs2CO3 (864 mg, 2.65 mmol, 3 eq), and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]- 4,5-dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (43 mg, 0.044 mmol, 0.05 eq) in dioxane (5 mL) was stirred at 100°C for 3 h under N2. The mixture was diluted with water (30 mL) and extracted with ethyl acetate (30 mL*2). The combined organic extracts were washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 5:1 petroleum ether/ethyl acetate) to give tert-butyl 2-(4- fluorophenyl)-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (150 mg, 0.47 mmol, 53% yield) as a yellow solid. LC/MS (ESI) m/z: 321.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 6.99 - 6.90 (m, 2H), 6.50 - 6.42 (m, 2H), 3.82 (s, 1H), 3.66 (dd, J=8.0, 11.6 Hz, 1H), 3.56 - 3.42 (m, 2H), 3.37 (d, J=14.8 Hz, 1H), 3.19 - 3.11 (m, 2H), 3.04 (s, 1H), 2.67 - 2.57 (m, 1H), 1.46 (s, 9H), 1.27 (d, J=6.4 Hz, 3H). Step 6: Preparation of (3S,3aS,6aS)-5-(4-fluorophenyl)-3-methyl-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aS,4S,6aR)-2-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (80 mg, 0.25 mmol, 1 eq) and trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 27.05 eq) in dichloromethane (1 mL) was stirred at 20 °C for 0.5 h. The mixture was concentrated to give (3S,3aS,6aS)-5-(4-fluorophenyl)-3-methyl- 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (85 mg, crude, TFA salt) as a brown oil. LC/MS (ESI) m/z: 221.0 [M+H]+. Step 7: Preparation of 4-[(3aS,4S,6aR)-2-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-1,5-naphthyridin-2-one The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,3aS,6aS)-5- (4-fluorophenyl)-3-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole and (6-chloro- 1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate. (yellow solid). LC/MS (ESI) m/z: 413.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.94 (d, J=8.8 Hz, 1H), 7.67 (d, J=8.8 Hz, 1H), 6.99 (t, J=8.8 Hz, 2H), 6.56 - 6.47 (m, 2H), 5.63 (s, 1H), 5.00 (s, 1H), 3.97 (dd, J=8.4, 11.6 Hz, 1H), 3.54 - 3.49 (m, 1H), 3.48 (s, 3H), 3.46 - 3.39 (m, 2H), 3.20 (d, J=7.6 Hz, 2H), 3.10 (dd, J=6.0, 9.6 Hz, 1H), 2.88 - 2.79 (m, 1H), 1.20 (d, J=6.4 Hz, 3H).
Synthesis of Compound 45 Step 1: Preparation of (3aR,6aS)-5-(4-fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole To a solution of tert-butyl (3aS,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (200 mg, 0.94 mmol, 1 eq) and 1-bromo-4-fluoro-benzene (247 mg, 1.41 mmol, 0.15 mL, 1.5 eq) in dioxane (4 mL) were added Cs2CO3 (921 mg, 2.83 mmol, 3 eq) and 1,3- bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (46 mg, 0.047 mmol, 0.05 eq), and the reaction mixture was stirred at 100 °C for 4 hours. The mixture was diluted with water (40 mL) and extracted with ethyl acetate (40 mL*2). The combined organic extracts were washed with brine (100 mL), dried with sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate). To the product was added trifluoroacetic acid (2 mL), and the resulting mixture was stirred at 20°C for 1 h. The mixture was concentrated to give (3aR,6aS)-5-(4- fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (200 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 207.2 [M+H]+. Step 2: Preparation of 6-chloro-4-((3aR,6aS)-5-(4- fluorophenyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile (Compound 45) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-(4- fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (yellow solid). LC/MS (ESI) m/z: 424.1 [M+H]+.1H NMR (400MHz, DMSO-d6) δ 8.00 (d, J = 9.2 Hz, 1H), 7.82 - 7.74 (m, 1H), 7.01 (t, J = 8.4 Hz, 2H), 6.53 (dd, J = 2.4, 4.8 Hz, 2H), 4.50 (dd, J = 6.4, 12.4 Hz, 2H), 4.14 (d, J = 12.4 Hz, 2H), 3.50 - 3.43 (m, 5H), 3.25 (d, J = 10.4 Hz, 2H), 3.15 (s, 2H). Synthesis of Compound 46 Step 1: Preparation of tert-butyl 4-(4-fluoro-N-(4-fluorophenyl)anilino)piperidine-1- carboxylate A mixture of tert-butyl 4-(4-fluoroanilino)piperidine-1-carboxylate (200 mg, 0.68 mmol, 1 eq), 1-fluoro-4-iodo-benzene (151 mg, 0.68 mmol, 0.078 mL, 1 eq), tert-butyl phosphorus (6 mg, 0.03 mmol, 0.007 mL, 4.42e-2 eq), diacetoxypalladium (7 mg, 0.03 mmol, 4.42e-2 eq), and potassium;2-methylpropan-2-olate (229 mg, 2.04 mmol, 3 eq) in toluene (10 mL) was stirred at 100 °C for 6 hours. The reaction mixture was filtered, then concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 10:1 petroleum ether/ethyl acetate) to give compound tert-butyl 4-(4-fluoro-N-(4- fluorophenyl)anilino)piperidine-1-carboxylate (188 mg, 0.48 mmol, 71% yield) as a white solid. LC/MS (ESI) m/z: 333.0 [M-55] +.1H NMR (400 MHz, DMSO-d6) δ 7.11 (t, J = 8.8 Hz, 4H), 6.82 (dd, J = 4.8, 8.8 Hz, 4H), 3.99 - 3.89 (m, 2H), 2.99 - 2.77 (m, 2H), 1.88 (d, J = 12.0 Hz, 2H), 1.33 (s, 9H), 1.10 - 0.99 (m, 2H), 0.89 - 0.77 (m, 1H). Step 2: Preparation of N,N-bis(4-fluorophenyl)piperidin-4-amine To a solution of tert-butyl 4-(4-fluoro-N-(4-fluorophenyl)anilino)piperidine-1-carboxylate (70 mg, 0.18 mmol, 1 eq) in dichloromethane (1 mL) was added dropwise trifluoroacetic acid (1.20 g, 10.50 mmol, 0.78 mL, 58.29 eq), and the resulting mixture was stirred at 25 °C for 30 minutes. The reaction mixture was concentrated under reduced pressure to give compound N,N-bis(4-fluorophenyl)piperidin-4-amine (60 mg, crude, TFA salt) as a green oil. LC/MS (ESI) m/z: 289.1 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-(4-fluoro-N-(4-fluorophenyl)anilino)-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from N,N-bis(4- fluorophenyl)piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 506.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.02 (d, J = 9.2 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 7.13 (t, J = 8.8 Hz, 4H), 6.88 (dd, J = 4.8, 8.8 Hz, 4H), 4.37 - 4.26 (m, 1H), 4.25 - 4.17 (m, 2H), 3.64 - 3.54 (m, 2H), 3.49 (s, 3H), 2.11 - 2.02 (m, 2H), 1.67 - 1.51 (m, 2H). Synthesis of Compound 48
Step 1: Preparation of tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a mixture of 1-bromo-4-(trifluoromethoxy)benzene (50 mg, 0.21 mmol, 0.030 mL, 1 eq), tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate (48 mg, 0.23 mmol, 1.1 eq) and Cs2CO3 (203 mg, 0.62 mmol, 3 eq) in dioxane (1 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (10 mg, 0.01 mmol, 0.05 eq), and the reaction mixture was stirred at 100 °C for 3 hours. The mixture was filtered, and the filtrate was diluted with water (2 mL) and extracted with ethyl acetate (2 mL*2). The combined organic extracts were concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 10:1 petroleum ether/ethyl acetate) to give tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate (20 mg, 0.054 mmol, 26% yield) as a yellow solid. LC/MS (ESI) m/z: 373.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.09 (d, J = 8.4 Hz, 2H), 6.50 - 6.40 (m, 2H), 3.81 - 3.63 (m, 2H), 3.55 - 3.39 (m, 2H), 3.17 - 3.03 (m, 4H), 2.42 - 2.26 (m, 2H), 1.49 (s, 9H).
Step 2: Preparation of (3aS,6aS)-5-[4-(trifluoromethoxy)phenyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole To a solution of tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (110 mg, 0.30 mmol, 1 eq) in dichloromethane (1 mL) was added dropwise trifluoroacetic acid (1.69 g, 14.86 mmol, 1.1 mL, 50.29 eq), and the resulting mixture was stirred at 25 °C for 30 minutes. The reaction mixture was concentrated under reduced pressure to give (3aS,6aS)-5-[4-(trifluoromethoxy)phenyl]- 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (120 mg, crude, TFA) as a yellow oil. LC/MS (ESI) m/z: 272.9 [M+H] +. Step 3: Preparation of 4-[(3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile
The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aS,6aS)-5-[4- (trifluoromethoxy)phenyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (yellow solid). LC/ MS (ESI) m/z: 490.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.05 - 7.99 (m, 1H), 7.81 (d, J = 9.2 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.57 (d, J = 8.8 Hz, 2H), 4.27 - 4.14 (m, 4H), 3.58 - 3.51 (m, 2H), 3.50 (s, 3H), 3.25 - 3.16 (m, 2H), 2.52 (s, 2H). Synthesis of Compound 49
Figure imgf000348_0001
Compound 49
Step 1: Preparation of tert-butyl (3aR,6aS)-2-(4-fluoro-2-methyl-phenyl)-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of 1-bromo-4-fluoro-2-methyl-benzene (300 mg, 1.59 mmol, 1 eq) and tert- butyl (3aR,6aS)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate (343 mg, 1.43 mmol, 0.9 eq) in dioxane (6 mL) was added K2CO3 (658 mg, 4.76 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (77 mg, 0.079 mmol, 0.05 eq), and the reaction mixture was stirred at 100 °C for 4 hr. The mixture was diluted with water (40 mL) and extracted with ethyl acetate (40 mL*2). The combined organic extracts were washed with brine (100 mL), dried with sodium sulfate, and concentrated. The resulting residue was purified by silica gel chromatography (gradient: 100/1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3aR,6aS)-2-(4-fluoro-2-methyl-phenyl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4- c]pyrrole-5-carboxylate (50 mg, crude) as a white solid. LC/MS (ESI) m/z: 349.1 [M+H] +. Step 2: Preparation of (3aS,6aR)-5-(4-fluoro-2-methyl-phenyl)-3a,6a-dimethyl- 2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrole To a solution of tert-butyl (3aR,6aS)-2-(4-fluoro-2-methyl-phenyl)-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (60 mg, 0.17 mmol, 1 eq) in dichloromethane (0.6 mL) was added trifluoroacetic acid (19 mg, 0.17 mmol, 0.013 mL, 1 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under reduced pressure to give (3aS,6aR)-5-(4-fluoro-2-methyl-phenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro- 1H-pyrrolo[3,4-c]pyrrole (65 mg, crude, trifluoroacetic acid salt) as a green solid. LC/MS (ESI) m/z: 249.0 [M+H] +. Step 3: Preparation of 4-[(3aS,6aR)-2-(4-fluoro-2-methyl-phenyl)-3a,6a-dimethyl- 1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 49)
Figure imgf000350_0001
Compound 49 The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aS,6aR)-5-(4- fluoro-2-methyl-phenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrole. (white solid). LC/MS (ESI) m/z: 466.1 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.64 - 7.57 (m, 1H), 7.49 (d, J = 8.8 Hz, 1H), 6.87 - 6.75 (m, 3H), 4.62 (d, J = 12.8 Hz, 2H), 4.24 (d, J = 12.8 Hz, 2H), 3.61 (s, 3H), 3.39 (d, J = 9.2 Hz, 2H), 3.10 (d, J = 9.2 Hz, 2H), 2.23 (s, 3H), 1.24 (s, 6H).
Synthesis of Compound 50 Step 1: Preparation of tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-hydroxy-piperidine-1- carboxylate To a mixture of tert-butyl 7-oxa-3-azabicyclo[4.1.0]heptane-3-carboxylate (3 g, 15.06 mmol, 1 eq) and 4-fluoroaniline (1.67 g, 15.06 mmol, 1.44 mL, 1 eq) in dichloroethane (30 mL) was added scandium(3+); trifluoromethanesulfonate (1.48 g, 3.01 mmol, 0.2 eq), and the reaction mixture stirred at 80 °C for 8 h under the N2. The mixture was partitioned between ethyl acetate (100 mL) and water (100 mL). The layers were separated, and the aqueous phase was further extracted with ethyl acetate (100 mL x 2). The combined organic extract was dried over sodium sulfate and concentrated. The crude product was purified by prep-HPLC (column: Phenomenex luna C18250*50mm*10 µm; mobile phase: [20-50% CH3CN in water (0.225% formic acid)]) to give tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-hydroxy-piperidine- 1-carboxylate (1.7 g, 5.48 mmol, 36% yield) as a yellow solid. LC/MS (ESI) m/z: 255.2 (M- 55)+.1H NMR (400 MHz, CDCl3) δ 6.92 - 6.68 (m, 2H), 6.64 - 6.52 (m, 2H), 4.32 - 4.16 (m, 1H), 4.12 - 3.92 (m, 1H), 3.36 (d, J = 4.0 Hz, 1H), 3.16 - 3.08 (m, 1H), 2.80 - 2.48 (m, 3H), 2.04 - 1.96 (m, 1H), 1.40 (s, 9H), 1.24 - 1.16 (m, 1H). Step 2: Preparation of tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1- carboxylate To a mixture of tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-hydroxy-piperidine-1-carboxylate (400 mg, 1.29 mmol, 1 eq) in tetrahydrofuran (5 mL) at 0°C was added sodium hydride (129 mg, 3.22 mmol, 60% purity, 2.5 eq), and the resulting mixture was stirred at 0 °C for 0.5 h. Iodomethane (549 mg, 3.87 mmol, 0.024 mL, 3 eq) was then added, and the reaction mixture was stirred at 20°C for 1 h under N2. The mixture was diluted with saturated aqueous ammonium chloride (20 mL), then extracted with ethyl acetate (3 × 20 mL). The combined organic extract was washed with brine (3 × 20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by silica gel chromatography (gradient: 30/1 to 3:1 petroleum ether/ethyl acetate) to give tert-butyl (3R,4R)-4-(4- fluoroanilino)-3-methoxy-piperidine-1-carboxylate (390 mg, 1.20 mmol, 93% yield) as a yellow oil. LC/MS (ESI) m/z: 347.0 (M+Na)+. 1H NMR (400 MHz, CDCl3) δ 6.94 - 6.83 (m, 2H), 6.68 - 6.56 (m, 2H), 4.25 - 4.04 (m, 1H), 3.89 - 3.75 (m, 1H), 3.45 (s, 3H), 3.26 (d, J=4.4 Hz, 1H), 3.17 - 2.75 (m, 3H), 2.20 - 2.08 (m, 1H), 1.48 (s, 9H), 1.43 - 1.32 (m, 1H). Step 3: Preparation of (3R,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine A mixture of tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1-carboxylate (150 mg, 0.46 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 29.21 eq) in dichloromethane (1 mL) and stirred at 25°C for 0.5 h. The reaction mixture was concentrated under vacuum to give (3R,4R)-N-(4-fluorophenyl)-3-methoxy-piperidin-4-amine (1.1 g, crude, trifluoroacetic) as a white solid. LC/MS (ESI) m/z: 225.2 [M+H]+. Step 4: Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 50) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,4R)-N-(4- fluorophenyl)-3-methoxy-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 442.2 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.08 - 9.2 (m, 1H), 7.84 - 9.2 (m, 1H), 6.92 - 8.8 (m, 2H), 6.68 - 4.4, 8.8 (m, 2H), 5.6 - 7.6 (m, 1H), 4.52 - 12.0 (m, 1H), 4.04 - 3.88 (m, 1H), 3.76 - 3.56 (m, 2H), 3.52 (s, 3H), 3.4 (s, 3H), 3.24 - 3.16 (m, 1H), 2.24 - 2.16 (m, 1H), 1.56 - 1.44 (m, 1H), 1.35 - 1.12 (m, 1H). Synthesis of Compound 51
Figure imgf000354_0001
Compound 51 Step 1: Preparation of tert-butyl (3R,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy- piperidine-1-carboxylate To a solution of tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-methoxy-piperidine-1-carboxylate (140 mg, 0.43 mmol, 1 eq) and formaldehyde (162 mg, 2.16 mmol, 0.15 mL, 40% purity, 5 eq) in methanol (1 mL) and tetrahydrofuran (1 mL) was added sodium cyanoborohydride (81 mg, 1.29 mmol, 3 eq), and the reaction mixture was stirred at 40 °C for 12 h. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 100/1 to 10:1 petroleum ether/ethyl acetate) to give tert-butyl (3R,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy-piperidine-1-carboxylate (50 mg, 147.75 µmol, 34% yield) as a yellow oil. LC/MS (ESI) m/z: 339.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.00 - 6.87 (m, 2H), 6.81 (dd, J = 4.4, 8.8 Hz, 2H), 4.71 - 4.29 (m, 1H), 4.23 - 3.91 (m, 1H), 3.56 - 3.46 (m, 1H), 3.41 (s, 3H), 3.29 (dt, J = 4.8, 9.6 Hz, 1H), 2.78 (s, 3H), 2.74 - 2.63 (m, 1H), 2.58 - 2.46 (m, 1H), 1.73 - 1.61 (m, 2H), 1.49 (s, 9H). Step 2: Preparation of (3R,4R)-N-(4-fluorophenyl)-3-methoxy-N-methylpiperidin-4- amine To a solution of tert-butyl (3R,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy-piperidine-1- carboxylate (120 mg, 0.35 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (1.85 g, 16.21 mmol, 1.20 mL, 45.71 eq), and the resulting mixture was stirred at 25 °C for 30 min. The reaction mixture was concentrated under reduced pressure to give (3R,4R)- N-(4-fluorophenyl)-3-methoxy-N-methyl-piperidin-4-amine (125 mg, crude, TFA salt) as a yellow oil. LC/MS (ESI) m/z: 239.1 [M+H]+. Step 3: Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoro-N-methyl-anilino)-3-methoxy- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 51)
The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,4R)-N-(4- fluorophenyl)-3-methoxy-N-methyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 456.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 8.8 Hz, 1H), 7.84 (d, J = 8.8 Hz, 1H), 7.04 - 6.95 (m, 2H), 6.89 - 6.82 (m, 2H), 4.66 (d, J = 12.0 Hz, 1H), 4.06 (d, J = 12.0 Hz, 1H), 3.95 - 3.79 (m, 2H), 3.62 (dt, J = 2.8, 12.4 Hz, 1H), 3.54 (s, 3H), 3.38 (s, 3H), 3.07 (dd, J = 9.6, 12.4 Hz, 1H), 2.76 (s, 3H), 1.95 - 1.76 (m, 2H).
Synthesis of Compounds 52 and 53 Step 1: Preparation of tert-butyl 2,5-dimethyl-4-oxo-piperidine-1-carboxylate To a solution of tert-butyl 2-methyl-4-oxo-piperidine-1-carboxylate (2.0 g, 9.38 mmol, 1.00 eq) in tetrahydrofuran (70 mL) at -78 °C was added dropwise lithium diisopropylamide in tetrahydrofuran (2 M, 5.16 mL, 1.10 eq), and the reaction mixture was stirred at -78 °C for 2 h. Iodomethane (1.73 g, 12.19 mmol, 1.30 eq) was then added, and the mixture was stirred at 25 °C for 12 h. The reaction mixture was quenched by addition saturated ammonium chloride solution (100 mL), and the resulting aqueous mixture was extracted with ethyl acetate (100 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 2,5-dimethyl-4- oxo-piperidine-1-carboxylate (2.1 g, crude) as a colorless oil. LC/MS (ESI) m/z: 172.2 [M- C4H9+H] +. Step 2: Preparation of benzyl (2S,5R)-2,5-dimethyl-4-oxo-piperidine-1-carboxylate To a solution of tert-butyl 2,5-dimethyl-4-oxo-piperidine-1-carboxylate (2.1 g, 9.24 mmol, 1.00 eq) in dichloromethane (10 mL) was added hydrogen chloride in dioxane (4 M, 10 mL, 4.33 eq), and the resulting mixture was stirred at 25 °C for 0.5 hours. The mixture was concentrated under reduced pressure. The resulting residue was dissolved in tetrahydrofuran (20 mL) and water (10 mL), then treated with NaHCO3 (3.85 g, 45.83 mmol, 5.00 eq) and benzyl chloroformate (3.13 g, 18.33 mmol, 2.00 eq). The reaction mixture was stirred at 25 °C for 0.5 h. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (100/1 to 30/1 petroleum ether/ethyl acetate) followed by semi- preparative reverse phase HPLC (32-62% acetonitrile + 0.1% formic acid in water, 11 min) to afford benzyl 2,5-dimethyl-4-oxo-piperidine-1-carboxylate (600 mg, 2.30 mmol, 25% yield) as a colorless oil.1H NMR (400 MHz, CDCl3) δ 7.44 - 7.29 (m, 5H), 5.28 - 5.07 (m, 2H), 4.64 -4.60 and 3.46 - 3.31 (m, 1H), 4.43 - 4.22 (m, 1H), 3.86 - 3.65 (m, 1H), 2.88 - 2.60 (m, 1H), 2.57 - 2.36 (m, 1H), 2.32 - 2.22 (m, 1H), 1.26 (dd, J = 5.6, 6.8 Hz, 3H), 1.18 (dd, J = 7.2, 9.6 Hz, 3H). This material was further purified by SFC [method: (column: DAICEL CHIRALPAK AY-H(250mm * 30mm,10um); mobile phase: 15% CH3OH in CO2] to afford benzyl (2S,5R)-2,5-dimethyl-4-oxo-piperidine-1-carboxylate (350 mg, 1.34 mmol, 58% yield) as a yellow oil.1H NMR δ 7.45 - 7.31 (m, 5H), 5.28 - 5.12 (m, 2H), 4.65 (dt, J = 3.1, 6.6 Hz, 1H), 3.90 - 3.78 (m, 1H), 3.75 - 3.65 (m, 1H), 2.84 (dd, J = 6.8, 15.4 Hz, 1H), 2.60 - 2.47 (m, 1H), 2.28 (dd, J = 3.3, 15.4 Hz, 1H), 1.26 (d, J = 6.8 Hz, 3H), 1.18 (d, J = 7.1 Hz, 3H). Step 3: Preparation of benzyl (2S,5R)-4-(4-fluoroanilino)-2,5-dimethyl-piperidine-1- carboxylate To a solution of benzyl (2S,5R)-2,5-dimethyl-4-oxo-piperidine-1-carboxylate (100 mg, 0.38 mmol, 1.00 eq) and 4-fluoroaniline (63 mg, 0.57 mmol, 1.50 eq) in methanol (2 mL) were added sodium cyanoborohydride (48 mg, 0.76 mmol, 2.00 eq) and acetic acid (2 mg, 38.27 umol, 0.10 eq), and the reaction mixture was stirred at 25 °C for 12 h. The reaction mixture was diluted with water 10 mL and extracted with ethyl acetate (10 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by semi-preparative reverse phase HPLC (70-80% acetonitrile + 0.225% formic acid in water, 7 min) to afford benzyl (2S,5R)- 4-(4-fluoroanilino)-2,5-dimethyl-piperidine-1-carboxylate (60 mg, 0.16 mmol, 43% yield) as a colorless oil.1H NMR (400MHz, CDCl3) δ 7.41 - 7.29 (m, 5H), 6.96 - 6.83 (m, 2H), 6.61 - 6.45 (m, 2H), 5.24 - 5.07 (m, 2H), 4.17 (t, J = 6.8 Hz, 1H), 3.78 - 3.61 (m, 1H), 3.40 (dd, J = 4.8, 14.0 Hz, 1H), 3.21 - 3.04 (m, 1H), 2.27 - 2.09 (m, 1H), 1.81 - 1.68 (m, 1H), 1.44 - 1.32 (m, 1H), 1.29 - 1.21 (m, 3H), 1.18 - 0.96 (m, 2H). Step 4: Preparation of benzyl (2S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl- piperidine-1-carboxylate To a solution of benzyl (2S,5R)-4-(4-fluoroanilino)-2,5-dimethyl-piperidine-1-carboxylate (60 mg, 0.16 mmol, 1.00 eq) in methanol (2 mL) were added sodium cyanoborohydride (15 mg, 0.25 mmol, 1.50 eq) and formaldehyde (136 mg, 1.68 mmol, 37% purity in water, 10.00 eq), and the reaction mixture was stirred at 25 °C for 12 h. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give benzyl (2S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-piperidine-1- carboxylate (62 mg, crude) as a white solid. LC/MS (ESI) m/z: 370.9 [M+H] +. Step 5: Preparation of (2S,5R)-N-(4-fluorophenyl)-N,2,5-trimethyl-piperidin-4-amine To a solution of benzyl (2S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-piperidine-1- carboxylate (62 mg, 0.16 mmol, 1.00 eq) in methanol (4 mL) was added palladium on activated carbon catalyst (20 mg, 10% purity) under nitrogen atmosphere. The suspension was degassed and purged with hydrogen (3X), and the reaction mixture was stirred under hydrogen (15 Psi) at 25 °C for 2 h. The reaction mixture was filtered and concentrated under reduced pressure to give (2S,5R)-N-(4-fluorophenyl)-N,2,5-trimethyl-piperidin-4-amine (40 mg, crude) as a white solid. Step 6: Preparation of 8-[(2S,4R,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-1- piperidyl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 52) and 8- [(2S,4S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-1-piperidyl]-5-methyl-6-oxo- 1,5-naphthyridine-2-carbonitrile (Compound 53) To a solution of (2S,5R)-N-(4-fluorophenyl)-N,2,5-trimethyl-piperidin-4-amine (40 mg, 0.16 mmol, 1.00 eq) and (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (62 mg, 0.18 mmol, 1.10 eq) in propan-2-ol (2 mL) was added N,N-diisopropylethylamine (43 mg, 0.33 mmol, 2.00 eq), and the reaction mixture was stirred at 100 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by semi-preparative reverse phase HPLC (48-78% CH3CN in water (0.225% formic acid)) to afford the desired products. 8-[(2S,4R,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-1-piperidyl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (Compound 52) (8.4 mg, 0.02 mol, 11% yield, yellow solid). LC/MS (ESI) m/z: 420.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.86 - 7.79 (m, 1H), 7.77 - 7.70 (m, 1H), 6.98 (t, J = 8.4 Hz, 2H), 6.83 (s, 2H), 6.38 (s, 1H), 3.97 (dd, J = 3.6, 12.0 Hz, 1H), 3.68 (s, 3H), 3.52 - 3.26 (m, 2H), 2.84 (s, 3H), 2.63 - 2.46 (m, 1H), 2.43 - 2.29 (m, 1H), 1.89 (d, J = 6.8 Hz, 2H), 1.20 (d, J = 5.6 Hz, 3H), 0.89 (d, J = 6.4 Hz, 3H). 8-[(2S,4S,5R)-4-(4-fluoro-N-methyl-anilino)-2,5-dimethyl-1-piperidyl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (Compound 53) (5.4 mg, 0.01 mmol, 7% yield, yellow solid). LC/MS (ESI) m/z: 420.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.80 (d, J = 8.8 Hz, 1H), 7.73 - 7.66 (m, 1H), 7.00 (t, J = 8.8 Hz, 2H), 6.81 (dd, J = 4.4, 9.2 Hz, 2H), 6.16 (s, 1H), 5.02 (d, J = 6.0 Hz, 1H), 4.04 - 3.92 (m, 1H), 3.76 - 3.61 (m, 4H), 3.50 (dd, J = 2.4, 12.8 Hz, 1H), 2.87 (s, 3H), 2.57 (m, 1H), 2.42 (d, J = 2.4 Hz, 1H), 1.57 (d, J = 12.4 Hz, 1H), 1.24 (d, J = 6.8 Hz, 3H), 1.12 (d, J = 7.2 Hz, 3H). Synthesis of Compound 54
Step 1: Preparation of (3aR,6aR)-5-benzyl-3-methyl-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aR,6aS)-2-benzyl-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (200 mg, 0.63 mmol, 1 eq) and HCl/dioxane (4 M, 1 mL, 6.33 eq) in dichloromethane (1 mL) was stirred at 20 °C for 1 h. The mixture was concentrated to give (3aR,6aR)-5-benzyl-3-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (200 mg, crude, HCl) as a white solid. LC/MS(ESI) m/z: 217.3 [M+H]+. Step 2: Preparation of (3aR,6aS)-2-benzyl-5-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole To a mixture of (3aR,6aR)-5-benzyl-3-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole (160.00 mg, 0.63 mmol, 1 eq, HCl), 1-bromo-4-fluoro-benzene (122 mg, 0.69 mmol, 0.076 mL, 1.1 eq), and Cs2CO3 (619 mg, 1.90 mmol, 3 eq) in dioxane (3 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (30.79 mg, 31.65 µmol, 0.05 eq), and the reaction mixture was stirred at 100°C for 3 h under N2. The mixture was filtered, and the filtrate was concentrated. The resulting residue was purified by silica gel chromatography (gradient: 40/1 to 5:1 petroleum ether/ethyl acetate) to give (3aR,6aS)-2-benzyl-5-(4-fluorophenyl)-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole (140 mg, 451.02 µmol, 71% yield) as a yellow oil. LC/MS (ESI) m/z: 311.2 [M+H]+. Step 3: Preparation of (3aR,6aS)-5-(4-fluorophenyl)-4-methyl-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole To a solution of (3aR,6aS)-2-benzyl-5-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole (140.00 mg, 0.45 mmol, 1 eq) in ethanol (3 mL) was added palladium on activated carbon catalyst (30 mg, 10% purity) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (50psi) at 50°C for 12 h. The mixture was filtered, and filtrate was concentrated to give (3aR,6aS)-5-(4-fluorophenyl)-4-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (70 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 220.8 [M+H]+. Step 4: Preparation of 4-[(3aR,6aS)-5-(4-fluorophenyl)-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile
The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3aR,6aS)-5-(4- fluorophenyl)-4-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole. (off-white solid). LC/MS (ESI) m/z: 438.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 7.99 (d, J=9.2 Hz, 1H), 7.77 (d, J=9.2 Hz, 1H), 7.24 (s, 1H), 7.11 (s, 1H), 7.03 - 6.98 (m, 2H), 6.53 (dd, J=4.0, 8.8 Hz, 2H), 4.53 - 4.44 (m, 2H), 4.20 (d, J=12.8 Hz, 1H), 3.97 - 3.86 (m, 2H), 3.61 - 3.55 (m, 1H), 3.47 (s, 3H), 3.10 - 3.04 (m, 1H), 2.81 - 2.74 (m, 1H), 2.52 (s, 1H), 1.16 (d, J=6.4 Hz, 3H). Synthesis of Compounds 55 and 56
Step 1: Preparation of tert-butyl (1R,3aS,6aR)-5-(6-chloro-3-cyano-1-methyl-2-oxo- 1,2-dihydro-1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate To a mixture of tert-butyl (3aR,6aS)-4-methyl-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c] pyrrole-5-carboxylate (0.10 g, 0.44 mmol, 1 eq) and 4,6-dichloro-1-methyl-2-oxo -1,5- naphthyridine-3-carbonitrile (112 mg, 0.44 mmol, 1 eq) in acetonitrile (3 mL) was added diisopropylethylamine (171 mg, 1.33 mmol, 3 eq), and the reaction mixture was stirred at 40 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (5:1 to 1:0 petroleum ether: ethyl acetate) to give tert-butyl (1R,3aS,6aR)-5-(6-chloro-3-cyano-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (50 mg, 0.11 mmol, 25% yield) as a yellow oil. LC/MS (ESI) m/z: 388.3 [M-56+H]+.1H NMR (400MHz, CDCl3) δ 7.54 (d, J = 8.8 Hz, 1H), 7.43 (d, J = 8.8 Hz, 1H), 4.57 - 4.45 (m, 2H), 4.16 - 4.07 (m, 2H), 3.87 - 3.78 (m, 1H), 3.68 - 3.58 (m, 1H), 3.51 (s, 3H), 3.27 - 3.19 (m, 1H), 3.06 - 2.95 (m, 1H), 2.60 - 2.50 (m, 1H), 1.39 (s, 9H), 1.26 - 1.21 (m, 3H). Step 2: Preparation of 6-chloro-1-methyl-4-((3aR,4R,6aR)-4- methylhexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile To a mixture of tert-butyl (1R,3aS,6aR)-5-(6-chloro-3-cyano-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (50 mg, 0.11 mmol, 1 eq) in dichloromethane (2 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated under reduced pressure to give 6-chloro-1-methyl-4-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4-c]pyrrol- 2(1H)-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile (50 mg, 0.11 mmol, 96% yield, trifluoroacetate) as a yellow oil. LC/MS (ESI) m/z: 344.2 [M+H] +. Step 3: Preparation of 4-[(3aR,4R,6aR)-5-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile To a mixture of 6-chloro-1-methyl-4-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4-c]pyrrol- 2(1H)-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile (50 mg, 0.11 mmol, 1 eq, trifluoroacetate) and 4-chloro-2-hydroxy-benzaldehyde (34 mg, 0.22 mmol, 2 eq) in 1,2- dichloroethane (3 mL) were added sodium triacetoxyborohydride (69 mg, 0.32 mmol, 3 eq) and triethylamine (33 mg, 0.32 mmol, 3 eq), and the reaction mixture was stirred at 60 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by semi-preparative reverse phase (column: Phenomenex Synergi Polar-RP 100*25mm*4um; mobile phase: [32-52% CH3CN in water (TFA)]) to give 4-[(3aR,4R,6aR)- 5-[(4-chloro-2-hydroxy-phenyl) methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. LC/MS (ESI) m/z: 484.2 [M+H]+. Step 4: Preparation of 4-[(3aR,4R,6aR)-5-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 55) and 4-[(3aS,4S,6aS)-5 -[(4-chloro- 2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2- yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 56) The mixture of enantiomers (40 mg, 0.08 mmol, 1 eq) was separated by supercritical fluid chromatography (Column: DAICEL CHIRALCEL OD(250mm*30mm,10um); mobile phase: [0.1%NH3OH in CH3OH]; B%: 60%-60%, 3.4 min; 40 min). 4-[(3aR,4R,6aR)-5-[(4-chloro-2-hydroxy-phenyl) methyl]-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (14.6 mg, 0.03 mmol, 36% yield) (Compound 55) as an off-white solid. LC/MS (ESI) m/z: 484.1 [M+H]+.1H NMR (400MHz, CDCl3) δ 7.60 - 7.52 (m, 1H), 7.48 - 7.41 (m, 1H), 6.79 (d, J = 8.0 Hz, 1H), 6.75 - 6.70 (m, 1H), 6.69 - 6.63 (m, 1H), 4.60 (d, J = 12.4 Hz, 1H), 4.40 (dd, J = 2.0, 12.4 Hz, 1H), 4.10 (d, J = 13.6 Hz, 1H), 4.01 - 3.89 (m, 2H), 3.54 (s, 3H), 3.35 - 3.25 (m, 2H), 2.96 - 2.85 (m, 1H), 2.61 - 2.48 (m, 2H), 2.30 - 2.19 (m, 1H), 1.27 (d, J = 5.6 Hz, 3H). The second peak was purified by semi-preparative reverse phase (column: Unisil 3-100 C18 Ultra 150*50mm*3 um; mobile phase: [5-35% CH3CN in water (0.225% formic acid)]) to give 4-[(3aS,4S,6aS)-5-[(4-chloro-2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a - hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (9.6 mg, 0.02 mmol, 23% yield) (Compound 56) as a white solid. LC/MS (ESI) m/z: 484.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.69 - 7.62 (m, 1H), 7.57 - 7.51 (m, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.84 - 6.80 (m, 1H), 6.79 - 6.74 (m, 1H), 4.75 - 4.59 (m, 1H), 4.55 - 4.41 (m, 1H), 4.19 (d, J = 13.6 Hz, 1H), 4.11 - 3.99 (m, 2H), 3.68 - 3.61 (m, 3H), 3.44 - 3.33 (m, 2H), 3.06 - 2.95 (m, 1H), 2.69 - 2.58 (m, 2H), 2.37 - 2.28 (m, 1H), 1.37 (d, J = 5.6 Hz, 3H). Synthesis of Compounds 57 and 58
Step 1: Preparation of tert-butyl (1S,3aR,6aS)-5-(4-chloro-2-hydroxybenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate To a mixture of tert-butyl (1R,3aS,6aR)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate (0.50 g, 2.21 mmol, 1 eq) and 4-chloro-2-hydroxy-benzaldehyde (371 mg, 2.37 mmol, 1.07 eq) in 1,2-dichloroethane (10 mL) were added sodium triacetoxyborohydride (1.40 g, 6.63 mmol, 3 eq) and triethylamine (447 mg, 4.42 mmol, 2 eq), and the reaction mixture was stirred at 60 °C for 12 h. The mixture was diluted with water (5 mL), then extracted with ethyl acetate (20 mL x 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate = 5:1 to 0:1) to give tert-butyl (1S,3aR,6aS)-5-(4-chloro-2-hydroxybenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (0.70 g, 1.91 mmol, 86% yield) as a yellow solid. LC/MS (ESI) m/z: 367.3 [M+H]+. Step 2: Preparation of 5-chloro-2-(((3aS,4S,6aS)-4-methylhexahydropyrrolo[3,4- c]pyrrol-2(1H)-yl)methyl)phenol To a mixture of tert-butyl (1S,3aR,6aS)-5-(4-chloro-2-hydroxybenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (0.30 g, 0.81 mmol, 1 eq) in dichloromethane (6 mL) was added trifluoroacetic acid (3 mL), and the reaction mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated under reduced pressure to give 5- chloro-2-(((3aS,4S,6aS)-4-methylhexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)methyl)phenol (0.30 g, 0.78 mmol, 96% yield, TFA salt) as a yellow oil. Step 3: Preparation of 4-[(3aS,4S,6aR)-2-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile To a mixture of 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (0.10 g, 0.39 mmol, 1 eq) and 5-chloro-2-(((3aS,4S,6aS)-4-methylhexahydropyrrolo[3,4-c]pyrrol-2(1H)- yl)methyl)phenol (0.30 g, 0.78 mmol, 2.00 eq, trifluoroacetate) in tetrahydrofuran (5 mL) was added diisopropylethylamine (152 mg, 1.18 mmol, 3 eq), and the reaction mixture was stirred at 60 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by semi-preparative reverse phase (column: Phenomenex Synergi Polar- RP 100*25mm*4 µm; mobile phase: [32-52% CH3CN in water (TFA)]) to give 4- [(3aS,4S,6aR)-2-[(4-chloro-2-hydroxy-phenyl) methyl]-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (0.15 g, 0.30 mmol, 78% yield) as an off-white solid. LC/MS (ESI) m/z: 484.2 [M+H]+.
Step 4: Preparation of 4-[(3aS,4S,6aR)-2-[(4-chloro-2-hydroxy-phenyl) methyl]-4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 57) and 4-[(3aR,4R,6aS)-2 -[(4-chloro- 2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5- yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 58) The mixture of enantiomers (0.15 g, 0.30 mmol, 1 eq) was separated by supercritical fluid chromatography (column: DAICEL CHIRALPAK AD (250mm*30mm,10µm); mobile phase: [0.1%NH4OH in isopropanol]; B%: 40%-40%, 3.2 min; 60 min). Absolute configuration was arbitrarily assigned. The first peak was purified further by semi-preparative reverse phase (column: Waters Xbridge 150*25mm* 5um; mobile phase: [45-75% CH3CN in water (NH4OH)]) to give 4- [(3aS,4S,6aR)-2-[(4-chloro-2-hydroxy-phenyl) methyl]-4-methyl-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (22.6 mg, 0.04 mmol, 14% yield) (Compound 57) as a yellow solid. LC/MS (ESI) m/z: 484.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.85 - 7.78 (m, 1H), 7.02 (d, J = 8.0 Hz, 1H), 6.64 (dd, J = 2.0, 8.0 Hz, 1H), 6.60 - 6.55 (m, 1H), 6.00 - 5.86 (m, 1H), 4.44 - 4.34 (m, 1H), 3.79 (br d, J = 12.4 Hz, 1H), 3.65 - 3.47 (m, 7H), 3.11 - 3.00 (m, 1H), 2.78 - 2.59 (m, 4H), 1.21 (d, J = 6.4 Hz, 3H). The second peak was purified further by semi-preparative reverse phase (column: Phenomenex C1875*30mm*3µm; mobile phase: [12-42% CH3CN in water (formic acid)]) to give 4-[(3aR,4R,6aS)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-4-methyl-1,3,3a,4,6,6a - hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (30.9 mg, 0.06 mmol, 77% yield) (Compound 58) as an off-white solid. LC/MS (ESI) m/z: 484.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.07 (br d, J = 8.0 Hz, 1H), 6.77 - 6.56 (m, 2H), 5.97 - 5.81 (m, 1H), 4.43 - 4.30 (m, 1H), 3.86 - 3.54 (m, 4H), 3.52 (s, 3H), 3.13 - 3.03 (m, 1H), 2.94 - 2.70 (m, 3H), 2.69 - 2.57 (m, 2H), 1.20 (d, J = 6.8 Hz, 3H). Synthesis of Compound 61 Preparation of 8-[(3aR,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (Compound 61) A solution of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-ylmethyl)-5-chloro- phenol (65 mg, 0.22 mmol, 1 eq, hydrochloride), triethylamine (113 mg, 1.12 mmol, 0.15 mL, 5.0 eq), and (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (74 mg, 0.22 mmol, 1 eq) in dichloromethane (5 mL) and DMF (1 mL) was stirred at 20 °C for 15 h. The reaction mixture was concentrated under reduced pressure. The product was purified by prep-HPLC (column: Waters Xbridge 150*25 mm* 5 µm; mobile phase: [35-65% CH3CN in water (NH4HCO3)]) followed by lyophilization to afford 8-[(3aR,6aR)-2-[(4- chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (36.1 mg, 82.82 µmol, 37% yield) as a white solid. LC/MS (ESI) m/z: 436.1 [M+H] +.1H NMR (400 MHz, DMSO-d6) δ 8.17 - 8.10 (m, 1H), 8.05 - 7.98 (m, 1H), 7.16 - 7.08 (m, 1H), 6.82 - 6.73 (m, 2H), 5.65 - 5.59 (m, 1H), 3.98 - 3.93 (m, 2H), 3.50 (br s, 5H), 2.97 - 2.90 (m, 2H), 2.71 - 2.64 (m, 4H), 2.40 - 2.35 (m, 2H). Synthesis of Compound 62 Step 1: Preparation of 1-benzyl-4-(2-bromoanilino)piperidine-4-carbonitrile To a stirred solution of 1-benzylpiperidin-4-one (10.0 g, 52.8 mmol, 9.8 mL, 1 eq) and 2- bromoaniline (9.10 g, 52.8 mmol, 1 eq) in AcOH (40 mL) was added TMSCN (5.5 g, 56.0 mmol, 7.0 mL, 1.06 eq), and the reaction mixture was stirred at 25°C for 16 hours. The reaction mixture was neutralized by addition of 8 N aqueous NaOH (120 mL) at 0 °C, and the resulting mixture was extracted with ethyl acetate (100 mL ×3). The organic extract was dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The crude product was purified by flash silica gel chromatography (gradient: of 0~10% ethyl acetate in petroleum ether) to give 1-benzyl-4-(2-bromoanilino) piperidine-4-carbonitrile (10.0 g, 27.0 mmol, 51% yield) as a white solid. LC/MS (ESI) m/z: 370.2 [M+H]+. Step 2: Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]-3-imine To a stirred solution of 1-benzyl-4-(2-bromoanilino)piperidine-4-carbonitrile (7.4 g, 19.98 mmol, 1 eq) in toluene (200 mL) were added AIBN (328 mg, 2.00 mmol, 0.1 eq) and Bu3SnH (6.4 g, 21.98 mmol, 5.8 mL, 1.1 eq), and the reaction mixture was stirred at 110 °C under N2 for 16 hours. After cooling to 25°C, the reaction mixture was quenched by addition of saturated KF (200 mL) and extracted with ethyl acetate (200 mL ×2). The combined organic extract was dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give 1'-benzylspiro[indoline-2,4'-piperidine]-3-imine (7.7 g, crude) as a yellow gum. LC/MS (ESI) m/z: 292.3 [M+H]+. Step 3: Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]-3-one A mixture of 1'-benzylspiro[indoline-2,4'-piperidine]-3-imine (7.70 g, 26.4 mmol, 1 eq) in aqueous HCl (1 M, 232 mL, 8.8 eq) was stirred at 100 °C for 1 hour. After cooling to 25°C, the mixture was basified with saturated aqueous NaHCO3 to pH = 8 and then extracted with CH2Cl2 (200 mL ×3). The combined organic layer was washed with brine (100 mL ×2), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The crude product was purified by flash silica gel chromatography (gradient: 0~1% CH3OH in CH2Cl2) to give 1'-benzylspiro[indoline-2,4'-piperidine]-3-one (3.00 g, 10.3 mmol, 38.83% yield) as a yellow solid. LC/MS (ESI) m/z: 293.3 [M+H]+. Step 4: Preparation of 1'-benzylspiro[indoline-2,4'-piperidine]-3-ol To a solution of 1'-benzylspiro[indoline-2,4'-piperidine]-3-one (500 mg, 1.71 mmol, 1 eq) in THF (5 mL) was added LiAlH4 (1 M, 1.8 mL, 1.1 eq) at 0°C, and the reaction mixture was stirred at 25°C for 1 hour. The reaction quenched by sequential addition of water (0.3 mL), 15% aq. NaOH (0.3 mL), and water (0.6 mL). The resulting suspension was dried over anhydrous Na2SO4 and then filtered. The filtrate was concentrated in vacuo to give 1'- benzylspiro[indoline-2,4'-piperidine]-3-ol (500 mg, crude) as a white solid. LC/MS (ESI) m/z: 295.3 [M+H]+. Step 5: Preparation of 1'-benzylspiro[indoline-2,4'-piperidine] A solution of 1'-benzylspiro[indoline-2,4'-piperidine]-3-ol (500 mg, 1.70 mmol, 1 eq) and Et3SiH (987 mg, 8.49 mmol, 1.3 mL, 5 eq) in CH2Cl2 (10 mL) was treated with TFA (1.9 g, 16.9 mmol, 1.3 mL, 10 eq) at 25°C for 1 hour. The mixture was basified with sat. aq. NaHCO3 to pH = 8, and the suspension was extracted with CH2Cl2 (20 mL ×3). The combined organic extract was washed with brine (15 mL ×3), dried over Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography (gradient: 0~50 % ethyl acetate in petroleum ether) to give 1'- benzylspiro[indoline-2,4'-piperidine] (370 mg, 1.33 mmol, 86.95% yield) as a white solid. LC/MS (ESI) m/z: 279.3 [M+H]+. Step 6: Preparation of 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine] To a mixture of 1'-benzylspiro[indoline-2,4'-piperidine] (450 mg, 1.62 mmol, 1 eq) and formaldehyde (655 mg, 8.08 mmol, 601 µL, 37% purity, 5 eq) in CH3OH (10 mL) were added AcOH (970 mg, 16.1 mmol, 924 µL, 10 eq) and borane 2-methylpyridine (864 mg, 8.08 mmol, 5 eq), and the reaction mixture was stirred at 25 °C for 10 hours. The mixture was basified with triethylamine to pH = 8, and then concentrated. The crude product was purified by flash silica gel chromatography (gradient: 0~50 % ethyl acetate in petroleum ether) to give 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine] (300 mg, 1.03 mmol, 63% yield) as a yellow oil. LC/MS (ESI) m/z: 293.1 [M+H]+. Step 7: Preparation of spiro[indoline-2,4'-piperidine] To a solution of 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine] (150 mg, 0.513 mmol, 1 eq) in CH3OH (5 mL) were added Pd(OH)2 (62 mg, 0.442 mmol, 20% purity, 0.8 eq) and AcOH (61 mg, 1.03 mmol, 58 µL, 2 eq) under N2, and the resulting mixture was degassed and purged with H2 (3X), then stirred at 50 °C for 2 hours under H2 (50 Psi). The reaction mixture was filtered, the filter cake was washed with CH3OH (200 mL), and the filtrate concentrated under reduced pressure to give 1-methylspiro[indoline-2,4'-piperidine] (100 mg, crude) as a white solid. LC/MS (ESI) m/z: 203.1 [M+H]+. Step 8: Preparation of 6-chloro-1-methyl-4-(1-methylspiro[indoline-2,4'-piperidine]- 1'-yl)-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 62) To a solution of 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (87 mg, 0.346 mol, 1 eq) in CH3CN (3 mL) were added 1-methylspiro[indoline-2,4'-piperidine] (70 mg, 0.346 mmol, 1 eq) and diisopropylethylamine (223 mg, 1.73 mmol, 305 µL, 5 eq), and the reaction mixture was stirred at 25°C for 1 hour. The reaction mixture was concentrated, and the crude product was purified by prep-HPLC (column: DAICEL CHIRALPAK AD (250 mm * 30 mm, 10 µm); mobile phase: [0.1% NH4OH in EtOH]; B%: 45%-45%, 5 min). Pure fraction was concentrated and lyophilized to give 6-chloro-1-methyl-4-(1- methylspiro[indoline-2,4'-piperidine]-1'-yl)-2-oxo-1,5-naphthyridine-3-carbonitrile (21.6 mg, 0.0506 mmol, 15% yield) as a yellow solid. LC/MS (ESI) m/z: 419.8 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.82 (d, J = 8.8 Hz, 1H), 7.10-6.94 (m, 2H), 6.55 (t, J = 7.2 Hz, 1H), 6.37 (d, J = 7.6 Hz, 1H), 4.26 ( d, J = 12.8 Hz, 2H), 3.54 (s, 3H), 3.49 (s, 2H), 3.07 (s, 2H), 2.63 (s, 3H), 2.18-2.15 (m, 2H), 1.60 (d, J = 12.8 Hz, 2H). Synthesis of Compound 63 Preparation of 6-chloro-1-methyl-2-oxo-4-spiro[3H-benzofuran-2,4'-piperidine]-1'- yl-1,5-naphthyridine-3-carbonitrile (Compound 63) A mixture of spiro[3H-benzofuran-2,4'-piperidine] (250 mg, 1.32 mmol, 1 eq) ,4,6-dichloro- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (336 mg, 1.32 mmol, 1 eq), and diisopropylethylamine (371.00 mg, 2.87 mmol, 0.5 mL, 2.17 eq) in acetonitrile (3 mL) was stirred at 25°C for 12 h. The mixture was concentrated under vacuum, and the crude product was triturated in dimethylformamide (1mL), washed with acetonitrile, and collected by filtration to afford 6-chloro-1-methyl-2-oxo-4-spiro[3H-benzofuran-2,4'-piperidine]-1'-yl-1,5- naphthyridine-3-carbonitrile (475 mg, 1.15 mmol, 87% yield) as a white solid. LC/MS (ESI) m/z: 407.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.82 (d, J = 9.2 Hz, 1H), 7.2 (d, J = 7.2 Hz, 1H), 7.12 - 7.08 (m, 1H), 6.88 - 6.8 (m, 2H), 4.00 - 3.84 (m, 4H), 3.52 (s, 3H), 3.12 (s, 2H), 2.08 (t, J = 5.6 Hz, 4H). Synthesis of Compound 64 Step 1: Preparation of tert-butyl 2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (2.0 g, 5.16 mmol, 1.0 eq) and potassium carbonate (7.13 g, 51.62 mmol, 10.0 eq) in acetonitrile (50 mL) was added 4-fluoroaniline (1.72 g, 15.49 mmol, 1.48 mL, 3.0 eq), and the reaction mixture was stirred at 90 °C for 15 h. The reaction mixture was cooled to room temperature and partitioned between ethyl acetate (200 mL) and water (30 mL). The organic layer was washed with brine (30 mL), dried over sodium sulfate, filtered, and concentrated. The resulting residue was purified by silica gel chromatography (gradient: 10/1 to 2/1 petroleum ether/ethyl acetate) followed by prep-TLC (SiO2, petroleum ether/ethyl acetate=2/1) to afford tert-butyl 2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.3 g, 979.19 umol, 19% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 6.98 - 6.92 (m, 2H), 6.45 - 6.38 (m, 2H), 3.78 - 3.66 (m, 2H), 3.46 - 3.40 (m, 2H), 3.15 - 3.03 (m, 4H), 2.41 - 2.24 (m, 2H), 1.49 (s, 9H). Step 2: Preparation of 5-(4-fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole To a solution of tert-butyl 2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole- 5-carboxylate (0.3 g, 0.98 mmol, 1 eq) in methanol (3 mL) and dichloromethane (5 mL) was added hydrochloric acid/dioxane (4 M, 10 mL), and the reaction mixture was stirred at 20 °C for 0.5 h. The reaction mixture was concentrated to afford 5-(4-fluorophenyl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole (0.23 g, 947.60 umol, 97% yield, HCl salt) as a yellow solid. LC/MS (ESI) m/z: 207.5 [M+H] +. Step 3: Preparation of 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 64)
To a solution of 5-(4-fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (71 mg, 0.29 mmol, 1.5 eq, hydrochloride) and N,N-diisopropylethylamine (152 mg, 1.18 mmol, 0.2 mL, 6.00 eq) in dichloromethane (10 mL) and DMF (1 mL) was added 4,6-dichloro-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (50 mg, 0.2 mmol, 1.0 eq), and the reaction mixture was stirred at 20 °C for 15 h. Water (20 mL) and ethyl acetate (20 mL) were added, and the resulting mixture was stirred at 20 °C for 10 minutes. The precipitate was filtered, and the product was washed with water (5 mL) and ethyl acetate (5 mL x 2), then concentrated at 40 °C to afford 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (48.9 mg, 115.37 umol, 59% yield) as a yellow solid. LC/MS (ESI) m/z: 424.1 [M+H] +.1H NMR (400 MHz, DMSO-d6) δ 8.07 - 7.97 (m, 1H), 7.85 - 7.75 (m, 1H), 7.08 - 6.93 (m, 2H), 6.55 - 6.44 (m, 2H), 4.28 - 4.15 (m, 4H), 3.54 - 3.45 (m, 5H), 3.31 - 3.28 (m, 2H), 3.21 - 3.15 (m, 2H). Synthesis of Compounds 65, 66, 67, and 60 Step 1: Preparation of 5-chloro-2-[(3-methyl-4-piperidyl)amino]phenol To a solution of tert-butyl 4-(4-chloro-2-hydroxy-anilino)-3-methyl-piperidine-1-carboxylate (100 mg, 0.29 mmol, 1.00 eq) in dichloromethane (5 mL) was added trifluoroacetic acid (33 mg, 0.29 mmol, 1.00 eq), and the reaction mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give 5-chloro-2-[(3-methyl-4- piperidyl)amino]phenol (100 mg, 0.28 mmol, 96% yield, trifluoroacetic acid salt) as a brown oil. Step 2: Preparation of 6-chloro-4-[4-(4-chloro-2-hydroxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a solution of 5-chloro-2-[(3-methyl-4-piperidyl)amino]phenol (100 mg, 0.28 mmol, 1.00 eq, trifluoroacetate) in acetonitrile (5 mL) were added 4,6-dichloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (71 mg, 0.28 mmol, 1.00 eq) and N,N-diisopropylethylamine (36 mg, 0.28 mmol, 1.00 eq), and the reaction mixture was stirred at 60 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150*50 mm*3 µm; mobile phase: [45-75% CH3CN in water (0.225% formic acid)]) to afford 6-chloro-4-[4-(4-chloro-2-hydroxy- anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (100 mg, 0.21 mmol, 77% yield) as a yellow solid. LC/MS (ESI) m/z: 458.1[M+H]+.
Step 3: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 65), 6- chloro-4-[(3R,4S)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (Compound 66), 6-chloro-4-[(3R,4R)-4-(4- chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile (Compound 67), and 6-chloro-4-[(3S,4S)-4-(4-chloro-2-hydroxy- anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 60) The isomers (100 mg, 0.21 mmol) were separated by SFC. First SFC method (column: DAICEL CHIRALPAK AD (250mm*30 mm, 10 µm); mobile phase: 60% ethanol (0.1% NH4OH) in CO2) to give a mixture of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-anilino)-3- methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6-chloro-4- [(3R,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile, and mixture of 6-chloro-4-[(3R,4S)-4-(4-chloro-2-hydroxy- anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6- chloro-4-[(3S,4S)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile. Further purification of each mixture as shown below afforded the individual isomers. SFC method (column: Daicel ChiralPak IG (250*30 mm, 10 um); mobile phase: 45% ethanol (0.1% NH4OH) in CO2) gave 6-chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (4.4 mg, 0.009 mmol, 14% yield) as a yellow solid, and 6-chloro-4-[(3R,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (5.1 mg, 0.01 mmol, 16% yield) as a yellow solid. SFC method (column: DAICEL CHIRALPAK IC (250mm*30 mm,10 um); mobile phase: 50% methanol (0.1% NH4OH) in CO2) gave 6-chloro-4-[(3R,4S)-4-(4-chloro-2-hydroxy- anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (7.4 mg, 0.01 mmol, 23% yield) as a yellow solid, and 6-chloro-4-[(3S,4S)-4-(4-chloro-2-hydroxy- anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (9.3 mg, 0.02 mmol, 29% yield) as a yellow solid. Absolute configuration was arbitrarily assigned. 6-Chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 65). LC/MS (ESI) m/z: 458.1 [M+H] +. 1HNMR (400 MHz, CD3OD) δ 8.01 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 6.72-6.66 (m, 3H), 4.18 - 3.93 (m, 1H), 3.85-3.82 (m, 2H), 3.80 - 3.73 (m, 1H), 3.72 - 3.70 (m, 1H), 3.63 (s, 3H), 2.46 - 2.45 (m, 1H), 2.25 - 2.22 (m, 1H), 2.05 - 2.01 (m, 1H), 0.96 (d, J = 6.8 Hz, 3H). 6-Chloro-4-[(3R,4S)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 66). LC/MS (ESI) m/z: 458.1 [M+H] +. 1HNMR (400 MHz, CD3OD) δ 8.02 (d, J = 8.8 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 6.72 - 6.66 (m, 3H), 4.18 - 4.17(m, 1H), 3.96 - 3.93(m, 2H), 3.84 - 3.82(m, 1H), 3.81 - 3.73(m, 1H), 3.63 (s, 3H), 2.45 - 2.43(m, 1H), 2.24 - 2.21(m, 1H), 2.01 - 2.00 (m, 1H),0.97 (d, J = 7.2 Hz, 3H) 6-Chloro-4-[(3R,4R)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 67). LC/MS (ESI) m/z: 458.1 [M+H] +. 1HNMR (400 MHz, DMSO-d6) δ 8.01 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 6.70 - 6.63 (m, 3H), 4.44 - 4.36 (m, 2H), 3.67 - 3.61 (m, 4H), 3.31 - 3.19 (m, 1H), 2.29 - 2.15 (m, 1H), 2.14 -2.11 (m, 1H), 1.66 -1.63 (m, 1H), 1.32 - 1.29 (m, 1H), 1.10 (d, J = 6.4 Hz, 3H). 6-Chloro-4-[(3S,4S)-4-(4-chloro-2-hydroxy-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 60). LC/MS (ESI) m/z: 458.0 [M+H] +. 1HNMR (400 MHz, DMSO-d6) δ 8.01 (d, J = 8.8 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 6.74 - 6.58 (m, 3H), 4.48 - 4.33 (m, 2H), 3.69 - 3.61 (m, 4H), 3.27 - 3.19 (m, 1H), 2.66 (s, 1H), 2.33 - 2.23 (m, 1H), 2.20 - 2.07 (m, 1H), 1.73 - 1.56 (m, 1H), 1.10 (d, J = 6.4 Hz, 3H). Synthesis of Compound 68
Figure imgf000385_0001
Compound 68 Step 1: Preparation of spiro[indoline-2,4'-piperidine]-3-one To a solution of 1'-benzylspiro[indoline-2,4'-piperidine]-3-one (150 mg, 0.513 mmol, 1 eq) in CH2Cl2 (3 mL) was added 1-chloroethyl carbonochloridate (733 mg, 5.13 mmol, 10 eq), and the reaction mixture was stirred at 45 °C for 10 hours. The reaction mixture was concentrated to give a residue. A mixture of the residue in CH3OH (5 mL) was stirred at 60 °C for 1 hour. The mixture was concentrated to give spiro[indoline-2,4'-piperidine]-3-one (100 mg, crude) as a yellow solid. LC/MS (ESI) m/z: 203.3 [M+H]+.
Step 2: Preparation of 6-chloro-1-methyl-2-oxo-4-(3-oxospiro[indoline-2,4'- piperidine]-1'-yl)-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile starting from spiro[indoline-2,4'-piperidine]-3-one. LC/MS (ESI) m/z: 419.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 8.8 Hz, 2H), 7.83 (d, J = 9.2 Hz, 1H), 7.57-7.38 (m, 2H), 6.92 (d, J = 8.4 Hz, 1H), 6.74 (t, J = 7.2 Hz, 1H), 4.27 (d, J = 13.6 Hz, 2H), 3.79 (t, J = 11.6 Hz, 2H), 3.55 (s, 3H), 2.10-2.09 (m, 2H), 1.51-1.50 (m, 2H).
Synthesis of Compounds 69 and 43
Step 1: Preparation of tert-butyl (3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate and tert-butyl (3aS,6aS)-2-(4- fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate The enantiomers were separated by SFC (column: DAICEL CHIRALCEL OJ(250mm*30mm,10um); mobile phase: 10% CH3OH (0.1% NH4OH) in CO2). Absolute configuration was arbitrarily assigned. tert-butyl (3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate (43 mg, 0.14 mmol, 43% yield, white solid) (Rt = 1.14 min). tert-butyl (3aS,6aS)-2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate (42 mg, 0.14 mmol, 42% yield, white solid) (Rt = 1.23 min).
Step 2: Preparation of 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 69) and 4-[(3aS,6aS)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 43) The title compounds were prepared in an analogous manner to 4-[(3aR,6aR)-2-(4- fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile. 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (yellow solid) (Compound 69). LC/MS (ESI) m/z: 424.0 [M+H] +.1H NMR (400MHz, DMSO-d6) δ 8.02 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.01 (t, J = 8.8 Hz, 2H), 6.60 - 6.43 (m, 2H), 4.28 - 4.17 (m, 4H), 3.52 - 3.46 (m, 5H), 3.39 (dd, J = 2.4, 3.6 Hz, 2H), 3.18 (t, J = 9.2 Hz, 2H). 4-[(3aS,6aS)-2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (yellow solid) (Compound 43). LC/MS (ESI) m/z: 424.0 [M+H] +.1H NMR (400MHz, DMSO-d6) δ 8.08 - 7.98 (m, 1H), 7.85 - 7.74 (m, 1H), 7.08 - 6.94 (m, 2H), 6.66 - 6.38 (m, 2H), 4.31 - 4.16 (m, 4H), 3.54 - 3.45 (m, 5H), 3.37 (s, 2H), 3.18 (t, J = 8.8 Hz, 2H). MS (ESI) m/z: 424.0 [M+1] + 1H NMR: (400MHz, DMSO-d6) δ = 8.02 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.01 (t, J = 8.8 Hz, 2H), 6.60 - 6.43 (m, 2H), 4.28 - 4.17 (m, 4H), 3.52 - 3.46 (m, 5H), 3.39 (dd, J = 2.4, 3.6 Hz, 2H), 3.18 (t, J = 9.2 Hz, 2H) Synthesis of Compound 71
Figure imgf000390_0001
Step 1: Preparation of ethyl 3-amino-6-chloro-pyridine-2- carboxylate To a solution of 3-amino-6-chloro-pyridine-2-carboxylic acid (10.0 g, 58.0 mmol, 1 eq) in EtOH (100 mL) was added SOCl2 (8.96 g, 75.3 mmol, 1.3 eq), and the reaction mixture was stirred at 85 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give ethyl 3-amino-6-chloro-pyridine-2-carboxylate (13.5 g, 56.9 mmol, 98% yield, HCl) as a yellow solid. LC/MS (ESI) m/z: 201.2 [M+H]+. Step 2: Preparation of ethyl 6-chloro-3-[(2-cyanoacetyl)amino] pyridine-2- carboxylate To a solution of ethyl 3-amino-6-chloro-pyridine-2-carboxylate (8.00 g, 33.7 mmol, 1 eq, HCl) in DMF (50 mL) were added triethylamine (17.1 g, 169 mmol, 5 eq), 2-cyanoacetic acid (8.61 g, 101 mmol, 3 eq) and 1-propanephosphonic anhydride (34.4 g, 54.0 mmol, 50% purity, 1.6 eq), and the reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was diluted with water (80 mL) and extracted with EtOAc (80 mL ×3). The combined organic extract was washed with brine (80 mL ×3), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give ethyl 6-chloro-3-[(2- cyanoacetyl)amino]pyridine-2-carboxylate (8.71 g, 24.1 mmol, 71% yield) as a yellow solid. LC/MS (ESI) m/z: 268.1 [M+H]+. Step 3: Preparation of 6-chloro-4-hydroxy-2-oxo-1H-1,5 –naphthyridine-3- carbonitrile To a solution of ethyl 6-chloro-3-[(2-cyanoacetyl)amino]pyridine-2-carboxylate (8.71 g, 32.5 mmol, 1 eq) in THF (90 mL) was added potassium bis(trimethylsilyl)amide (1 M, 39.05 mL, 1.2 eq) at -78 °C, and the reaction mixture was stirred at -78°C under N2 for 1 hour. The reaction mixture was quenched by addition of 1M aqueous HCl to adjust pH=3, then concentrated under reduced pressure. The resulting residue was suspended in water (150 mL), and the suspension was cooled in an ice bath, then filtered. The solid was dried in vacuo to give 6-chloro-4-hydroxy-2-oxo-1H-1,5-naphthyridine-3-carbonitrile (5.33 g, 20.9 mmol, 64% yield) as a yellow solid. LC/MS (ESI) m/z: 221.9 [M+H]+. Step 4: Preparation of 6-chloro-4-hydroxy-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile To a solution of 6-chloro-4-hydroxy-2-oxo-1H-1,5-naphthyridine-3-carbonitrile (5.33 g, 20.9 mmol, 87% purity, 1 eq) in DMF (80 mL) was added NaH (4.81 g, 120 mmol, 60% purity, 5.74 eq) at 0 °C, and the reaction mixture was stirred at 0 °C under N2 for 0.5 hour. Iodomethane (10.2 g, 72.2 mmol, 3.45 eq) was then added, and the reaction mixture was stirred at 25 °C under N2 for 16 hours. The mixture was slowly poured into water (100 mL), and the pH of the resulting mixture was adjusted to pH=3 with 1N aqueous HCl. The resulting suspension was filtered, and the filter cake was collected and dried under vacuum to give 6-chloro-4-hydroxy-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (5.14 g, 21.8 mmol, 91% yield) as a yellow solid. LC/MS (ESI) m/z: 235.9 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 8.8 Hz, 1H), 7.87 (d, J = 9.2 Hz, 1H), 3.54 (s, 3H). Step 5: Preparation of 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a solution of 6-chloro-4-hydroxy-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (5.14 g, 21.8 mmol, 1 eq) and N,N-diisopropylethylamine (16.9 g, 131 mmol, 6 eq) in CH3CN (100 mL) were added benzyl(triethyl)ammonium chloride (9.94 g, 43.6 mmol, 2 eq) and POCl3 (16.7 g, 109 mmol, 5 eq) at 0 °C, and the reaction mixture was stirred at 25 °C under N2 for 16 hours. The mixture was concentrated under reduced pressure. The resulting residue was poured into ice and saturated NaHCO3 solution (30 mL), and the mixture was extracted with CH2Cl2 (30 mL ×3). The combined organic extracts were washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by flash silica gel chromatography (gradient: 0~3% THF/CH2Cl2) to give 4,6-dichloro-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (3.03 g, 11.9 mmol, 55% yield) as a yellow solid. LC/MS (ESI) m/z: 253.8 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J = 9.2 Hz, 1H), 8.00 (d, J = 8.8 Hz, 1H), 3.65 (s, 3H). Step 6: Preparation of 6-chloro-1-methyl-2-oxo-4-spiro[indane-2,4'-piperidine]-1'-yl- 1,5-naphthyridine-3-carbonitrile A mixture of spiro[indane-2,4'-piperidine] (300 mg, 1.34 mmol, 1 eq, hydrochloric acid), 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (341 mg, 1.34 mmol, 1 eq), and diisopropylethylamine (693 mg, 5.36 mmol, 0.934 mL, 4 eq) in acetonitrile (3 mL) was stirred at 40 °C for 3 h. The mixture was concentrated under vacuum, and the resulting precipitate was triturated in dimethylformamide (1 mL), washed with acetonitrile (5 mL), and collected by filtration to afford 6-chloro-1-methyl-2-oxo-4-spiro[indane-2,4'-piperidine]-1'- yl-1,5-naphthyridine-3-carbonitrile (471 mg, 1.15 mmol, 86% yield) as a yellow solid. LC/MS (ESI) m/z: 405.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.04 (d, J = 9.2 Hz, 1H), 7.60 (d, J = 9.2 Hz, 1H), 7.24 - 7.2 (m, 2H), 7.12 (dd, J = 3.2, 5.6 Hz, 2H), 3.84 (d, J = 4.8 Hz, 4H), 3.52 (s, 3H), 2.92 (s, 4H), 1.88 - 1.76 (m, 4H).
Synthesis of Compound 72
Figure imgf000394_0001
Compound 72 Step 1: Preparation of tert-butyl 2-[(4-chlorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (2.0 g, 5.16 mmol, 1 eq) and (4-chlorophenyl)methanamine (2.19 g, 15.49 mmol, 1.89 mL, 3.0 eq) in acetonitrile (50 mL) was added potassium carbonate (7.13 g, 51.62 mmol, 10.0 eq), and the reaction mixture was stirred at 90 °C for 15 h. The reaction mixture was cooled to room temperature and partitioned between ethyl acetate (50 mL) and water (10 mL). The organic layer was washed with brine (10 mL), dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography (Petroleum ether/Ethyl acetate=10/1, 1/1, 0/1) to give the crude product (2.0 g). The crude product was purified by prep-TLC (dichloromethane/Ethyl acetate/methanol = 2/10/1) to afford tert-butyl 2-[(4- chlorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.77 g, 2.29 mmol, 44% yield) as a yellow solid.1H NMR (400 MHz, CDCl3) δ 7.34 (m, 4H), 3.90 - 3.77 (m, 2H), 3.64 - 3.45 (m, 2H), 3.00 - 2.77 (m, 4H), 2.65 - 2.51 (m, 2H), 2.35 - 2.14 (m, 2H), 1.47 - 1.45 (m, 9H). Step 2: Preparation of 5-[(4-chlorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole To a solution of tert-butyl 2-[(4-chlorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (0.1 g, 0.3 mol, 1 eq) in methanol (5 mL) was added hydrochloric acid/dioxane (4 M, 10 mL) and the reaction mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated to afford 5-[(4-chlorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole (81 mg, 296.49 µmol, hydrochloride) as a yellow solid. LC/MS (ESI) m/z: 237.1 [M+H] +.
Step 3: Preparation of 6-chloro-4-[2-[(4-chlorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 72) To a solution of 5-[(4-chlorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole (81 mg, 0.3 mmol, 1.51 eq, hydrochloride) and N,N-diisopropylethylamine (152 mg, 1.18 mmol, 0.2 mL, 6.00 eq) in dichloromethane (10 mL) and DMF (1 mL) was added 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (50 mg, 0.2 mmol, 1.0 eq), and the reaction mixture was stirred at 20 °C for 15 h. The reaction mixture was partitioned between water (20 mL) and ethyl acetate (20 mL). The mixture was stirred at 20 °C for 10 min. The precipitate was filtered, and the product was washed with water (5 mL) and ethyl acetate (5 mL x 2), then concentrated at 40 °C to afford 6-chloro-4-[2-[(4- chlorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (34.1 mg, 72.91 µmol, 37% yield) as an off-white solid. LC/MS (ESI) m/z: 470.1 [M+H] +.1H NMR (400 MHz, DMSO-d6) δ 8.03 - 7.96 (m, 1H), 7.81 - 7.74 (m, 1H), 7.41 - 7.31 (m, 4H), 4.11 - 4.01 (m, 4H), 3.91 - 3.78 (m, 2H), 3.51 - 3.47 (m, 3H), 2.94 - 2.84 (m, 2H), 2.69 - 2.64 (m, 2H), 2.38 - 2.31 (m, 2H).
Synthesis of Compound 73 Preparation of 8-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 73) To a solution of 5-(4-fluorophenyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (87.40 mg, 0.36 mmol, 1.5 eq, hydrochloride) and N,N-diisopropylethylamine (186 mg, 1.44 mmol, 0.25 mL, 6.00 eq) in dichloromethane (10 mL) and DMF (1 mL) was added (6-cyano-1- methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (80 mg, 0.24 mmol, 1.0 eq), and the reaction mixture was stirred at 20 °C for 15 h. Water (20 mL) and ethyl acetate (20 mL) were added, and the mixture was stirred at 20 °C for 10 minutes. The precipitate was filtered, and the product was washed with water (5 mL) and ethyl acetate (5 mL x 2), then concentrated at 40 °C to afford 8-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (40.8 mg, 100.83 umol, 42% yield) as a yellow solid. LC/MS (ESI) m/z: 390.1 [M+H] +.1H NMR (400MHz, DMSO-d6) δ 8.20 (s, 1H), 8.06 - 7.98 (m, 1H), 7.07 - 6.95 (m, 2H), 6.57 - 6.43 (m, 2H), 5.70 - 5.63 (m, 1H), 3.91 (br s, 2H), 3.78 - 3.57 (m, 2H), 3.54 - 3.50 (m, 3H), 3.49 - 3.44 (m, 2H), 3.24 - 3.11 (m, 4H). Synthesis of Compounds 74 and 75 Step 1: Preparation of tert-butyl 5-oxo-2H-pyrrole-1-carboxylate To a solution of (1-tert-butoxycarbonylpyrrol-2-yl)boronic acid (40.00 g, 189.55 mmol, 1 eq) in tetrahydrofuran (500 mL) at 0 °C was added a solution of NaBO3·4H2O (72.91 g, 473.88 mmol, 2.5 eq) in water (300 mL) under argon, and the reaction mixture was stirred at 20 °C for 3 h. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (300 mL x 2). The combined organic extracts were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 5-oxo-2H-pyrrole-1- carboxylate (25.00 g, 136.46 mmol, 72% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 7.21 - 7.14 (m, 1H), 6.19 - 6.09 (m, 1H), 4.37 - 4.31 (m, 2H), 1.55 (s, 9H). Step 2: Preparation of tert-butyl 2-[tert-butyl(dimethyl)silyl]oxypyrrole-1-carboxylate To a solution of tert-butyl 5-oxo-2H-pyrrole-1-carboxylate (25.00 g, 136.46 mmol, 1 eq) in dichloromethane (500 mL) under argon at 0 °C was added triethylamine (41.42 g, 409.38 mmol, 56.98 mL, 3.0 eq) followed by [tert-butyl(dimethyl)silyl] trifluoromethanesulfonate (43.29 g, 163.75 mmol, 1.2 eq), and the reaction mixture was stirred at 20 °C for 2 h. The mixture was concentrated at 30 °C, and the resulting residue was purified by silica gel column chromatography (gradient: 100/0 to 10/1 petroleum ether/ethyl acetate) to afford tert- butyl 2-[tert-butyl(dimethyl)silyl]oxypyrrole-1-carboxylate (32.00 g, 107.58 mmol, 79% yield) as a yellow oil. Step 3: Preparation of tert-butyl 2-methyl-5-oxo-2H-pyrrole-1-carboxylate To a solution of tert-butyl 2-[tert-butyl(dimethyl)silyl]oxypyrrole-1-carboxylate (32.00 g, 107.58 mmol, 1 eq) in dichloromethane (400 mL) under nitrogen was added iodomethane (45.81 g, 322.73 mmol, 20.09 mL, 3.0 eq), and the mixture was cooled to 0 °C. Silver trifluoroacetate (71.29 g, 322.73 mmol, 3.0 eq) was added, and the reaction mixture was stirred at 20 °C for 15 h. The mixture was filtered, and the cake was washed with dichloromethane (30 mL x 2). The filtrate was concentrated at 30 °C, and the resulting residue was purified by silica gel column chromatography (gradient: 1/0 to 10/1 petroleum ether/ ethyl acetate) to afford tert-butyl 2-methyl-5-oxo-2H-pyrrole-1-carboxylate (5.30 g, 26.87 mmol, 25% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 7.14 - 7.07 (m, 1H), 6.12 - 6.01 (m, 1H), 4.68 - 4.57 (m, 1H), 1.58 - 1.54 (m, 9H), 1.44 (d, J = 6.8 Hz, 3H). Step 4: Preparation of tert-butyl (1R,3aS,6aR)-5-benzyl-1-methyl-3- oxohexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate To a solution of tert-butyl 2-methyl-5-oxo-2H-pyrrole-1-carboxylate (5.30 g, 26.87 mmol, 1 eq) and trifluoroacetic acid (1.53 g, 13.44 mmol, 0.5 eq) in dichloromethane (100 mL) under nitrogen 0 °C was added a solution of N-(methoxymethyl)-1-phenyl-N- (trimethylsilylmethyl)methanamine (10.13 g, 36.28 mmol, 85% purity, 1.35 eq) in dichloromethane (20 mL) dropwise, and the reaction mixture was stirred at 20 °C for 15 h. The mixture was concentrated at 30 °C, and the resulting residue was purified by silica gel column chromatography (gradient: 50/1 to 1/1 petroleum ether/ ethyl acetate) to afford tert- butyl (1R,3aS,6aR)-5-benzyl-1-methyl-3-oxohexahydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate (8.00 g, 24.21 mmol, 90% yield) as a yellow oil. LC/MS (ESI) m/z: 331.1 [M+H]+. Step 5: Preparation of tert-butyl (1R,3aS,6aR)-5-benzyl-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate To a mixture of tert-butyl (1R,3aS,6aR)-5-benzyl-1-methyl-3-oxohexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate (8.00 g, 24.21 mmol, 1 eq) in tetrahydrofuran (50 mL) under nitrogen at 0°C was added borane dimethyl sulfide complex solution (10 M, 6.0 mL, 2.48 eq) dropwise, and the reaction mixture was stirred at 90 °C for 0.5 h. The reaction was quenched by addition of CH3OH (50 mL) and refluxed. The mixture was concentrated in vacuum, and the resulting residue was purified by semi-preparative reverse phase separation (column: Phenomenex luna C18250*80mm*10 um; mobile phase: [20-45% CH3CN in water (TFA)]) to afford tert-butyl (1R,3aS,6aR)-5-benzyl-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate (3.60 g, 11.38 mmol, 47% yield) as a colorless oil. LC/MS (ESI) m/z: 317.1 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.28 (s, 5H), 4.05 (s, 2H), 3.69 (s, 3H), 3.32 (s, 2H), 3.03 (d, J = 8.0 Hz, 1H), 2.67 (s, 1H), 2.47 (d, J = 4.0 Hz, 2H), 1.33 (s, 9H), 1.01 (d, J = 6.4 Hz, 3H). Step 6: Preparation of tert-butyl (1R,3aS,6aR)-1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate To a solution of tert-butyl (1R,3aS,6aR)-5-benzyl-1-methylhexahydropyrrolo[3,4-c]pyrrole- 2(1H)-carboxylate (2.00 g, 6.32 mmol, 1 eq) in methanol (30 mL) was added palladium on activated carbon (0.20 g, 10% purity) under nitrogen, and the suspension was degassed under vacuum and purged with hydrogen several times. The mixture was stirred under hydrogen (20 psi) at 60 °C for 12 h. The reaction mixture was filtered, and the filtrate was concentrated to afford tert-butyl (1R,3aS,6aR)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (1.20 g, 5.30 mmol, 84% yield) as a colorless oil. Step 7: Preparation of tert-butyl (1S,3aR,6aS)-5-(4-fluorobenzyl)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate To a mixture of 4-fluorobenzaldehyde (219 mg, 1.77 mmol, 2 eq), tert-butyl (1R,3aS,6aR)-1- methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (200 mg, 0.88 mmol, 1 eq), and triethylamine (268 mg, 2.65 mmol, 0.37 mL, 3 eq) in 1,2-dichloroethane (20 mL) was added sodium cyanoborohydride (111 mg, 1.77 mmol, 2 eq) in one portion under nitrogen, and the reaction mixture was stirred at 60 °C for 10 h. The residue was poured into ice-water (w/w = 1/1) (10 mL), and the aqueous phase was extracted with dichloromethane (20 mL x 2). The combined organic extract was washed with brine (20 mL), dried with anhydrous sodium sulfate, filtered, and concentrated in vacuum to afford tert-butyl (1S,3aR,6aS)-5-(4- fluorobenzyl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (130 mg, 0.39 mmol, 44% yield) as a yellow gum. LC/MS (ESI) m/z: 334.9 [M+H]+. Step 8: Preparation of (1S,3aS,6aS)-5-(4-fluorobenzyl)-1- methyloctahydropyrrolo[3,4-c]pyrrole To a mixture of tert-butyl (1S,3aR,6aS)-5-(4-fluorobenzyl)-1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate (120 mg, 0.36 mmol, 1 eq) in dichloromethane (10 mL) was added trifluoroacetic acid (456 mg, 4.00 mmol, 2 mL, 11.15 eq) in one portion under nitrogen, and the reaction mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated in reduced pressure to afford (1S,3aS,6aS)-5-(4-fluorobenzyl)-1- methyloctahydropyrrolo[3,4-c]pyrrole (120 mg, 0.34 mmol, 96% yield, TFA salt) as a yellow gum. Step 9: Preparation of 8-[(3aS,4S,6aR)-2-[(4-fluorophenyl)methyl] -4-methyl- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile To a mixture of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (115 mg, 0.34 mmol, 1 eq) and (1S,3aS,6aS)-5-(4-fluorobenzyl)-1- methyloctahydropyrrolo[3,4-c]pyrrole (120 mg, 0.34 mmol, 1 eq, trifluoroacetate) in isopropyl alcohol (10 mL) was added diisopropylethylamine (134 mg, 1.03 mmol, 3 eq) in one portion under nitrogen, and the reaction mixture was stirred at 100 °C for 10 h. The mixture was concentrated in reduced pressure, and the resulting residue was purified by semi- preparative reverse phase separation (column: Phenomenex Synergi Polar-RP 100*25mm*4um; mobile phase: [26-46% CH3CN in water (TFA)]) to afford 8- [(3aS,4S,6aR)-2-[(4-fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (120 mg, 0.29 mmol, 83% yield) as a yellow gum. LC/MS (ESI) m/z: 418.2 [M+H]+. Step 10: Preparation of 8-[(3aS,4S,6aR)-2-[(4-fluorophenyl)methyl] -4-methyl- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (Compound 74) and 8-[(3aR,4R,6aS)-2-[(4- fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 75) The mixture of enantiomers (100 mg, 0.24 mmol, 1 eq) was separated by supercritical fluid chromatography (column: DAICEL CHIRALPAK AS(250mm*30mm,10um); mobile phase: [0.1%NH4OH in CH3OH]; B%: 65%-65%, 4.2 min; 50 min). Absolute configuration was arbitrarily assigned. The first peak was purified further by semi-preparative reverse phase (column: Waters Xbridge 150*25mm*5um; mobile phase: [36-66% CH3CN in water (NH4OH)]) to give 8- [(3aS,4S,6aR)-2-[(4-fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (14.6 mg, 0.03 mmol, 14% yield) (Compound 74) as an off-white solid. LC/MS (ESI) m/z: 418.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.82 (d, J = 8.8 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 7.35 - 7.29 (m, 2H), 7.02 (t, J = 8.8 Hz, 2H), 5.91 (s, 1H), 5.33 - 5.18 (m, 1H), 3.81 - 3.74 (m, 1H), 3.68 - 3.54 (m, 6H), 3.05 (br s, 3H), 2.76 - 2.66 (m, 1H), 2.53 - 2.43 (m, 1H), 2.39 - 2.29 (m, 1H), 1.10 (d, J = 6.4 Hz, 3H). The second peak to give 8-[(3aR,4R,6aS)-2-[(4-fluorophenyl)methyl]-4-methyl-1,3,3a,4,6,6a -hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (17.7 mg, 0.04 mmol, 17% yield) (Compound 75) as an off-white solid. LC/MS (ESI) m/z: 418.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.72 (d, J = 8.8 Hz, 1H), 7.60 (d, J = 8.8 Hz, 1H), 7.26 - 7.20 (m, 2H), 6.92 (t, J = 8.8 Hz, 2H), 5.82 (s, 1H), 5.22 - 5.08 (m, 1H), 3.71 - 3.65 (m, 1H), 3.58 - 3.45 (m, 6H), 2.96 (br s, 3H), 2.63 (br s, 1H), 2.46 - 2.35 (m, 1H), 2.31 - 2.17 (m, 1H), 1.00 (d, J = 6.4 Hz, 3H). Synthesis of Compounds 76 and 70
Step 1: Preparation of tert-butyl (1R,3aS,6aR)-5-(6-cyano-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
Figure imgf000405_0001
To a mixture of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (295 mg, 0.88 mmol, 1 eq) and tert-butyl (1R,3aS,6aR)-1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate (200 mg, 0.88 mmol, 1 eq) in n-butyl alcohol (20 mL) was added diisopropylethylamine (343 mg, 2.65 mmol, 3 eq) in one portion under nitrogen, and the reaction mixture was stirred at 100 °C for 10 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 5/1 to 0/1 petroleum ether/ ethyl acetate) to afford tert-butyl (1R,3aS,6aR)-5-(6- cyano-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate (210 mg, 0.51 mmol, 58% yield) as a yellow gum. LC/MS (ESI) m/z: 354.1 [M-56+H]+. Step 2: Preparation of 5-methyl-8-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4- c]pyrrol-2(1H)-yl)-6-oxo-5,6-dihydro-1,5-naphthyridine-2-carbonitrile
Figure imgf000405_0002
racemic To a mixture of tert-butyl (1R,3aS,6aR)-5-(6-cyano-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridin-4-yl)-1-methylhexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (200 mg, 0.49 mmol, 1 eq) in dichloromethane (20 mL) was added trifluoroacetic acid (3.08 g, 27.01 mmol, 2 mL, 55.30 eq) under nitrogen, and the reaction mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated under vacuum to give 5-methyl-8-((3aR,4R,6aR)-4- methylhexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-6-oxo-5,6-dihydro-1,5-naphthyridine-2- carbonitrile (180 mg, 0.43 mmol, 87% yield, TFA salt) as a yellow gum. LC/MS (ESI) m/z: 309.8 [M+H]+. Step 3: Preparation of 8-[(3aR,4R,6aR)-5-[(4-fluorophenyl)methyl] -4-methyl- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile To a mixture of 4-fluorobenzaldehyde (106 mg, 0.85 mmol, 2 eq), triethylamine (215 mg, 2.13 mmol, 5 eq), and 5-methyl-8-((3aR,4R,6aR)-4-methylhexahydropyrrolo[3,4-c]pyrrol- 2(1H)-yl)-6-oxo-5,6-dihydro-1,5-naphthyridine-2-carbonitrile (180 mg, 0.43 mmol, 1 eq, trifluoroacetate) in 1,2-dichloroethane (10 mL) was added sodium triacetoxyborohydride (270 mg, 1.28 mmol, 3 eq) in one portion under nitrogen, and the reaction mixture was stirred at 60 °C for 10 h. The residue was poured into ice-water (w/w = 1/1) (10 mL), and the aqueous phase was extracted with dichloromethane (20 mL x 2). The combined organic extract was washed with brine (20 mL), dried with anhydrous sodium sulfate, filtered, and concentrated in vacuum. The resulting residue was purified by semi-preparative reverse phase separation (column: Phenomenex Synergi Polar-RP 100*25mm*4µm; mobile phase: [26-46% CH3CN in water (TFA)]) to afford 8-[(3aR,4R,6aR)-5-[(4-fluorophenyl)methyl] -4- methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-5-methyl-6-oxo-1,5-naphthyridine- 2-carbonitrile (150 mg, 0.36 mmol, 85% yield) as a yellow gum. LC/MS (ESI) m/z: 418.2 [M+H]+. Step 4: Preparation of WC-ARV-JM-004-E, 8-[(3aR,4R,6aR)-5-[(4- fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 76) and 8-[(3aS,4S,6aS)- 5-[(4-fluorophenyl)methyl] -4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 70) The mixture of enantiomers (150 mg, 0.35 mmol, 1 eq) was separated by supercritical fluid chromatography (column: DAICEL CHIRALCEL OJ(250mm*30mm,10um); mobile phase: [0.1%NH4OH in CH3OH]; B%: 40%-40%, 2.9 min; 25 min). Absolute configuration was arbitrarily assigned. 8-[(3aR,4R,6aR)-5-[(4-fluorophenyl) methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-2-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (44.1 mg, 0.10 mmol, 28% yield, 98% purity) (Compound 76) as an off-white solid. LC/MS (ESI) m/z: 418.2 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 7.71 (d, J = 8.8 Hz, 1H), 7.58 (d, J = 8.8 Hz, 1H), 7.23 (br t, J = 6.4 Hz, 2H), 6.93 (t, J = 8.8 Hz, 2H), 5.80 (s, 1H), 4.02 - 3.78 (m, 3H), 3.63 - 3.51 (m, 5H), 3.26 - 3.07 (m, 2H), 2.92 - 2.76 (m, 1H), 2.59 - 2.45 (m, 1H), 2.43 - 2.28 (m, 1H), 2.17 - 2.00 (m, 1H), 1.31 - 1.14 (m, 3H). 8-[(3aS,4S,6aS)-5-[(4-fluorophenyl)methyl]-4-methyl-1,3,3a,4,6,6a-hexahydropyrrolo [3,4- c]pyrrol-2-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (95.2 mg, 0.22 mmol, 62% yield, 98% purity) (Compound 70) as a yellow solid. LC/MS (ESI) m/z: 418.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ: 7.76 (d, J = 8.8 Hz, 1H), 7.65 (d, J = 8.8 Hz, 1H), 7.46 - 7.38 (m, 2H), 7.10 - 7.02 (m, 2H), 5.95 (s, 1H), 4.49 (br d, J = 12.8 Hz, 1H), 4.11 (br d, J = 11.2 Hz, 1H), 3.90 - 3.71 (m, 3H), 3.57 (s, 3H), 3.38 - 3.19 (m, 3H), 3.10 - 2.93 (m, 2H), 2.70 - 2.56 (m, 1H), 1.70 - 1.56 (m, 3H). Synthesis of Compound 78 Step 1: Preparation of tert-butyl 2-benzyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate To a solution of tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (0.4 g, 1.03 mmol, 1.0 eq) and potassium carbonate (1.43 g, 10.32 mmol, 0.64 mL, 10.0 eq) in acetonitrile (20 mL) was added benzylamine (331 mg, 3.10 mmol, 0.33 mL, 3.0 eq), and the reaction mixture was stirred at 90 °C for 15 h. The reaction mixture was cooled to room temperature and partitioned between ethyl acetate (50 mL) and water (10 mL). The organic layer was washed with brine (10 mL), dried over sodium sulfate, filtered, and concentrated. The resulting residue was purified by prep-TLC (SiO2, Petroleum ether/Ethyl acetate=2/3) to afford tert-butyl 2-benzyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.15 g, 496.02 µmol, 48% yield) as a yellow oil. LC/MS (ESI) m/z: 303.5 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.36 - 7.28 (m, 5H), 3.99 - 3.81 (m, 2H), 3.63 - 3.46 (m, 2H), 3.02 - 2.83 (m, 4H), 2.73 - 2.50 (m, 2H), 2.33 - 2.15 (m, 2H), 1.46 (s, 9H). Step 2: Preparation of tert-butyl 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole- 5-carboxylate To a stirred solution of tert-butyl 2-benzyl-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate (150.00 mg, 0.5 mmol, 1 eq) in ethanol (10 mL) was added palladium on activated carbon catalyst (0.05 g, 10% purity) under nitrogen, and the reaction mixture was degassed with hydrogen (3X) and stirred at 20 °C for 15 h under hydrogen (15 psi). The reaction mixture was degassed with hydrogen (3X) and stirred at 20 °C for 15 h under hydrogen (15 psi). The reaction mixture was filtered, and the cake was washed with ethanol (10 mL x 2). The filtrate was concentrated under reduced pressure to afford tert-butyl 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate (90 mg, 423.95 µmol, 85% yield) as a white solid. LC/MS (ESI) m/z: 156.9 [M-55] +.1H NMR (400 MHz, CDCl3) δ 4.43 - 4.16 (m, 1H), 3.76 - 3.53 (m, 2H), 2.91 (br s, 6H), 2.45 - 2.19 (m, 2H), 1.53 - 1.39 (m, 9H). Step 3: Preparation of tert-butyl 2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate A solution of tert-butyl 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate (90 mg, 0.42 mmol, 1 eq) and 4-chloro-2-hydroxy-benzaldehyde (79 mg, 0.51 mmol, 1.2 eq) in ethanol (10 mL) and dichloromethane (5 mL) was stirred at 20 °C for 2 h. Sodium cyanoborohydride (79 mg, 1.27 mmol, 3.0 eq) was then added, and the reaction mixture was stirred at 20 °C for 13 h under nitrogen. The product was purified by prep-TLC (petroleum ether/ethyl acetate=1/2) followed by prep-TLC (dichloromethane/methanol=10/1) to afford tert-butyl 2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (40 mg, 113.36 µmol, 27% yield) as a yellow solid. LC/MS (ESI) m/z: 353.0 [M+H] +.1H NMR (400 MHz, CDCl3) δ 6.99 - 6.95 (m, 1H), 6.94 - 6.90 (m, 1H), 6.78 (dd, J = 1.8, 7.8 Hz, 1H), 4.12 - 3.96 (m, 2H), 3.67 - 3.57 (m, 2H), 3.19 - 3.07 (m, 2H), 3.02 - 2.93 (m, 2H), 2.76 - 2.65 (m, 2H), 2.34 - 2.25 (m, 2H), 1.47 - 1.46 (m, 9H). Step 4: Preparation of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5- ylmethyl)-5-chloro-phenol To a solution of tert-butyl 2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (40 mg, 0.11 mmol, 1 eq) in dichloromethane (5 mL) was added hydrochloric acid/methanol (4 M, 10 mL) and the mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated under reduced pressure to afford 2- (2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-ylmethyl)-5-chloro-phenol (33 mg, crude, hydrochloride) as a yellow solid. LC/MS (ESI) m/z: 253.0 [M+H] +.
Step 5: Preparation of 4-[(3aR,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 78) A solution of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-ylmethyl)-5-chloro- phenol (33 mg, 0.11 mmol, 1 eq, hydrochloride), triethylamine (57 mg, 0.57 mmol, 0.08 mL, 5.0 eq), and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (29 mg, 0.11 mmol, 1 eq) in dichloromethane (5 mL) and DMF (1 mL) was stirred at 20 °C for 15 h. The reaction mixture was concentrated under reduced pressure, and the product was purified by prep-HPLC (column: Waters Xbridge 150*25 mm* 5µm; mobile phase: [45-75% CH3CN in water (NH4HCO3)]). Pure fractions were lyophilized to afford 4-[(3aR,6aR)-2-[(4-chloro-2- hydroxy-phenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (13.3 mg, 28.28 µmol, 25% yield) as a white solid. LC/MS (ESI) m/z: 470.1 [M+H] +.1H NMR (400 MHz, DMSO-d6) δ 8.02 - 7.96 (m, 1H), 7.81 - 7.73 (m, 1H), 7.15 - 7.10 (m, 1H), 6.83 - 6.73 (m, 2H), 4.15 - 4.05 (m, 4H), 4.01 - 3.89 (m, 2H), 3.49 (s, 3H), 3.00 - 2.91 (m, 2H), 2.71 - 2.67 (m, 2H), 2.44 - 2.35 (m, 2H).
Synthesis of Compounds 79 and 80 Step 1: Preparation of tert-butyl 4-(4-fluoro-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate To a solution of tert-butyl 4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate (300 mg, 0.97 mmol, 1 eq) in methanol (5 mL) were added paraformaldehyde (292 mg, 9.73 mmol, 0.26 mL, 10 eq) and sodium cyanoborohydride (61 mg, 0.97 mmol, 1 eq), and the reaction mixture was stirred at 25 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (gradient: 1/0 to 20/1 petroleum ether/ethyl acetate) to afford tert-butyl 4-(4-fluoro-N-methyl- anilino)-3-methyl-piperidine-1 -carboxylate (200 mg, 0.62 mmol, 64% yield) as a yellow oil. LC/MS (ESI) m/z: 323.2 [M+H]+. Step 2: Preparation of N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine To a solution of tert-butyl 4-(4-fluoro-N-methyl-anilino)-3-methyl-piperidine-1-carboxylate (50 mg, 0.15 mmol, 1 eq) in dichloromethane (5 mL) was added trifluoroacetic acid (17 mg, 0.15 mmol, 0.01 mL, 1 eq), and the reaction mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give N-(4-fluorophenyl)-N,3- dimethyl-piperidin-4-amine (50 mg, 148.67 µmol, 96% yield, TFA salt) as a brown oil. Step 3: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6-chloro-4-[(3R,4R)- 4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile To a solution of N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine (50 mg, 148.67 µmol, 1 eq, trifluoroacetic acid) in acetonitrile (5 mL) were added diisopropylethylamine (19.21 mg, 148.67 µmol, 25.89 µL, 1 eq) and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3 - carbonitrile (30.22 mg, 118.93 µmol, 0.8 eq), and the reaction mixture was stirred at 60 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10µm; mobile phase: [33-63% CH3CN in water (formic acid)]) to afford the desired products. 6-Chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (19.8 mg, 42.85 µmol, 29% yield) (Compound 79) was obtained as a yellow solid. LC/MS (ESI) m/z: 440.0 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.82 (d, J = 9.2 Hz, 1H), 7.09 - 6.99 (m, 2H), 6.86 (dd, J = 4.4, 9.2 Hz, 2H), 4.33 (d, J = 12.0 Hz, 1H), 4.06 - 3.96 (m, 1H), 4.06 - 3.95 (m, 1H), 3.84 (dd, J = 2.4, 12.8 Hz, 1H), 3.53 (s, 3H), 3.46 - 3.40 (m, 1H), 2.82 (s, 3H), 2.68 - 2.62 (m, 1H), 2.33 (s, 1H), 1.74 ( d, J = 11.2 Hz, 1H), 0.87 (d, J = 6.4 Hz, 3H). 6-Chloro-4-[(3R,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (5.8 mg, 12.37 µmol, 8% yield) (Compound 80) was obtained as a yellow solid. LC/MS (ESI) m/z: 440.0 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.07 - 6.96 (m, 2H), 6.90 - 6.83 (m, 2H), 4.28 (d, J = 12.4 Hz, 2H), 3.73 (dt, J = 3.6, 11.1 Hz, 1H), 3.66 - 3.55 (m, 1H), 3.53 (s, 3H), 3.17 (s, 1H), 2.70 - 2.66 (m, 3H), 2.28 (dd, J = 4.0, 6.4 Hz, 1H), 1.97 - 1.84 (m, 1H), 1.77 - 1.65 (m, 1H), 0.83 (d, J = 6.4 Hz, 3H). Synthesis of Compound 81
Step 1: Preparation of (3S,4R)-N-(4-fluorophenyl)-3-methyl-piperidin-4-amine To a solution of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate (100.00 mg, 324.26 µmol, 1 eq) in dichloromethane (5 mL) was added trifluoroacetic acid (36.97 mg, 324.26 µmol, 24.01 µL, 1 eq), and the reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give (3S,4R)-N-(4- fluorophenyl)-3-methyl-piperidin-4-amine (100 mg, 310.27 µmol, 96% yield, TFA salt) as brown oil. Step 2: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 81) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4- fluorophenyl)-3-methyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 426.1 [M+H]+. 1HNMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 6.90 (t, J = 8.8 Hz, 2H), 6.76 - 6.65 (m, 2H), 5.60 (d, J = 8.8 Hz, 1H), 4.01 (d, J = 2.4 Hz, 1H), 3.85 ( d, J = 4.8 Hz, 2H), 3.77 - 3.59 (m, 2H), 3.52 (s, 3H), 2.41 - 2.21 (m, 1H), 2.13 - 2.04 (m, 1H), 1.84 (ddd, J = 3.2, 9.6 Hz, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 82 Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 82) The title compound was made in an analogous manner to 6-chloro-4-[(3S,4R)-4-(4- fluoroanilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 426.1 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 6.95 - 6.85 (m, 2H), 6.68 - 6.56 (m, 2H), 5.48 (d, J = 9.2 Hz, 1H), 4.32 - 4.15 (m, 2H), 3.65 - 3.54 (m, 1H), 3.52 (s, 3H), 3.26 - 3.09 (m, 2H), 2.20 - 2.09 (m, 1H), 2.08 - 1.92 (m, 1H), 1.57 - 1.40 (m, 1H), 0.98 (d, J = 6.4 Hz, 3H).
Synthesis of Compound 83 Step 1: Preparation of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl-methyl- amino]piperidine-1-carboxylate To a mixture of tert-butyl 4-(methylamino)piperidine-1-carboxylate (500 mg, 2.33 mmol, 1.0 eq) and 4-chloro-2-hydroxy-benzaldehyde (365 mg, 2.33 mmol, 1.0 eq) in dichloroethane (5 mL) and CH3OH (5 mL) was added acetic (560 mg, 9.33 mmol, 4.0 eq), and the resulting mixture was stirred at 25 °C for 5 hours. NaBH(OAc)3 (2.47 g, 11.67 mmol, 5.0 eq) was then added, and the reaction mixture was stirred at 25 °C for 3 hours. The reaction mixture was filtered and concentrated in vacuo. The resulting residue was purified by prep-HPLC (column: Xtimate C18150*40mm*10um; mobile phase: [0-30% CH3CN in water (0.225% formic acid)]). Pure fractions were combined, then lyophilized to afford tert-butyl 4-[(4- chloro-2-hydroxy-phenyl)methyl-methyl-amino]piperidine-1-carboxylate (335 mg, 0.906 mmol, 39% yield) as a white solid. LC/MS (ESI) m/z: 355.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.04 - 6.92 (m, 2H), 6.88 - 6.80 (m, 1H), 4.34 - 4.17 (m, 2H), 4.02 - 3.82 (m, 2H), 3.08 - 2.93 (m, 1H), 2.78 - 2.68 (m, 2H), 2.46 (s, 3H), 2.01 (d, J = 11.6 Hz, 2H), 1.66 - 1.61 (m, 2H), 1.47 (s, 9H). Step 2: Preparation of 5-chloro-2-[[methyl(4-piperidyl)amino]methyl]phenol To a mixture of tert-butyl 4-[(4-chloro-2-hydroxy-phenyl)methyl-methyl-amino]piperidine-1- carboxylate (120 mg, 0.338 mmol, 1.0 eq) in dichloromethane (1 mL) was added HCl/dioxane (4 M, 1 mL, 11.8 eq) in one portion at 25°C. The mixture was stirred at 25°C for 1 hour. The reaction mixture was concentrated in vacuum to afford 5-chloro-2- [[methyl(4-piperidyl)amino]methyl]phenol (86 mg, 0.311 mmol, 92% yield) as a white solid. LC/MS (ESI) m/z: 255.2 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-[(4-chloro-2-hydroxy-phenyl)methyl-methyl- amino]-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 83) To a mixture of 5-chloro-2-[[methyl(4-piperidyl)amino]methyl]phenol (95 mg, 0.374 mmol, 1.0 eq) and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (95 mg, 0.374 mmol, 1.0 eq) in CH3CN (4 mL) at 20°C was added diisopropylethylamine (1.87 mmol, 326 µL, 5.0 eq), and the reaction mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated in vacuo, and the resulting residue was purified by prep-HPLC (column: Xtimate C18100*30mm*10 µm; mobile phase: [15-45% CH3CN in water (0.225% formic acid)]). Pure fractions were combined, then lyophilized to afford 6-chloro-4-[4-[(4-chloro-2- hydroxy-phenyl)methyl-methyl-amino]-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (80.3 mg, 0.168 mmol, 45% yield) as a yellow solid. LC/MS (ESI) m/z: 472.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.14 (d, J = 8.0 Hz, 1H), 6.79 (dd, J = 2.0, 8.0 Hz, 1H), 6.76 (d, J = 2.0 Hz, 1H), 4.29 (d, J = 13.2 Hz, 2H), 3.77 (s, 2H), 3.55 (s, 2H), 3.52 (s, 3H), 2.96 - 2.90 (m, 1H), 2.22 (s, 3H), 1.96 (d, J = 10.4 Hz, 2H), 1.90 - 1.77 (m, 2H). Synthesis of Compound 84 Step 1: 3.2 Preparation of tert-butyl 4-(4-fluoro-N-methyl-anilino)piperidine-1- carboxylate To a solution of tert-butyl 4-(4-fluoroanilino)piperidine-1-carboxylate (300 mg, 1.02 mmol, 1.0 eq) in DMF (2 mL) at 25°C was added K2CO3 (155 mg, 1.12 mmol, 1.1 eq) and iodomethane (2.55 mmol, 159 µL, 2.5 eq) dropwise, and the reaction mixture was stirred at 60 °C for 12 hours under N2. The reaction mixture was filtered, and the residue was purified by prep-HPLC (column: Xtimate C18150*40mm*10um; mobile phase: [30-60% CH3CN in water (0.225% formic acid)]). Pure fractions were combined, then lyophilized to afford tert- butyl 4-(4-fluoro-N-methyl-anilino)piperidine-1-carboxylate (100 mg, 292 mmol, 29% yield) as a white solid. LC/MS (ESI) m/z: 309.3 [M+H]+. Step 2: Preparation of N-(4-fluorophenyl)-N-methyl-piperidin-4-amine To tert-butyl 4-(4-fluoro-N-methyl-anilino)piperidine-1-carboxylate (100 mg, 0.324 mmol, 1.0 eq) in dichloromethane (1 mL) was added HCl/dioxane (4 M, 1 mL, 12.3 eq) in one portion at 25 °C. The mixture was stirred at 25°C for 1 hour. The reaction mixture was concentrated in vacuum to afford N-(4-fluorophenyl)-N-methyl-piperidin-4-amine (67 mg, 0.322 mmol, 99% yield) as a white solid. LC/MS (ESI) m/z: 209.0 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-(4-fluoro-N-methyl-anilino)-1-piperidyl]-1- methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile (Compound 84) To a mixture of N-(4-fluorophenyl)-N-methyl-piperidin-4-amine (67 mg, 0.322 mmol, 1.0 eq) and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (82 mg, 0.322 mmol, 1.0 eq) in CH3CN (2 mL) at 25°C was added diisopropylamine (1.61 mmol, 280 µL, 5.0 eq), and the reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated in vacuum, and the resulting residue was purified by prep-HPLC (column: Xtimate C18100*30mm*10um; mobile phase: [0-60% CH3CN in water (0.225% formic acid)]). Pure fractions were combined, then lyophilized to afford 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (64.3 mg, 0.146 mmol, 45% yield) as a yellow solid. LC/MS (ESI) m/z: 426.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.09 - 6.97 (m, 2H), 6.93 - 6.86 (m, 2H), 4.29 (d, J = 12.8 Hz, 2H), 4.02 (tt, J = 3.6, 11.6 Hz, 1H), 3.57 - 3.48 (m, 5H), 2.70 (s, 3H), 1.98 (dq, J = 3.2, 12.0 Hz, 2H), 1.79 (d, J = 10.4 Hz, 2H). Synthesis of Compound 85 Step 1: Preparation of spiro[indoline-2,4'-piperidine] To a solution of 1'-benzylspiro[indoline-2,4'-piperidine] (230 mg, 0.826 mmol) in CH3OH (5 mL) was added Pd(OH)2 (100 mg, 0.712 mmol) and AcOH (99.23 mg, 1.65 mmol, 94.50 µL) under N2. The mixture was degassed and purged with H2 (3X) and stirred at 50 °C for 3 hours under H2 (50 Psi). The reaction mixture was filtered, and the filter cake was washed with CH3OH (200 mL). The filtrate was concentrated under reduced pressure to give spiro[indoline-2,4'-piperidine] (150 mg, crude) as a white solid. LC/MS (ESI) m/z: 189.3 [M+H]+. Step 2: Preparation of 6-chloro-1-methyl-2-oxo-4-spiro[indoline-2,4'-piperidine]-1'- yl-1,5-naphthyridine-3-carbonitrile (Compound 85) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile starting from spiro[indoline-2,4'-piperidine]. (yellow solid). LC/MS (ESI) m/z: 406.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.00 (d, J = 7.2 Hz, 1H), 6.91 (t, J = 7.6 Hz, 1H), 6.53-6.43 (m, 2H), 6.09 (s, 1H), 4.04-3.91 (m, 2H), 3.89- 3.77 (m, 2H), 3.53 (s, 3H), 2.90 (s, 2H), 1.93-1.83 (m, 4H). Synthesis of Compound 86 Step 1: Preparation of 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine]-3-one To a solution of 1'-benzylspiro[indoline-2,4'-piperidine]-3-one (500 mg, 1.71 mmol) in DMF (5 mL) at 0°C was added NaH (68 mg, 1.71 mmol, 60% purity), and the reaction mixture was stirred at 25 °C for 1 hour. Iodomethane (242 mg, 1.71 mmol, 106 µL) was then added at 0°C, and mixture was stirred at 25 °C for 1 hour. The reaction mixture was poured into saturated aqueous NH4Cl (10 mL), and the resulting mixture was extracted with ethyl acetate (10 mL×3). The combined organic extracts were washed with brine (10 mL×3), dried over Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography (gradient: 0~50% ethyl acetate in petroleum ether) to afford 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine]-3-one (480 mg, 1.25 mmol, 73% yield) as a yellow oil. LC/MS (ESI) m/z: 307.2 [M+H]+. Step 2: Preparation of 1-methylspiro[indoline-2,4'-piperidine]-3-one To a solution of 1'-benzyl-1-methyl-spiro[indoline-2,4'-piperidine]-3-one (50 mg, 0.163 mmol, 1 eq) in dichloroethane (2 mL) was added 1-chloroethyl carbonochloridate (122 mg, 0.855 mmol, 5.24 eq), and the reaction mixture was stirred at 50°C for 3 hours. The mixture was concentrated to give a residue. A mixture of the residue in MeOH (5 mL) was stirred at 25°C for 0.5 hour. The mixture was concentrated to give 1-methylspiro[indoline-2,4'- piperidine]-3-one (30 mg, crude) as a yellow solid. LC/MS (ESI) m/z: 217.0 [M+H]+. Step 3: Preparation of 6-chloro-1-methyl-4-(1-methyl-3-oxo-spiro[indoline-2,4'- piperidine]-1'-yl)-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 86) The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-fluoro-N- methyl-anilino)-1-piperidyl]-1-methyl-2-oxo-1, 5-naphthyridine-3-carbonitrile starting from 1-methylspiro[indoline-2,4'-piperidine]-3-one. (yellow solid). LC/MS (ESI) m/z: 434.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 9.2 Hz, 1H), 7.83 (d, J = 9.0 Hz, 1H), 7.54 (d, J = 1.2 Hz, 1H), 7.48 (d, J = 7.6 Hz, 1H), 6.98 (d, J = 8.4 Hz, 1H), 6.73 (t, J = 7.4 Hz, 1H), 4.20-4.12 (m, 4H), 3.55 (s, 3H), 2.93 (s, 3H), 2.36-2.29 (m, 2H), 1.62 (d, J = 13.6 Hz, 2H). Synthesis of Compounds 87 and 42 Step 1: Preparation of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1- carboxylate and tert-butyl (3R,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1- carboxylate To a solution of 4-fluoroaniline (1 g, 9.00 mmol, 0.86 mL, 1 eq) in methanol (10 mL) were added tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (1.92 g, 9.00 mmol, 1 eq), sodium cyanoborohydride (565 mg, 9.00 mmol, 1 eq), and acetic acid (54 mg, 0.9 mmol, 51.47 uL, 0.1 eq), and the reaction mixture was stirred at 25 °C for 12 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by column chromatography on silica gel (gradient: 1/1 to 13/1 petroleum ether/ethyl acetate) to afford the desired products. tert-Butyl (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate (500 mg, 1.62 mmol, 18.00% yield) was obtained as a yellow solid.1HNMR (400 MHz, DMSO-d6) δ 6.93 - 6.83 (m, 1H), 6.88 (t, J = 8.8 Hz, 1H), 6.65 - 6.58 (m, 2H), 5.34 (d, J = 8.8 Hz, 1H), 3.78 - 3.44 (m, 3H), 3.19 - 2.94 (m, 2H), 2.05 - 1.96 (m, 1H), 1.57 - 1.46 (m, 2H), 1.39 (s, 9H), 0.77 (d, J = 7.2 Hz, 3H). tert-Butyl (3R,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate (500 mg, 1.62 mmol, 18.02% yield) was obtained as a yellow solid.1HNMR (400 MHz, DMSO-d6) δ 6.90 - 6.85 (m, 2H), 6.57 - 6.53 (m, 2H), 5.30 - 5.27 (m, 1H), 3.88 - 3.84 (m, 2H), 2.99 - 2.94 (m, 2H), 2.85 - 2.81 (m, 1H), 2.64 - 2.60 (m, 1H), 1.92 - 1.88 (m, 1H), 1.47 - 1.45 (m, 1H), 1.42 (s, 9H), 1.09 - 1.05 (m, 1H), 0.90 (d, J = 7.2 Hz, 3H) Step 2: Preparation of tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl- piperidine-1-carboxylate A mixture of tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate (200 mg, 0.65 mmol, 1 eq), sodium cyanoborohydride (61 mg, 0.97 mmol, 1.5 eq), and formaldehyde (58 mg, 0.71 mmol, 0.053 mL, 37% purity, 1.1 eq) in CH3OH (2 mL) was stirred at 25 °C for 12 h. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*2). The combined organic extracts were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (gradient: 100:1 to 1:1 petroleum ether/ethyl acetate) to give tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate (180 mg, 0.56 mmol, 86% yield) as a white solid. LC/MS (ESI) m/z: 267.2 (M- 55)+. Step 3: Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile A mixture of tert-butyl (3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate (100 mg, 0.31 mmol, 1 eq) and trifluoroacetic acid (35 mg, 0.31 mmol, 0.023 mL, 1 eq) in dichloromethane (1 mL) was stirred at 25°C for 15 minutes. The mixture was concentrated under vacuum to give (3S,4R)-N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4- amine (105 mg, crude, TFA salt) as a red oil. LC/MS (ESI) m/z: 223.1 [M+H]+. Step 4: Preparation of 6-chloro-4-[(3R,4R)-4-(4-fluoroanilino)-3-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile A mixture of (3S,4R)-N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine (105 mg, 0.31 mmol, 1 eq, trifluoroacetic acid), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (79 mg, 0.31 mmol, 1 eq), and diisopropylethylamine (121 mg, 0.94 mmol, 0.16 mL, 3 eq) in acetonitrile (1 mL) was stirred at 40 °C for 12 h. The mixture was concentrated under vacuum, and the precipitate was triturated in DMF (2 mL) and collected by filtration to afford 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (59 mg, 0.13 mmol, 43% yield) as a yellow solid. LC/MS (ESI) m/z: 440.0 [M+H]+. Step 5: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 87) and 6- chloro-4-[(3R,4S)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (Compound 42) The enantiomers were separated by SFC (column: DAICEL CHIRALPAK AD(250mm*30mm, 10 µm); mobile phase: ethanol (0.1% NH4OH) in CO2). The absolute configuration was arbitrarily assigned. 6-Chloro-4-[(3S,4R)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (30.3 mg, 0.07 mmol, 51% yield) (Compound 87) was obtained as a yellow solid. LC/MS (ESI) m/z: 440.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.82 (d, J = 9.2 Hz, 1H), 7.08 - 7.01 (m, 2H), 6.87 (dd, J = 4.4, 9.2 Hz, 2H), 4.38 - 4.29 (m, 1H), 4.06 - 3.94 (m, 2H), 3.89 - 3.79 (m, 1H), 3.53 (s, 3H), 3.47 - 3.39 (m, 2H), 2.82 (s, 3H), 2.61 (s, 1H), 1.80 - 1.69 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H). 6-Chloro-4-[(3R,4S)-4-(4-fluoro-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (11.9 mg, 0.03 mmol, 20% yield) (Compound 42) was obtained as a yellow solid. LC/MS (ESI) m/z: 440.0 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 7.04 (t, J = 8.8 Hz, 2H), 6.86 (dd, J = 4.4, 8.8 Hz, 2H), 4.39 - 4.27 (m, 1H), 4.09 - 3.91 (m, 2H), 3.84 (d, J = 12.8 Hz, 1H), 3.53 (s, 3H), 3.48 - 3.38 (m, 2H), 2.82 (s, 3H), 2.62 (d, J = 3.2 Hz, 1H), 1.74 (d, J = 11.2 Hz, 1H), 0.87 (d, J = 6.8 Hz, 3H). Synthesis of Compounds 89 Step 1: Preparation of (3aS,6aR)-5-benzyl-3a,6a-dimethyl-4,6-dihydrofuro[3,4- c]pyrrole-1,3-dione
To a solution of tert-butyl (3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c] pyrrole-5-carboxylate (0.5 g, 2.08 mmol, 1 eq) and (4-fluorophenyl)boronic acid (582.17 mg, 4.16 mmol, 2 eq) in dichloromethane (20 mL) were added copper acetate (755.73 mg, 4.16 mmol, 2 eq) and triethylamine (421.02 mg, 4.16 mmol, 579.12 µL, 2 eq), and the reaction mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 5/1 to 1/1 petroleum ether/ethyl acetate) to afford tert-butyl (3aS,6aR)-2-(4-fluorophenyl)- 3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.4 g, 1.20 mmol, 57% yield) as a yellow oil. LC/MS (ESI) m/z: 335.1 [M+H]+. Step 2: Preparation of (3aR,6aS)-5-(4-fluorophenyl)-3a,6a-dimethyl- 2,3,4,6- tetrahydro-1H-pyrrolo[3,4-c]pyrrole To a solution of tert-butyl (3aS,6aR)-2-(4-fluorophenyl)-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (100 mg, 0.30 mmol, 1 eq) in dichloromethane (3 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at 25 °C for 1 hour. The mixture was concentrated under reduced pressure to afford (3aR,6aS)-5-(4- fluorophenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c] pyrrole (100 mg, crude, trifluoroacetate) as a yellow oil.
Step 3: Preparation of 6-chloro-4-[2-(4-fluorophenyl)-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 89) To a solution of (3aR,6aS)-5-(4-fluorophenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H- pyrrolo[3,4-c]pyrrole (50 mg, 0.14 mmol, 1 eq, trifluoroacetate) and 4,6-dichloro-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile (36.47 mg, 143.54 µmol, 1 eq) in acetonitrile (3 mL) was added N,N-diisopropylethylamine (37.10 mg, 0.29 mmol, 50.00 µL, 2 eq), and the reaction mixture was stirred at 60 °C for 12 hours. The mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative High Performance Liquid Chromatography (column: Phenomenex Luna C18150*25 mm*10 um; mobile phase: [56-86% CH3CN in water (0.225 formic acid)]) to afford 6-chloro-4-[2-(4-fluorophenyl)- 3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (38.6 mg, 85.41 µmol, 60% yield) as a yellow solid. LC/MS (ESI) m/z: 452.2 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 7.99 (d, J = 9.2 Hz, 1H), 7.77 (d, J = 9.2 Hz, 1H), 7.01 (t, J = 8.8 Hz, 2H), 6.46 (dd, J = 4.4, 9.2 Hz, 2H), 4.32 (d, J = 12.8 Hz, 2H), 4.20 (d, J = 12.8 Hz, 2H), 3.48 (s, 3H), 3.45 (d, J = 10.0 Hz, 2H), 3.23 (d, J = 10.0 Hz, 2H), 1.17 (s, 6H). Synthesis of Compound 90 Preparation of 8-[(3aS,6aR)-2-(4-fluorophenyl)-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (Compound 90) To a solution of (3aR,6aS)-5-(4-fluorophenyl)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo [3,4-c]pyrrole (50 mg, 0.14 mmol, 1 eq, trifluoroacetate) and (6-cyano-1-methyl-2-oxo-1,5- naphthyridin-4-yl) trifluoromethanesulfonate (47.83 mg, 143.54 umol, 1 eq) in acetonitrile (3 mL) was added N,N-diisopropylethylamine (55.65 mg, 430.62 umol, 75.01 uL, 3 eq), and the reaction mixture was stirred at 60 °C for 12 hours. The mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative High Performance Liquid Chromatography (column: Phenomenex Luna C18150*25 mm*10 um; mobile phase: [54-84% CH3CN in water (0.225 formic acid)]) to afford 8-[(3aS,6aR)-2-(4-fluorophenyl)- 3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (22.9 mg, 54.85 umol, 38% yield) as an off-white solid. LC/MS (ESI) m/z: 418.2 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.15 (d, J = 8.8 Hz, 1H), 8.01 (d, J = 8.8 Hz, 1H), 7.00 (t, J = 8.8 Hz, 2H), 6.50 - 6.41 (m, 2H), 5.59 (s, 1H), 4.01 - 3.62 (m, 4H), 3.49 (s, 3H), 3.40 (d, J = 9.6 Hz, 2H), 3.23 (d, J = 9.6 Hz, 2H), 1.16 (s, 6H). Synthesis of Compound 91 Step 1: Preparation of 5-chloro-2-(4-piperidylamino)phenol To a stirred solution of tert-butyl 4-(4-chloro-2-hydroxy-anilino)piperidine-1-carboxylate (200 mg, 0.612 mmol, 1 eq) in CH2Cl2 (1 mL) was added 4 M HCl/dioxane (1 mL), and the reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 5-chloro-2-(4-piperidylamino)phenol (131 mg, 0.498 mmol, 81% yield, HCl) as a black solid. LC/MS (ESI) m/z: 227.2 [M+H]+. Step 2: Preparation of 6-chloro-4-[4-(4-chloro-2-hydroxy-anilino)-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 91) To a stirred solution of 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (100 mg, 0.394 mmol, 1 eq) in CH3CN (3 mL) were added diisopropylethylamine (153 mg, 1.18 mmol, 3 eq) and 5-chloro-2-(4-piperidylamino)phenol (103.58 mg, 0.394 mmol, 1 eq, HCl), and the reaction mixture was stirred at 60 °C for 0.5 hour. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The resulting crude product was purified by prep-HPLC (column: Phenomenex luna 30*30 mm*10 um+YMC AQ 100*30*10 um; mobile phase: [30-80% CH3CN in water (NH4HCO3)]). Pure fractions were combined and dried by lyophilization to give 6-chloro-4-[4-(4-chloro-2-hydroxy-anilino)-1-piperidyl]- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (27.5 mg, 0.060 mmol, 15% yield) as a yellow solid. LC/MS (ESI) m/z: 444.0 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 9.78 (s, 1H), 8.07 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 6.72-6.59 (m, 3H), 4.56 (d, J = 8.8 Hz, 1H), 4.26-4.16 (m, 2H), 3.72-3.50 (m, 6H), 2.17-2.03 (m, 2H), 1.79-1.62 (m, 2H). Synthesis of Compound 92 Step 1: Preparation of tert-butyl 4-[(4-fluorophenyl)methyl-methyl-amino]piperidine- 1-carboxylate To a solution of tert-butyl 4-(methylamino)piperidine-1-carboxylate (500 mg, 2.33 mmol) in CH3OH (10 mL) were added 4-fluorobenzaldehyde (289 mg, 2.33 mmol, 245 µL), AcOH (1.40 g, 23 mmol, 1.33 mL) and borane 2-methylpyridine (1.25 g, 11. mmol), and the reaction mixture was stirred at 20 °C for 10 hours The reaction mixture was concentrated, and the crude product was purified by flash silica gel chromatography (gradient: 0~40 % ethyl acetate in petroleum ether). Pure fraction was lyophilized to give tert-butyl 4-[(4- fluorophenyl)methyl-methyl-amino]piperidine-1-carboxylate (350 mg, 1.09 mmol, 47% yield) as colorless oil. LC/MS (ESI) m/z: 323.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.40- 7.27 (m, 2H), 7.02 (d, J = 8.4 Hz, 2H), 4.32-4.04 (m, 2H), 3.74-3.44 (m, 2H), 2.81-2.54 (m, 3H), 2.24-2.18 (m, 2H), 1.90-1.74 (m, 2H), 1.66 (s, 3H), 1.47 (s, 9H). Step 2: Preparation of N-[(4-fluorophenyl)methyl]-N-methyl-piperidin-4-amine A solution of tert-butyl 4-[(4-fluorophenyl)methyl-methyl-amino] piperidine-1-carboxylate (120 mg, 0.372 mmol) in HCl/dioxane (5 mL, 4 M) was stirred at 20°C for 1 hour. The reaction was concentrated to give N-[(4-fluorophenyl)methyl]-N-methyl-piperidin-4-amine (96 mg, crude, HCl) as a white solid. LC/MS (ESI) m/z: 223.3 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-[(4-fluorophenyl)methyl-methyl-amino]-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 92) To a solution of N-[(4-fluorophenyl)methyl]-N-methyl-piperidin-4-amine (96 mg, 0.371 mmol, HCl) in CH3CN (3 mL) were added 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (94 mg, 0.371 mmol) and diisopropylethylamine (239 mg, 1.85 mmol, 323.0 µL), and the reaction mixture was stirred at 50°C for 1 hour. The reaction mixture was concentrated, and the crude product was purified by prep-HPLC (column: Xtimate C18 100*30 mm * 10 µm; mobile phase: [15-45% CH3CN in water (0.225% formic acid)]). Pure fraction was lyophilized to give 6-chloro-4-[4-[(4-fluorophenyl)methyl-methyl-amino]-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (80.6 mg, 0.178 mmol, 48% yield) as a yellow solid. LC/MS (ESI) m/z: 440.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.37-7.34 (m, 2H), 7.15-7.11(t, J = 8.8 Hz, 2H), 4.27 (d, J = 12.8 Hz, 2H), 3.58 (s, 2H), 3.52 (s, 3H), 3.41 (s, 2H), 2.87-2.75 (m, 1H), 2.12 (s, 3H), 1.99-1.89 (m, 2H), 1.80-1.77 (m, 2H). Synthesis of Compound 96 Step 1: Preparation of tert-butyl (3aR,6aS)-2-(6-chloro-3-cyano-1-methyl-2-oxo-1,5- naphthyridin-4-yl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl (3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c] pyrrole-5-carboxylate (0.1 g, 0.42 mmol, 1 eq) and 4,6-dichloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (105.71 mg, 416.08 umol, 1 eq) in acetonitrile (5 mL) was added diisopropylethylamine (161.32 mg, 1.25 mmol, 217.42 µL, 3 eq), and the mixture was stirred at 80 °C for 4 hours. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (gradient: 10/1 to 1/1petroleum ether/ethyl acetate) to afford tert-butyl (3aR,6aS)-2-(6-chloro-3-cyano-1-methyl-2-oxo-1,5-naphthyridin- 4-yl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.1 g, 218.36 µmol, 52% yield) as a light yellow solid. LC/MS (ESI) m/z: 480.0 [M+Na]+. Step 2: Preparation of 4-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H- pyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a solution of tert-butyl (3aR,6aS)-2-(6-chloro-3-cyano-1-methyl-2-oxo-1,5-naphthyridin- 4-yl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (30 mg, 65.51 µmol, 1 eq) in dichloromethane (3 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at 25 °C for 1 hour. The mixture was concentrated under reduced pressure to afford 4-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6- tetrahydro-1H-pyrrolo[3,4-c]pyrrol- 5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (30 mg, crude, trifluoroacetate) as a light yellow oil.
Step 3: Preparation of 4-[(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 96) To a solution of 4-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl] -6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (30 mg, 63.58 µmol, 1 eq, trifluoroacetate) in methanol (3 mL) was added sodium acetate (26.08 mg, 0.32 mmol, 5 eq), and the mixture was stirred at 25 °C for 0.5 hour.4-Chloro-2-hydroxy-benzaldehyde (9.95 mg, 63.58 µmol, 1 eq) and sodium cyanoborohydride (5.99 mg, 95.37 µmol, 1.5 eq) were then added, and the reaction mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative High Performance Liquid Chromatography (column: Phenomenex Synergi C18150*25 mm* 10 µm; mobile phase: [7-40% CH3CN in water (0.225% formic acid)]) to afford 4- [(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (26.2 mg, 51.71 µmol, 81% yield) as an off-white solid. LC/MS (ESI) m/z: 498.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.41 (brs, 1H), 8.04 (d, J = 9.2 Hz, 1H), 7.78 (d, J = 9.2 Hz, 1H), 7.03 (d, J = 8.0 Hz, 1H), 6.68 (dd, J = 2.0, 8.0 Hz, 1H), 6.61 (d, J = 2.0 Hz, 1H), 4.54 (d, J = 12.4 Hz, 2H), 3.87 (d, J = 12.4 Hz, 2H), 3.58 (s, 2H), 3.51 (s, 3H), 2.94 (d, J = 9.6 Hz, 2H), 2.39 (d, J = 9.6 Hz, 2H), 1.10 (s, 6H). Synthesis of Compound 97 Step 1: Preparation of dimethyl 1-benzylpyrrolidine-3,4-dicarboxylate To a stirred solution of dimethyl (E)-but-2-enedioate (30 g, 208.15 mmol, 1.0 eq) and trifluoroacetic acid (11.87 g, 104.08 mmol, 7.71 mL, 0.5 eq) in dichloromethane (500 mL) at 0 °C was added dropwise a solution of N-(methoxymethyl)-1-phenyl-N- (trimethylsilylmethyl)methanamine (58.14 g, 208.15 mmol, 85% purity, 1.0 eq) in dichloromethane (50 mL) over 30 minutes. After completion of addition, the cold bath was removed, and the reaction mixture was stirred at 20 °C for 14.5 h. The reaction mixture was diluted with dichloromethane (50 mL), then washed with a saturated solution of sodium bicarbonate (20 mL x 2). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (gradient: 100/0 to 10/1 petroleum ether/ethyl acetate) to afford dimethyl 1- benzylpyrrolidine-3,4-dicarboxylate (45 g, 162.27 mmol, 78% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 7.38 - 7.28 (m, 5H), 3.73 - 3.69 (m, 6H), 3.64 - 3.59 (m, 2H), 3.50 - 3.43 (m, 2H), 2.95 - 2.87 (m, 2H), 2.80 (dd, J = 5.8, 9.4 Hz, 2H). Step 2: Preparation of O1-tert-butyl O3,O4-dimethyl pyrrolidine-1,3,4-tricarboxylate To a stirred solution of dimethyl 1-benzylpyrrolidine-3,4-dicarboxylate (15.0 g, 54.09 mmol, 1.0 eq) and di-tert-butyl dicarbonate (17.71 g, 81.14 mmol, 18.64 mL, 1.5 eq) in ethanol (150 mL) was added palladium on activated carbon catalyst (1.5 g, 10% purity) under nitrogen. The reaction mixture was degassed with hydrogen (3X) and stirred at 20 °C for 15 h under hydrogen (15 psi). The reaction mixture was degassed with hydrogen (3X) and stirred at 20 °C for 35 h under hydrogen (15 psi). The reaction mixture was filtered, and the cake was washed with ethanol (20 mL x 2). The resulting filtrate was concentrated under reduced pressure to afford O1-tert-butyl O3,O4-dimethyl pyrrolidine-1,3,4-tricarboxylate (13 g, 45.25 mmol, 83.65% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 3.90 - 3.64 (m, 8H), 3.62 - 3.28 (m, 4H), 1.53 - 1.39 (m, 9H). Step 3: Preparation of tert-butyl 3,4-bis(hydroxymethyl)pyrrolidine-1-carboxylate To a solution of O1-tert-butyl O3,O4-dimethyl pyrrolidine-1,3,4-tricarboxylate (13 g, 45.25 mmol, 1 eq) in tetrahydrofuran (300 mL) was added lithium borohydride (11.91 g, 546.74 mmol, 12.08 eq) over 30 minutes, and the reaction mixture was stirred at 20 °C for 15 hours under nitrogen. To the reaction mixture was added dropwise water (100 mL) and methanol (200 mL) over 30 minutes, then stirred at 20 °C for 1 h. The precipitate was filtered, and the cake was washed with THF (100 mL x 2) and ethyl acetate (100 mL x 2). The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (gradient: petroleum ether/ethyl acetate=10/1 to ethyl acetate/methanol = 13/1) to afford tert-butyl 3,4-bis(hydroxymethyl)pyrrolidine-1- carboxylate (10.0 g, 43.24 mmol, 95.55% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 5.24 - 4.75 (m, 1H), 3.82 - 3.69 (m, 2H), 3.64 - 3.46 (m, 4H), 3.08 - 2.93 (m, 2H), 2.27 - 2.12 (m, 2H), 1.50 - 1.38 (m, 9H). Step 4: Preparation of tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1- carboxylate To a solution of tert-butyl 3,4-bis(hydroxymethyl)pyrrolidine-1-carboxylate (10.0 g, 43.24 mmol, 1 eq) and TEA (26.25 g, 259.42 mmol, 36.11 mL, 6.0 eq) in dichloromethane (300 mL) was added a solution of methanesulfonyl chloride (17.79 g, 155.30 mmol, 12.02 mL, 3.59 eq) in dichloromethane (10 mL) at 0 °C for 10 minutes. The reaction mixture was then stirred at 20 °C for 15 hours. The reaction mixture was treated with saturated aqueous ammonium chloride solution (100 mL) and extracted with ethyl acetate (200 mL x 2). The organic extract was washed with saturated aqueous NaHCO3 solution (100 mL), brine (100 mL x 3), dried over sodium sulfate, and concentrated under reduced pressure to afford tert- butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (16.0 g, 41.29 mmol, 96% yield) as a yellow oil.1H NMR (400 MHz, CDCl3) δ 4.36 - 4.19 (m, 4H), 3.70 - 3.59 (m, 2H), 3.31 - 3.19 (m, 2H), 3.09 - 3.02 (m, 6H), 2.63 - 2.43 (m, 2H), 1.49 - 1.44 (m, 9H). Step 5: Preparation of tert-butyl 2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl 3,4-bis(methylsulfonyloxymethyl)pyrrolidine-1-carboxylate (0.33 g, 0.85 mmol, 1.0 eq) and potassium carbonate (1.18 g, 8.52 mmol, 0.64 mL, 10.0 eq) in acetonitrile (10 mL) was added (4-fluorophenyl)methanamine (319 mg, 2.56 mmol, 0.29 mL, 3.0 eq), and the reaction mixture was stirred at 100 °C for 15 h. The reaction mixture was cooled to room temperature and partitioned between ethyl acetate (50 mL) and water (10 mL). The organic layer was washed with brine (10 mL), dried over sodium sulfate, filtered, and concentrated. The resulting residue was purified by prep-TLC (SiO2, petroleum ether/ethyl acetate=2/3) to afford tert-butyl 2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.13 g, 405.74 µmol, 48% yield) as a white solid. LC/MS (ESI) m/z: 321.4 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.40 - 7.28 (m, 2H), 7.08 - 6.97 (m, 2H), 3.94 - 3.81 (m, 2H), 3.65 - 3.58 (m, 1H), 3.56 - 3.49 (m, 1H), 3.03 - 2.86 (m, 4H), 2.71 - 2.52 (m, 2H), 2.35 - 2.17 (m, 2H), 1.46 (s, 9H). Step 6: Preparation of 5-[(4-fluorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H- pyrrolo[3,4-c]pyrrole
To a solution of tert-butyl 2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (60 mg, 0.19 mmol, 1 eq) in dichloromethane (5 mL) and methanol (2 mL) was added hydrochloric acid/dioxane (4 M, 10 mL), and the reaction mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated to afford 5-[(4- fluorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole (48 mg, 186.96 umol, 99.83% yield, hydrochloride) as a yellow solid. LC/MS (ESI) m/z: 221.3 [M+H] +. Step 7: Preparation of 8-[(3aR,6aR)-2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (Compound 97) To a solution of 5-[(4-fluorophenyl)methyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole (48 mg, 0.19 µmol, 1 eq, hydrochloride) and N,N-diisopropylethylamine (120 mg, 0.93 mmol, 0.16 mL, 5.0 eq) in dichloromethane (5 mL) and DMF (1 mL) was added (6- cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (62 mg, 0.19 mmol, 1 eq), and the reaction mixture was stirred at 20 °C for 8 h. Brine (10 mL) was added, and the resulting mixture was stirred for 10 minutes, then extracted with ethyl acetate (50 mL x 2). The combined organic extract was washed with brine (10 mL x 2), dried with anhydrous sodium sulfate, filtered, and concentrated in vacuum. The residue was purified by prep-TLC (dichloromethane/ethyl acetate/methanol=3/7/1) to afford 8-[(3aR,6aR)-2-[(4- fluorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (29.1 mg, 72.13 µmol, 39% yield) as an off-white solid. LC/MS (ESI) m/z: 404.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.81 - 7.74 (m, 1H), 7.69 - 7.61 (m, 1H), 7.50 - 7.38 (m, 2H), 7.13 - 7.03 (m, 2H), 5.81 - 5.73 (m, 1H), 4.07 - 4.00 (m, 2H), 3.99 - 3.77 (m, 2H), 3.63 - 3.59 (m, 3H), 3.28 - 3.16 (m, 2H), 3.00 - 2.85 (m, 2H), 2.55 - 2.42 (m, 2H), 1.51 - 1.39 (m, 2H). Synthesis of Compounds 98 and 88 Step 1: Preparation of tert-butyl 4-(4-chloro-2-hydroxy-anilino)-3-methyl-piperidine- 1-carboxylate To a solution of 2-amino-5-chloro-phenol (2 g, 13.93 mmol, 1 eq) and tert-butyl 3-methyl-4- oxo-piperidine-1-carboxylate (2.97 g, 13.93 mmol, 1 eq) in methanol (20 mL) was added acetic acid (83.65 mg, 1.39 mmol, 79.67 µL, 0.1 eq), and the resulting mixture was stirred at 50 °C for 12 hours. Sodium cyanoborohydride (1.75 g, 27.86 mmol, 2 eq) was then added, and the reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (gradient: 1/0 to 6/1 petroleum ether/ethyl acetate) to afford tert- butyl 4-(4-chloro-2-hydroxy-anilino)-3-methyl-piperidine-1- carboxylate (3.89 g, 11.40 mmol, 82% yield) as a brown oil. LC/MS (ESI) m/z: 341.0 [M+H]+. Step 2: Preparation of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl- piperidine-1-carboxylate To a solution of tert-butyl 4-(4-chloro-2-hydroxy-anilino)-3-methyl-piperidine-1-carboxylate (3 g, 8.80 mmol, 1 eq) in methanol (10 mL) were added paraformaldehyde (2.64 g, 88.02 mmol, 2.42 mL, 10 eq) and sodium cyanoborohydride (1.11 g, 17.60 mmol, 2 eq), and the reaction mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (gradient: 1/0 to 25/1 petroleum ether/ethyl acetate) to afford tert-butyl 4-(4- chloro-2-hydroxy-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate (3 g, 8.45 mmol, 96% yield) as a brown oil. LC/MS (ESI) m/z: 355.0 [M+H]+. Step 3: Preparation of 5-chloro-2-[methyl-(3-methyl-4-piperidyl)amino]phenol To a solution of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-piperidine-1- carboxylate (200 mg, 0.56 mmol, 1 eq) in dichloromethane (5 mL) was added HCl in dioxane (4 M, 5 mL), and the reaction mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give 5-chloro-2-[methyl-(3-methyl-4- piperidyl)amino]phenol (160 mg, 0.55 mmol, 97% yield, HCl salt) as a brown solid. Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-N-methyl-anilino)- 3-methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 98) and 6-chloro-4-[(3R,4R)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 88) To a solution of 5-chloro-2-[methyl-(3-methyl-4-piperidyl)amino]phenol (160 mg, 0.55 mmol, 1 eq, HCl salt) in propan-1-ol (5 mL) was added 4,6-dichloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (139.59 mg, 0.55 mmol, 1 eq), N,N-diisopropylethylamine (213.03 mg, 1.65 mmol, 287.10 µL, 3 eq), and the reaction mixture was stirred at 80 °C for 0.5 h. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative High Performance Liquid Chromatography (column: Phenomenex Synergi C18150*25 mm* 10 um; mobile phase: [37-73% CH3CN in water (0.225% formic acid)]) to afford the desired products. 6-Chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (42.2 mg, 81.74 µmol, 15% yield) (Compound 98) was obtained as a yellow solid. LC/MS (ESI) m/z: 472.1 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 8.8 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 7.19 (d, J = 8.4 Hz, 1H), 6.93 - 6.72 (m, 2H), 4.29 (d, J = 10.4 Hz, 1H), 4.11 - 3.99 (m, 1H), 3.67 - 3.59 (m, 1H), 3.52 (s, 3H), 3.30 - 3.20 (m, 2H), 2.54 (s, 3H), 2.20 - 1.99 (m, 2H), 1.70 - 1.62 (m, 1H), 0.85 (d, J = 6.8 Hz, 3H). 6-Chloro-4-[(3R,4R)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (36.8 mg, 75.88 µmol, 14% yield) (Compound 88) was obtained as a yellow solid. LC/MS (ESI) m/z: 472.1 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 9.89 (brs, 1H), 8.07 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.8 Hz, 1H), 6.80 (d, J = 2.4 Hz, 1H), 6.77 - 6.65 (m, 1H), 4.37 - 4.16 (m, 2H), 3.53 (s, 3H), 3.46 -3.42 (m, 2H), 3.08 - 2.97 (m, 1H), 2.63 (s, 3H), 2.30 - 2.15 (m, 1H), 2.02 - 1.91 (m, 1H), 1.80 - 1.72 (m, 1H), 0.91 (d, J = 6.4 Hz, 3H). Synthesis of Compound 99 Preparation of 4-[(3aS,6aR)-2-[(4-chlorophenyl)methyl]-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (Compound 99) The title compound was prepared in an analogous manner to 4-[(3aS,6aR)-2-[(4-chloro-2- hydroxy-phenyl)methyl]-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-6- chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from 4-[(3aS,6aR)-3a,6a- dimethyl-2,3,4,6- tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile. (off-white solid). LC/MS (ESI) m/z: 482.1 [M+H]+.1HNMR (400 MHz, DMSO-d6) δ 8.02 (d, J = 8.8 Hz, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.27 - 7.21 (m, 2H), 7.21 - 7.14 (m, 2H), 4.51 (d, J = 12.0 Hz, 2H), 3.91 (d, J = 12.0 Hz, 2H), 3.51 (s, 3H), 3.50 (s, 2H), 2.84 (d, J = 9.2 Hz, 2H), 2.29 (d, J = 9.2 Hz, 2H), 1.08 (s, 6H). Synthesis of Compounds 100 and 59 Preparation of 6-chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3- methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6-chloro-4- [(3R,4S)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-1-piperidyl]-1-methyl-2- oxo-1,5-naphthyridine-3-carbonitrile The enantiomers were separated by SFC (Column: Daicel ChiralPak IG (250*30mm, 10um); Mobile phase: 60% isopropanol (0.1% NH4OH) in supercritical CO2). The absolute configuration was arbitrarily assigned. 6-Chloro-4-[(3S,4R)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (17.6 mg, 0.03 mmol, 26% yield) (Compound 100) was obtained as a yellow solid. LC/MS (ESI) m/z: 472.1 [M+H] +.1H NMR (400 MHz, CD3OD) δ 8.01 (d, J = 9.2 Hz, 1H), 7.68 (d, J = 9.2 Hz, 1H), 7.25 (d, J = 8.4 Hz, 1H), 6.93 - 6.79 (m, 2H), 4.61 - 4.57 (m, 1H), 4.51 - 4.40 (m, 1H), 4.21 (m, 1H), 3.73 (dd, J = 2.4, 13.2 Hz, 1H), 3.62 (s, 3H), 3.37 - 3.34 (m, 1H), 2.62 (s, 3H), 2.33 - 2.24 (m, 1H), 2.23 - 2.14 (m, 1H), 1.74 - 1.63 (m, 1H), 0.98 (d, J = 7.2 Hz, 3H). 6-Chloro-4-[(3R,4S)-4-(4-chloro-2-hydroxy-N-methyl-anilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (24.8 mg, 0.05 mol, 38% yield) (Compound 59) was obtained as a yellow solid. LC/MS (ESI) m/z: 472.1 [M+H] +.1H NMR (400 MHz, CD3OD) δ 8.01 (d, J = 9.2 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 7.26 (d, J = 8.4 Hz, 1H), 6.91 - 6.81 (m, 2H), 4.63 - 4.55 (m, 1H), 4.49 - 4.40 (m, 1H), 4.20 (m, 1H), 3.73 (dd, J = 2.4, 13.2 Hz, 1H), 3.62 (s, 3H), 3.34 (d, J = 2.4 Hz, 1H), 2.63 (s, 3H), 2.32 - 2.24 (m, 1H), 2.24 - 2.15 (m, 1H), 1.74 - 1.65 (m, 1H), 0.98 (d, J = 6.8 Hz, 3H). Synthesis of Compound 101 Step 1: Preparation of tert-butyl (3aR,6aS)-2-(6-cyano-1-methyl-2-oxo-1,5- naphthyridin-4-yl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5- carboxylate To a solution of tert-butyl (3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4- c]pyrrole-5-carboxylate (100 mg, 0.42 mmol, 1 eq) and (6-cyano-1-methyl-2-oxo-1,5- naphthyridin-4-yl) trifluoromethanesulfonate (138.65 mg, 0.42 mmol, 1 eq) in CH3CN (5 mL) was added N,N-diisopropylethylamine (107.55 mg, 0.83 mmol, 144.95 µL, 2 eq), and the reaction mixture was stirred at 80 °C for 12 hours. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (3/1 to 0/1 petroleum ether/ethyl acetate) to afford tert-butyl (3aR,6aS)-2-(6-cyano-1-methyl-2-oxo- 1,5-naphthyridin-4-yl)-3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (150 mg, 354.19 µmol, 85% yield) was as a colorless oil. LC/MS (ESI) m/z: 260.1 [M+H]+. Step 2: 8-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile To a solution of tert-butyl (3aR,6aS)-2-(6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl)- 3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrole-5-carboxylate (30 mg, 0.07 mmol, 1 eq) in dichloromethane (5 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 190.66 eq), and the reaction mixture was stirred at 20 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give crude 8-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6- tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (30 mg).
Step 3: Preparation of 8-[(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]-3a,6a- dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (Compound 101) To a solution of 8-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (30 mg, 0.07 mmol, 1 eq, trifluoroacetic acid) and 4-chloro-2-hydroxy-benzaldehyde (16.11 mg, 0.1 mmol, 1.5 eq) in CH3OH (5 mL) were added sodium acetate (16.88 mg, 0.21 mmol, 3 eq), acetic acid (411.87 µg, 6.86 µmol, 3.92e-1 µL, 0.1 eq), and sodium triacetoxyborohydride (29.07 mg, 0.14 mmol, 2 eq), and the reaction mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC [column: Phenomenex Synergi C18150*25mm* 10um; mobile phase: (7-37% CH3CN in water (0.225% formic acid))] to afford 8-[(3aS,6aR)-2-[(4-chloro-2-hydroxy-phenyl)methyl]- 3a,6a-dimethyl-1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (10.3 mg, 22.14 µmol, 32 % yield) as a white solid. LC/MS (ESI) m/z: 464.0 [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.04 (s, 1H), 7.83 - 7.79 (m, 1H), 7.70 (d, J = 8.8 Hz, 1H), 7.09 - 7.00 (m, 1H), 6.93 - 6.87 (m, 1H), 6.81 (br d, J = 8.0 Hz, 1H), 5.86 (s, 1H), 4.16 (br d, J = 11.6 Hz, 2H), 4.06 - 3.94 (m, 2H), 3.63 (s, 3H), 3.52 - 3.43 (m, 2H), 3.26 - 3.12 (m, 2H), 3.09 - 2.86 (m, 2H), 1.25 (s, 6H). Synthesis of Compound 102 Preparation of 8-[(3aS,6aR)-2-[(4-fluorophenyl)methyl]-3a,6a-dimethyl-1,3,4,6- tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile To a solution of 8-[(3aS,6aR)-3a,6a-dimethyl-2,3,4,6-tetrahydro-1H-pyrrolo[3,4-c]pyrrol-5- yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (60 mg, 0.14 mmol, 1 eq, trifluoroacetic acid) and 4-fluorobenzaldehyde (25.54 mg, 0.21 mmol, 21.64 µL, 1.5 eq) in CH3OH (5 mL) were added sodium acetate (33.76 mg, 0.41 mmol, 3 eq), acetic acid (823.73 ug, 13.72 µmol, 7.85e-1 µL, 0.1 eq), and sodium triacetoxyborohydride (58.14 mg, 0.27 mmol, 2 eq), and the reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC [column: Phenomenex Synergi C18150*25mm* 10µm; mobile phase: (4-37% CH3CN in water (0.225% formic acid))] to afford 8-[(3aS,6aR)-2-[(4-fluorophenyl)methyl]-3a,6a-dimethyl- 1,3,4,6-tetrahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (29.9 mg, 68.30 µmol, 50 % yield) as a white solid. LC/MS (ESI) m/z: 432.2 [M+H]+.1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.80 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 7.41 (br t, J = 6.4 Hz, 2H), 7.05 (br t, J = 8.0 Hz, 2H), 5.84 (s, 1H), 4.09 (br d, J = 11.6 Hz, 2H), 3.91 (s, 2H), 3.62 (s, 3H), 3.52 (br d, J = 11.6 Hz, 2H), 3.11 (br d, J = 10.4 Hz, 2H), 2.90 (br d, J = 10.4 Hz, 2H), 1.22 (s, 6H). Synthesis of Compound 103 Step 1: Preparation of tert-butyl 4-(4-chloro-2-hydroxy-anilino)piperidine-1- carboxylate To a solution of 2-amino-5-chloro-phenol (1 g, 6.97 mmol, 1 eq) in dichloromethane (10 mL) was added tert-butyl 4-oxopiperidine-1-carboxylate (1.39 g, 6.97 mmol, 1 eq), and the reaction mixture was stirred at 20 °C for 11 hours. Sodium triacetoxyborohydride (2.95 g, 13.93 mmol, 2 eq) was then added and the mixture was stirred at 20 °C for 1 hour. The reaction mixture was diluted with water (10 mL) and extracted with dichloromethane (10 mL x 3). The combined organic extracts were washed with brine (10 mL x 2), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by silica gel column chromatography (gradient: 30/1 to 8/1 petroleum ether/ethyl acetate) to give tert-butyl 4-(4-chloro-2-hydroxy-anilino)piperidine-1-carboxylate (1.8 g, 5.51 mmol, 79% yield) as black brown solid. LC/MS (ESI) m/z: 271.1 [M-C4H9+H]+.1H NMR (400 MHz, CDCl3) δ 6.80 (m, 2H), 6.63 (d, J = 8.8 Hz, 1H), 4.11 - 3.98 (m, 2H), 3.34 (s, 1H), 2.91 (t, J = 11.6 Hz, 2H), 1.99 (d, J = 12.0 Hz, 2H), 1.48 (s, 9H), 1.44 - 1.24 (m, 3H). Step 2: Preparation of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)piperidine- 1-carboxylate To a solution of tert-butyl 4-(4-chloro-2-hydroxy-anilino)piperidine-1- carboxylate (300 mg, 0.92 mmol, 1 eq) in dichloromethane (3 mL) was added paraformaldehyde (138 mg, 4.59 mmol, 5 eq) and the mixture was stirred at 20 °C for 11 hour. Sodium triacetoxyborohydride (389 mg, 1.84 mmol, 2 eq) was then added, and the reaction mixture was stirred at 20 °C for 1 hour. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (20 mL x 3). The combined organic extracts were washed with brine (15 mL x2), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by prep-TLC (SiO2, petroleum ether/ethyl acetate = 3/1) to give tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)piperidine-1- carboxylate (200 mg, 0.59 mmol, 64% yield) as brown oil. LC/MS (ESI) m/z: 341.2 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 7.16 - 6.96 (m, 2H), 6.88 - 6.82 (m, 1H), 4.16 - 4.01 (m, 2H), 2.99 - 2.82 (m, 1H), 2.77 - 2.61 (m, 5H), 1.91 - 1.72 (m, 3H), 1.45 (s, 9H), 1.42 - 1.33 (m, 2H). Step 3: Preparation of 5-chloro-2-[methyl(4-piperidyl)amino]phenol To a solution of tert-butyl 4-(4-chloro-2-hydroxy-N-methyl-anilino)piperidine-1-carboxylate (100 mg, 0.29 mmol, 1 eq) in dichloromethane (2 mL) was added trifluoroacetic acid (1 mL), and the reaction mixture was stirred at 20 °C for 0.5 hour. The mixture was concentrated under reduced pressure to give 5-chloro-2-[methyl(4-piperidyl)amino]phenol (70 mg, crude) as a brown oil. LC/MS (ESI) m/z: 241.2 [M+H] +. Step 4: Preparation of 6-chloro-4-[4-(4-chloro-2-hydroxy-N-methyl-anilino)-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a solution of 5-chloro-2-[methyl(4-piperidyl)amino]phenol (70 mg, 0.29 mmol, 1.23 eq) in DMF (2 mL) were added triethylamine (96 mg, 0.94 mmol, 4 eq) and 4,6-dichloro-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (60 mg, 0.24 mmol, 1 eq), and the reaction mixture was stirred at 20 °C for 1 hour. The reaction mixture was diluted with water (10 mL) and extracted with dichloromethane (5 mL x 3). The combined organic extracts were washed with brine (5 mL x2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC [column: Unisil 3-100 C18 Ultra 150 x 50mm x 3 µm; mobile phase: (23-53% CH3CN in water (0.225% formic acid))] to give 6-chloro-4-[4-(4-chloro-2-hydroxy-N-methyl-anilino)-1-piperidyl]-1-methyl- 2-oxo-1,5-naphthyridine-3-carbonitrile (57.7 mg, 0.12 mmol, 52% yield) as yellow solid. LC/MS (ESI) m/z: 458.1 [M+H] +.1H NMR (400 MHz, DMSO-d6) δ 9.32 (s, 1H), 8.05 (d, J = 9.2 Hz, 1H), 7.79 (d, J = 8.8 Hz, 1H), 7.04 (d, J = 8.4 Hz, 1H), 6.88 - 6.73 (m, 2H), 4.23 (d, J = 12.4 Hz, 2H), 3.51 (s, 3H), 3.39 (m, 3H), 2.60 (s, 3H), 1.92 - 1.75 (m, 4H). Synthesis of Compound 104 Step 1: Preparation of tert-butyl 4-[(5-chloro-2-pyridyl)-methyl-amino]piperidine-1- carboxylate To a solution of 5-chloro-2-fluoro-pyridine (300 mg, 2.28 mmol, 1 eq) and tert-butyl 4- (methylamino)piperidine-1-carboxylate (500 mg, 2.33 mmol, 1.02 eq) in dimethylsulfoxide (5 mL) was added diisopropylethylamine (300 mg, 2.32 mmol, 1.02 eq), and the reaction mixture was stirred at 100 °C for 12 h. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (5 mL x 3). The combined organic extracts were washed with brine (5 mL x 3), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by prep-TLC (SiO2, petroleum ether: ethyl acetate = 3: 1) to afford tert-butyl 4-[(5-chloro-2-pyridyl)-methyl-amino]piperidine-1-carboxylate (120 mg, 0.37 mmol, 16% yield) as a colorless oil. LC/MS (ESI) m/z: 356.3 [M+H]+. Step 2: Preparation of 5-chloro-N-methyl-N-(4-piperidyl)pyridin-2-amine To a solution of tert-butyl 4-[(5-chloro-2-pyridyl)-methyl-amino]piperidine-1-carboxylate (120 mg, 0.37 mmol, 1 eq) in dichloromethane (2 mL) was added trifluoroacetic acid (1.54 g, 13.51 mmol, 36.67 eq), and the reaction mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated under reduced pressure to afford 5-chloro-N-methyl-N-(4- piperidyl)pyridin-2-amine (120 mg, 0.35 mmol, 96% yield, TFA salt) as a yellow solid. Step 3: Preparation of 6-chloro-4-[4-[(5-chloro-2-pyridyl)-methyl-amino]-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 104) To a solution of 5-chloro-N-methyl-N-(4-piperidyl)pyridin-2-amine (120 mg, 0.35 mmol, 1.13 eq, trifluoroacetate) in DMF (3 mL) were added triethylamine (130 mg, 1.28 mmol, 4.09 eq) and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (80 mg, 0.31 mmol, 1 eq), and the reaction mixture was stirred at 25 °C for 1 h. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (5 mL x 3). The combined organic extract was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by prep-HPLC [column: Unisil 3-100 C18 Ultra 150*50mm*3 um; mobile phase: (45-75% CH3CN in water (0.225% formic acid))] to afford 6-chloro-4-[4-[(5-chloro-2-pyridyl)-methyl-amino]-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (59.2 mg, 0.13 mmol, 42% yield) as a yellow solid. LC/MS (ESI) m/z: 443.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.13 - 8.05 (m, 2H), 7.82 (d, J = 8.8 Hz, 1H), 7.58 (d, J = 9.2 Hz, 1H), 6.78 (d, J = 8.8 Hz, 1H), 4.74 (t, J = 10.8 Hz, 1H), 4.33 (d, J = 12.4 Hz, 2H), 3.58 - 3.49 (m, 5H), 2.86 (s, 3H), 2.11 - 2.00 (m, 2H), 1.78 (d, J = 10.4 Hz, 2H). Synthesis of Compound 105 Step 1: Preparation of tert-butyl 2-[(4-chloro-2-hydroxy-phenyl) methyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate A mixture of 4-chloro-2-hydroxy-benzaldehyde (200 mg, 1.28 mmol), tert-butyl 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5-carboxylate (272 mg, 1.28 mmol), and triethylamine (129 mg, 1.28 mmol) in 1,2-dichloroethane (5 mL) was stirred at 20 °C for 1 h. Sodium cyanoborohydride (161 mg, 2.56 mmol) was then added, and the reaction mixture was stirred for 1 h. The reaction was quenched with water (30 mL) and extracted with dichloromethane (15 mL x 3). The combined organic extract was dried over sodium sulfate, filtered, concentrated in vacuum. Purification by prep-HPLC column: [Phenomenex luna C18 150*40mm* 15um; mobile phase: (8-38% CH3CN in water (0.225% formic acid))] afforded tert-butyl 2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (230 mg, 0.65 mmol, 50% yield) as a pink solid. LC/MS (ESI) m/z: 353.2 [M+H] +. Step 2: Preparation of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo [3,4-c]pyrrol-5- ylmethyl)-5-chloro-phenol To a solution of tert-butyl 2-[(4-chloro-2-hydroxy-phenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (200 mg, 0.57 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (323 mg, 2.83 mmol, 0.21 mL), and the reaction mixture was stirred at 20 °C for 1 h. The mixture was concentrated to afford crude 2-(2,3,3a,4,6,6a- hexahydro-1H-pyrrolo [3,4-c]pyrrol-5-ylmethyl)-5-chloro-phenol (200 mg, 0.55 mmol, TFA salt) as a pale-yellow oil. Step 3: Preparation of 6-chloro-4-[2-[(4-chloro-2-hydroxy-phenyl)methyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (Compound 105) To a solution of 2-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-ylmethyl)-5-chloro- phenol (150 mg, 0.41 mmol, 1.04 eq, TFA salt) in DMF (3 mL) were added triethylamine (159 mg, 1.57 mmol, 0.22 mL, 4 eq) and 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (100 mg, 0.39 mmol, 1 eq), and the reaction mixture was stirred at 25 °C for 1 h. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (5 mL x 3). The combined organic extract was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by prep-HPLC [column: Phenomenex luna C18150*25mm* 10 µm; mobile phase: (10-40% CH3CN in water (0.225% formic acid))] to afford 6-chloro-4-[2-[(4-chloro-2- hydroxy-phenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (43.9 mg, 0.084 mmol, 21% yield, formic acid salt) as a white solid. LC/MS (ESI) m/z: 470.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.16 (s, 1H), 8.03 (d, J = 9.2 Hz, 1H), 7.79 (d, J = 9.2 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 6.73 (dd, J = 2.0, 8.0 Hz, 1H), 6.67 (d, J = 2.0 Hz, 1H), 4.25 (d, J = 2.8 Hz, 4H), 3.62 (s, 2H), 3.51 (s, 3H), 2.95 (s, 2H), 2.71 - 2.66 (m, 2H), 2.64 - 2.59 (m, 2H). Synthesis of Compound 106 Step 1: Preparation of ethyl 3-[acetyl(methyl)amino]pyridine-2- carboxylate To a mixture of ethyl 3-acetamidopyridine-2-carboxylate (10 g, 48.03 mmol, 1 eq) and cesium carbonate (31.30 g, 96.06 mmol, 2 eq) in DMF (100 mL) was added iodomethane (13.63 g, 96.06 mmol, 6.0 mL, 2 eq), and the reaction mixture was stirred at 25 °C for 12 h. The mixture was poured into ice water (700 mL) and stirred for 5 minutes. The aqueous phase was extracted with ethyl acetate (300 mL × 4). The combined organic extract was washed with brine (150 mL × 2), dried with anhydrous sodium sulfate, filtered, and concentrated in vacuum. The resulting residue was purified by semi-preparative reverse phase HPLC [(column: Phenomenex luna C18 (250*70 mm, 10 um); mobile phase: (5-35% CH3CN in water (0.225% formic acid))] to afford ethyl 3-[acetyl(methyl)amino]pyridine-2- carboxylate (9.3 g, 41.85 mmol, 87% yield) as a yellow solid. LC/MS (ESI) m/z: 223.2 [M+H]+. Step 2: Preparation of ethyl 3-[acetyl(methyl)amino]-1-oxido-pyridin-1- ium-2- carboxylate To a mixture of ethyl 3-[acetyl(methyl)amino]pyridine-2-carboxylate (9.2 g, 41.40 mmol, 1 eq) and urea hydrogen peroxide (5.84 g, 62.10 mmol, 1.5 eq) in dichloromethane (92 mL) at 0 °C was added (2,2,2-trifluoroacetyl) 2,2,2-trifluoroacetate (13.04 g, 62.10 mmol, 8.6 mL, 1.5 eq) dropwise, and the reaction mixture was stirred for 2 h at 20 °C. The reaction was quenched with 10% sodium bicarbonate in water (150 mL), and the resulting mixture was extracted with dichloromethane (100 mL × 3). The combined organic extract was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated to afford ethyl 3-[acetyl(methyl)amino]-1-oxido-pyridin-1-ium-2-carboxylate (9.4 g, crude) as a yellow solid. Step 3: Preparation of ethyl 3-[acetyl(methyl)amino]-6-cyano-pyridine-2- carboxylate To a mixture of ethyl 3-[acetyl(methyl)amino]-1-oxido-pyridin-1-ium-2-carboxylate (9.4 g, 39.46 mmol, 1 eq) and trimethylsilyl cyanide (4.70 g, 47.35 mmol, 5.9 mL, 1.2 eq) in dichloromethane (100 mL) at -10 °C was added benzoyl chloride (7.76 g, 55.24 mmol, 6.4 mL, 1.4 eq) dropwise under nitrogen, and the reaction mixture was stirred at -10 °C for 15 minutes. Triethylamine (5.99 g, 59.18 mmol, 8.2 mL, 1.5 eq) was then added dropwise, and the resulting mixture was stirred for 3 h. The mixture was poured into ice water (100 mL) and stirred for 5 minutes. The aqueous phase was extracted with dichloromethane (100 mL × 2), and the combined organic extract was washed with brine (50 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by silica gel chromatography (gradient: 10-50% EtOAc in petroleum ether) to afford ethyl 3-[acetyl (methyl)amino]-6-cyano-pyridine-2-carboxylate (7.86 g, 31.79 mmol, 81% yield) as a yellow solid.1H NMR (400 MHz, CDCl3) δ 7.98 - 7.62 (m, 2H), 4.48 - 4.26 (m, 2H), 3.48 - 3.06 (m, 3H), 2.25 - 1.72 (m, 3H), 1.34 (t, J = 7.2 Hz, 3H). Step 4: Preparation of 8-hydroxy-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile To a solution of ethyl 3-[acetyl(methyl)amino]-6-cyano-pyridine-2-carboxylate (7.86 g, 31.79 mmol, 1 eq) in tetrahydrofuran (80 mL) at -78 °C was added potassium bis(trimethylsilyl)amide (1 M, 39.7 mL, 1.25 eq) dropwise under nitrogen, and the reaction mixture was stirred at -70 °C for 15 minutes, then warmed to 25 °C and stirred for 4 h. The mixture was poured into ice water (160 mL) and stirred for 5 minutes. The aqueous phase was extracted with ethyl acetate (80 mL). The aqueous phase was adjusted to pH=5~6 with 1M hydrochloric acid, and the resulting precipitate was collected by filtration and dried under high vacuo to afford 8-hydroxy-5-methyl-6- oxo-1,5-naphthyridine-2-carbonitrile (5.95 g, crude) as a yellow solid. Step 5: Preparation of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate To a solution of 8-hydroxy-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (2 g, 9.94 mmol, 1 eq) in dichloromethane (20 mL) at 0 °C was added triethylamine (2.01 g, 19.88 mmol, 2 eq) and dimethylaminopyridine (121 mg, 1.0 mmol, 0.1 eq) followed by trifluoromethanesulfonic anhydride (3.37 g, 11.93 mmol, 1.2 eq), and the reaction mixture was stirred at 20 °C for 3h. The mixture was diluted with water (30 mL) and extracted with dichloromethane (50 mL x 2). The organic extract was washed with brine (40 mL), dried over anhydrous sodium sulfate, filtered and then concentrated. The resulting residue was purified by silica gel column chromatography (gradient: 5-50% EtOAc in petroleum ether) to afford (6-cyano-1-methyl-2- oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (1.9 g, 5.70 mmol, 57% yield) as a light yellow solid. Step: 6 Preparation of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate To a solution of (6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (50 mg, 150.04 µmol, 1 eq) and tert-butyl 2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole- 5-carboxylate (47.78 mg, 225.06 µmol, 1.5 eq) in acetonitrile (3 mL) was added N,N- diisopropylethylamine (58.18 mg, 450.12 µmol, 78.40 µL, 3 eq), and the reaction mixture was stirred at 85 °C for 12 h. The mixture was diluted with ethyl acetate (10 mL*3) and water (10 mL), and the organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by prep-TLC (SiO2, CH2Cl2/CH3OH = 10:1) to afford tert-butyl2-(6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4- yl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (55 mg, 93% yield) as a white solid. LC/MS (ESI) m/z: 396.2 [M+H] +.1H NMR (400 MHz, CDCl3) δ 7.79 (d, J = 8.4 Hz, 1H), 7.66 (d, J = 8.8 Hz, 1H), 5.81 (s, 1H), 4.12 (s, 2H), 3.86 - 3.64 (m, 4H), 3.62 (s, 3H), 3.34 (d, J = 9.2 Hz, 2H), 3.01 (s, 2H), 1.48 (s, 9H). Step 7: Preparation of 8-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl)-5- methyl-6-oxo-1,5-naphthyridine-2-carbonitrile. A solution of tert-butyl 2-(6-cyano-1-methyl-2-oxo-1,5-naphthyridin-4-yl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (170 mg, 429.89 µmol, 1 eq) in TFA (1 mL) and dichloromethane (4 mL) was stirred at 20 °C for 1 h. The mixture was concentrated to give 8-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl)-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (126 mg, crude, TFA salt) as a white solid. LC/MS (ESI) m/z: 296.1[M+H] +. Step 8: Preparation of 8-[2-[(4-fluorophenyl)methyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (Compound 106) To a solution of 8-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl)-5-methyl-6-oxo- 1,5-naphthyridine-2-carbonitrile (70 mg, 171.00 µmol, 1 eq, TFA) and 4-fluorobenzaldehyde (25.47 mg, 205.20 µmol, 21.58 µL, 1.2 eq in dichloroethane (5 mL) was added TFA (86.52 mg, 854.99 µmol, 119.00 µL, 5 eq) followed by sodium cyanoborohydride (16.12 mg, 256.50 µmol, 1.5 eq), and the reaction mixture was stirred at 20 °C for 2 h. The mixture was concentrated, and the resulting residue was diluted with water (20mL) and extracted with ethyl acetate (20 mL*3). The organic extract was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated. The resulting residue was purified by prep-HPLC [column: Phenomenex Synergi C18150*25 mm* 10 um; mobile phase: (7-37% CH3CN in water (0.225% formic acid))] to afford 8-[2-[(4- fluorophenyl)methyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-5-yl]-5-methyl-6-oxo-1,5- naphthyridine-2-carbonitrile (56.6 mg, 139.90 µmol, 82% yield) as a white solid. LC/MS (ESI) m/z: 403.9 [M+H] +.1H NMR (400 MHz, CD3OD) δ 8.08 - 7.95 (m, 2H), 7.41 (dd, J = 5.6, 8.0 Hz, 2H), 7.09 (t, J = 8.4 Hz, 2H), 5.84 (d, J = 13.6Hz, 1H), 3.99 - 3.85 (m, 6H), 3.63 (d, J = 6.8 Hz, 3H), 3.17 - 3.07 (m, 4H), 2.81 (s, 2H). Synthesis of Compound 107 Preparation of 8-[5-[1-(4-fluorophenyl)ethyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrol-2-yl]-5-methyl-6-oxo-1,5-naphthyridine-2-carbonitrile (Compound 107) To a solution of 8-(2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrol-5-yl)-5-methyl-6-oxo- 1,5-naphthyridine-2-carbonitrile (50 mg, 122.14 µmol, 1 eq, TFA) and 1-(1-bromoethyl)-4- fluoro-benzene (37.20 mg, 183.21 µmol, 1.5 eq) in acetonitrile (3 mL) was added N,N- diisopropylethylamine (47.36 mg, 366.42 µmol, 63.82 µL, 3 eq), and the reaction mixture was stirred at 85 °C for 12 h. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL*3). The organic extract was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The resulting residue was purified by prep-HPLC [column: Phenomenex Luna C18150* 25 mm* 10 um; mobile phase: (9-39% CH3CN in water (0.225% formic acid))] to afford 8-[5-[1-(4-fluorophenyl)ethyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrol-2-yl]-5-methyl-6-oxo-1,5-naphthyridine-2- carbonitrile (11.5 mg, 27.55 µmol, 22% yield) as a white solid. LC/MS (ESI) m/z: 418.1[M+H] +.1H NMR (400 MHz, CDCl3) δ 7.80 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 7.39 ( s, 2H), 7.04 (t, J = 8.4 Hz, 2H), 5.89 (s, 1H), 3.90 - 3.76 (m, 4H), 3.63 (s, 3H), 3.57 - 3.47 (m, 1H), 3.24 - 3.04 (m, 3H), 2.98 - 2.87 (m, 1H), 2.72 - 2.64 (m, 1H), 2.60 - 2.51 (m, 1H), 1.51 (d, J = 4.4 Hz, 3H). Synthesis of Compound 235 Exemplary Synthesis of 6-chloro-4-((3S,4R)-4-((4-fluorophenyl)amino)-3- methylpiperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate and tert-butyl (3R,4S)-4-(4-fluoroanilino)-3-methyl-piperidine-1-carboxylate The racemic cis isomer tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1- carboxylate (600 mg) was purified by SFC (column: DAICEL CHIRALPAK AD (250mm*30mm,10um); mobile phase: [0.1% ammonium hydroxide in ethanol]; B%: 15%- 15%,5.7; 102min.) to afford tert-butyl (3S,4R)-4-(4-fluoroanilino)-3-methyl-piperidine-1- carboxylate (298 mg, 7%) as a white solid and tert-butyl (3R,4S)-4-(4-fluoroanilino)-3- methyl-piperidine-1-carboxylate (200 mg, 5%) as a white solid. LC/MS (ESI) m/z: 308.2 [M+H]+. Step 2: Preparation of 6-chloro-4-[(3S,4R)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-4-(4- fluoroanilino)-3-methyl-piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 425.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.6 (d, J = 8.8 Hz, 1H), 6.92 (m, 1H), 6.8 (m, 1H), 6.72 (m, 2H), 5.60 (d, J = 8.9 Hz, 1H), 4.08 (m, 1H), 3.85 ( d, J = 4.8 Hz, 2H), 3.74 (m, J = 3.6, 8.4 Hz, 1H), 3.69 - 3.60 (m, 1H), 3.52 (s, 3H), 2.34 - 2.26 (m, 1H), 2(m, 1H), 1.84 (m, J = 3.4, 6.8, 9.2 Hz, 1H), 0.86 (d, J = 7.2 Hz, 3H). Synthesis of Compound 236 Exemplary Synthesis of 4-[(3aS,6aS)-5-[4-(trifluoromethoxy)phenyl]- octahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate and tert-butyl (3aS,6aS)-2- [4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate The racemic mixture of tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate was further purified by SFC. (column: DAICEL CHIRALCEL OJ (250mm*30mm,10um); mobile phase: 15% [0.1% ammonium hydroxide in methanol in supercritical carbon dioxide]; 15%-15% ; 117min) . tert-Butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate was obtained as a yellow solid. LC/MS (ESI) m/z: 373.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.13 (d, J = 8.4 Hz, 2H), 6.51 (d, J = 9.2 Hz, 2H), 3.54 (dd, J = 6.4, 9.2 Hz, 2H), 3.45 - 3.36 (m, 2H), 3.09 - 2.99 (m, 4H), 2.36 - 2.31 (m, 2H), 1.41 (s, 9H). tert-Butyl (3aS,6aS)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate was obtained as a yellow solid. LC/MS (ESI) m/z: 373.1 [M+H]+; 1H NMR (400MHz, DMSO-d6) δ 7.13 (d, J = 8.8 Hz, 2H), 6.55 - 6.46 (m, 2H), 3.54 (dd, J = 6.0, 9.2 Hz, 2H), 3.44 - 3.36 (m, 2H), 3.09 - 2.98 (m, 4H), 2.36 - 2.29 (m, 2H), 1.41 (s, 9H). Step 2: Preparation of 4-[(3aS,6aS)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5- carboxylate. (yellow solid). LC/MS (ESI) m/z: 490.2 [M+H]+; 1H NMR (400 MHz, DMSO- d6) δ 8.02 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.63 - 6.49 (m, 2H), 4.31 - 4.14 (m, 4H), 3.58 - 3.48 (m, 5H), 3.21 (t, J = 9.6 Hz, 2H), 2.49 - 2.40 (m, 2H). Synthesis of Compound 237 Exemplary Synthesis of 6-chloro-4-((3R,4S)-4-((4-fluorophenyl)amino)-3- methylpiperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile
The title compound was prepared in an analogous manner to 6-chloro-4-((3S,4R)-4-((4- fluorophenyl)amino)-3-methylpiperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile stating from 6-chloro-4-[(3R,4S)-4-(4-fluoroanilino)-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS (ESI) m/z: 439.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.2 (d, J = 9.0 Hz, 1H), 7.80 (d, J = 9.0 Hz, 1H), 6.96 - 6.84 (m, 2H), 6.72 - 6.66 (m, 2H), 5.72 - 5.49 (m, 1H), 4.08 - 3.98 (m, 1H), 3.84 (br d, J = 4.8 Hz, 2H), 3.72 (td, J = 4.0, 8.1 Hz, 1H), 3.68 - 3.60 (m, 1H), 3.52 (s, 3H), 2.34 - 2.30 (m, 1H), 2 (m, 1H), 1.88 - 1.78 (m, 1H), 0.86 (d, J = 7.0 Hz, 3H). Synthesis of Compound 238 Exemplary Synthesis of 4-[(3aR,6aR)-5-[4-(trifluoromethoxy)phenyl]- octahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 4-[(3aS,6aS)-5-[4- (trifluoromethoxy)phenyl]-octahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,2- dihydro-1,5-naphthyridine-3-carbonitrile starting from tert-butyl (3aR,6aR)-2-[4- (trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate. (yellow solid). LC/MS (ESI) m/z: 490.0 [M+H]+; 1H NMR (400MHz, DMSO-d6) δ 8.02 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.57 (d, J = 9.2 Hz, 2H), 4.28 - 4.18 (m, 4H), 3.55 - 3.49 (m, 5H), 3.21 (t, J = 9.2 Hz, 2H), 2.45 (s, 2H). Synthesis of Compound 239 Exemplary Synthesis of 4-(5-fluorospiro[3H-benzofuran-2,4'-piperidine]-1'-yl)-1,6- dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of ethyl 3-amino-6-bromo-pyridine-2-carboxylate To a solution of ethyl 3-aminopyridine-2-carboxylate (10 g, 60.18 mmol, 1 eq) in acetonitrile (100 mL) was added N-bromosuccinimide (10.82 g, 60.78 mmol, 1.01 eq). The mixture was stirred at 25 °C for 1.5 h, then concentrated under reduced pressure to afford ethyl 3-amino-6- bromo-pyridine-2-carboxylate (14.3 g, 97%) as a yellow solid. LC/MS (ESI) m/z: 245.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.33 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 8.8 Hz, 1H), 5.82 (s, 2H), 4.44 (J = 7.2 Hz, 2H), 1.44 (J = 7.2 Hz, 3H). Step 2: Preparation of ethyl 3-amino-6-methyl-pyridine-2-carboxylate A mixture of ethyl 3-amino-6-bromo-pyridine-2-carboxylate (4.15 g, 16.93 mmol, 1 eq), cyclopentyl(diphenyl)phosphane;dichloropalladium;iron (1.24 g, 1.69 mmol, 0.1 eq), 2,4,6- trimethyl-1,3,5,2,4,6-trioxatriborinane (12.75 g, 50.80 mmol, 14.20 mL, 50% purity, 3 eq), and sodium bicarbonate (3.59 g, 33.87 mmol, 2 eq) in dioxane (40 mL) and water (1.5 mL) was degassed and purged with nitrogen (3X). The mixture was then stirred at 90 °C for 12 h under nitrogen atmosphere. The residue was extracted with ethyl acetate (3 x 100 mL), and the combined organic layers were washed with brine (3 x 100 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by column chromatography (petroleum ether/ethyl acetate = 1/1) to give ethyl 3-amino-6- methyl-pyridine-2-carboxylate (5.65 g, 92%) as a yellow solid. LC/MS (ESI) m/z: 181.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.13 - 7.10 (m, 1H), 7.01 (d, J = 8.4 Hz, 1H), 4.47 ( J = 7.2 Hz, 2H), 2.51 (s, 3H), 1.46 (t, J = 7.2 Hz, 3H). Step 3: Preparation of ethyl 3-[(2-cyanoacetyl)amino]-6-methyl-pyridine-2-carboxylate 2-Cyanoacetic acid (1.72 g, 20.20 mmol, 1.3 eq) was added to acetic anhydride (20 mL), and the reaction mixture was stirred at 80 °C for 1 hr. Ethyl 3-amino-6-methyl-pyridine-2- carboxylate (2.8 g, 15.54 mmol, 1 eq) was then added, and the reaction mixture was stirred at 80 °C for 1 h, then concentrated under reduced pressure. The resulting residue was purified by column chromatography (petroleum ether/ethyl acetate = 2/1) to give ethyl 3-[(2- cyanoacetyl)amino]-6-methyl-pyridine-2-carboxylate (5 g, 65%) as a white solid. LC/MS (ESI) m/z: 248.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 11.62 (s, 1H), 8.93 - 8.89 (m, 1H), 7.40 (d, J = 8.8 Hz, 1H), 4.60 - 4.53 (m, 2H), 3.65 - 3.61 (m, 2H), 2.64 (s, 3H), 1.49 (t, J = 7.2 Hz, 3H). Step 4: Preparation of 4-hydroxy-6-methyl-2-oxo-1H-1,5-naphthyridine-3-carbonitrile A mixture of ethyl 3-[(2-cyanoacetyl)amino]-6-methyl-pyridine-2-carboxylate (5 g, 20.22 mmol, 1 eq) and 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine (3.39 g, 22.24 mmol, 3.35 mL, 1.1 eq) in ethanol (40 mL) was degassed and purged with nitrogen for 3 times, then the mixture was stirred at 25 °C for 1 h under nitrogen atmosphere. The pH was adjusted to 5 with 1M hydrochloric acid, and the resulting suspension was filtered. The filtered cake was dried in vacuum to afford 4-hydroxy-6-methyl-2-oxo-1H-1,5-naphthyridine-3-carbonitrile (3.62 g, 89%) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 11.38 - 11.26 (m, 1H), 7.91 (d, J = 8.4Hz, 1H), 7.75 (d, J = 8.8Hz, 1H), 2.69 (s, 3H). Step 5: Preparation of 4-hydroxy-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a mixture of 4-hydroxy-6-methyl-2-oxo-1H-1,5-naphthyridine-3-carbonitrile (1.6 g, 7.95 mmol, 1 eq) in DMF (20 mL) was added sodium hydride (795.23 mg, 19.88 mmol, 60%, 2.5 eq), and the resulting mixture was stirred at 0°C for 0.5 h. Methyl iodide (3.39 g, 23.86 mmol, 1.49 mL, 3 eq) was then added, and the reaction mixture was stirred at 0 °C for 0.5 h under nitrogen. The reaction was quenched and the pH adjusted to 5 by addition of 1 M hydrochloric acid. The resulting mixture was filtered, and the filter cake was dried to afford 4-hydroxy-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (1.8 g, crude) as a yellow solid. LC/MS (ESI) m/z: 216.1 [M+H]+. Step 6: Preparation of 4-chloro-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a mixture of 4-hydroxy-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (300 mg, 1.39 mmol, 1 eq) and N-ethyl-N-isopropyl-propan-2-amine (1.08 g, 8.36 mmol, 1.46 mL, 6 eq) in acetonitrile (5 mL) were added phosphorus oxychloride (1.07 g, 6.97 mmol, 0.65 mL, 5 eq) and benzyltriethylammonium chloride (635 mg, 2.79 mmol, 2 eq) in one portion at 20 °C under nitrogen, and the reaction mixture was stirred at 20 °C for 12 h. The mixture was concentrated, and the resulting residue was poured into ice and saturated aqueous sodium bicarbonate (10 mL). The aqueous mixture was extracted with ethyl acetate (2 x 15 mL), and the combined organic layers were washed with brine (30 mL), dried with sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate = 30/1 to 0:1) to give 4-chloro-1,6-dimethyl- 2-oxo-1,5-naphthyridine-3-carbonitrile (60 mg, 18%) as a yellow solid. LC/MS m/z: 233.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 8.8 Hz, 1H), 7.77 (d, J = 8.8 Hz, 1H), 3.64 (s, 3H), 2.62 (s, 3H). Step 7: Preparation of 4-(5-fluorospiro[3H-benzofuran-2,4'-piperidine]-1'-yl)-1,6- dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile To a mixture of 5-fluorospiro[3H-benzofuran-2,4'-piperidine] (92 mg, 0.3 mmol, 1.34 eq, trifluoroacetic acid) and diisopropylethylamine (371 mg, 2.87 mmol, 0.5 mL, 13.41 eq) in acetonitrile (4 mL) was added 4-chloro-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (50 mg, 0.2 mmol, 1 eq), and the reaction mixture was stirred at 25°C for 12 h. The mixture was concentrated, and the resulting residue was purified by prep-HPLC (column: Phenomenex C18150*25 mm*10um; mobile phase: [45-75% acetonitrile in water (ammonium bicarbonate)]) to afford 4-(5-fluorospiro[3H-benzofuran-2,4'-piperidine]-1'-yl)- 1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (18.0 mg, 21%) as an off-white solid. LC/MS (ESI) m/z: 405.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J = 8.8 Hz, 1H), 7.60 (d, J = 8.8 Hz, 1H), 7.09 (dd, J = 2.4, 8.4 Hz, 1H), 6.95 - 6.88 (m, 1H), 6.79 (dd, J = 4.4, 8.8 Hz, 1H), 4.05 - 3.95 (m, 2H), 3.95 - 3.84 (m, 2H), 3.52 (s, 3H), 3.15 (s, 2H), 2.56 (s, 3H), 2.08 (t, J = 5.2 Hz, 4H).
Synthesis of Compound 240 Exemplary Synthesis of 4-[(3aR,6aR)-2-(4-fluorophenyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-1,6-dimethyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 4-(5-fluorospiro[3H-benzofuran- 2,4'-piperidine]-1'-yl)-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert- butyl (3S,4S)-3,4-bis(p-tolylsulfonyloxymethyl)pyrrolidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 404.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.85 (d, J = 8.8 Hz, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.01 (t, J = 8.8 Hz, 2H), 6.53 - 6.46 (m, 2H), 4.34 - 4.19 (m, 4H), 3.53 - 3.43 (m, 5H), 3.18 (t, J = 9.2 Hz, 2H), 2.55 (s, 3H), 2.47 - 2.42 (m, 2H). Synthesis of Compound 241 Exemplary Synthesis of 2,4-dimethyl-7-[(3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]-1-piperidyl]-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate and tert-butyl (3R,4S)-3-methyl-4- [N-methyl-4-(trifluoromethoxy)anilino]piperidine-1-carboxylate Racemic tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (700 mg) was purified by SFC (column: DAICEL CHIRALPAK IG (250mm*30mm, 10 um); mobile phase: [0.1% ammonium hydroxide IPA]; B%: 5%-5%,3.3; 65min), and further purified by HPLC (column: Phenomenex Luna C18200*40mm*10um; mobile phase: [water(formic acid)- acetonitrile]; B%: 65%-95%, 10 min). tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (130 mg, 19%) was obtained as a yellow oil. LC/MS (ESI) m/z: 389.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.14 (d, J = 8.8 Hz, 2H), 6.80 - 6.76 (m, 2H), 4.19 - 4.02 (m, 1H), 3.87 (d, J = 12.4 Hz, 1H), 3.83 - 3.70 (m, 1H), 3.44 (q, J = 7.2 Hz, 1H), 3.17 - 2.95 (m, 1H), 2.79 (s, 3H), 2.14 (s, 1H), 2.08 - 1.96 (m, 1H), 1.50 (dd, J = 2.4, 12.4 Hz, 1H), 1.40 (s, 9H), 0.81 (d, J = 6.8 Hz, 3H). tert-butyl (3R,4S)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (120 mg, 17%) was obtained as a yellow oil. LC/MS (ESI) m/z: 389.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.14 (d, J = 8.8 Hz, 2H), 6.80 - 6.74 (m, 2H), 4.18 - 4.02 (m, 1H), 3.87 (d, J = 12.0 Hz, 1H), 3.83 - 3.70 (m, 1H), 3.15 - 2.94 (m, 1H), 2.79 (s, 3H), 2.13 (s, 1H), 2.05 - 1.96 (m, 1H), 1.50 (dd, J = 2.4, 12.4 Hz, 1H), 1.40 (s, 9H), 0.81 (d, J = 6.8 Hz, 3H). Step 2: Preparation of 2,4-dimethyl-7-[(3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]-1-piperidyl]-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile The title compounds was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7- spiro[3H-benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate. (yellow solid). LC/MS (ESI) m/z: 492.2 [M+H]+; 1H NMR (400 MHz, DMSO- d6) δ 7.17 (d, J = 8.8 Hz, 2H), 6.86 (d, J = 9.2 Hz, 2H), 4.56 - 4.49 (m, 1H), 4.32 (d, J = 13.2 Hz, 1H), 4.14 - 4.07 (m, 1H), 3.79 (dd, J = 2.4, 13.2 Hz, 1H), 3.49 (s, 3H), 3.46 - 3.38 (m, 1H), 2.85 (s, 3H), 2.72 (s, 3H), 2.54 - 2.53 (m, 1H), 2.39 - 2.33 (m, 1H), 1.80 - 1.72 (m, 1H), 0.84 (d, J = 7.2 Hz, 3H). Synthesis of Compound 242 Exemplary Synthesis of 6-chloro-1-methyl-2-oxo-4-(5-(trifluoromethoxy)-1,3- dihydrospiro[indene-2,4'-piperidin]-1'-yl)-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of methyl 1-benzyl-4-[[4- (trifluoromethoxy)phenyl]methyl]piperidine-4-carboxylate To methyl 1-benzylpiperidine-4-carboxylate (3 g, 12.86 mmol, 1 eq) in tetrahydrofuran (30 mL) was added [bis(trimethylsilyl)amino]lithium (1 M, 19.29 mL, 1.5 eq) at -70°C, and the resulting mixture was stirred for 0.5 h.1-(Bromomethyl)-4-(trifluoromethoxy)benzene (3.28 g, 12.86 mmol, 2.06 mL, 1 eq) was then added, and the reaction mixture was stirred at 20 °C for 11.5 h under nitrogen. The reaction was diluted with saturated aqueous ammonium chloride (20 mL) and water (20 ml), and the resulting mixture was extracted with ethyl acetate (4 x 20 mL). The combined organic extracts were washed with brine (80 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by prep- HPLC (column: UniSil 10-120 C1850x250 mm; mobile phase: [water (formic acid) in acetonitrile]; 22%-52%, 23 min) to afford methyl 1-benzyl-4-[[4- (trifluoromethoxy)phenyl]methyl]piperidine-4-carboxylate formic acid (4.3 g, 74%) as a yellow oil. LC/MS(ESI) m/z: 408.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.16 (s, 1H), 7.38 - 7.20 (m, 7H), 7.18 - 7.11 (m, 2H), 3.58 (s, 3H), 3.45 (s, 2H), 2.81 (s, 2H), 2.75 - 2.66 (m, 2H), 2.04 - 1.87 (m, 4H), 1.60 - 1.47 (m, 2H). Step 2: Preparation of 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1- one A mixture of methyl 1-benzyl-4-[[4-(trifluoromethoxy)phenyl]methyl]piperidine-4- carboxylate (4.3 g, 9.48 mmol, 1 eq, formic acid) in trifluoromethanesulfonic acid (40 mL) was degassed and purged with nitrogen (3X), then stirred at 80 °C for 3 h under nitrogen atmosphere. The reaction mixture was poured into water (200 mL), and the resulting mixture was treated with sodium bicarbonate to adjust the pH to 7~8. The mixture was extracted with ethyl acetate (3 x 200 mL), and the combined organic layers were washed with brine (400 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex luna C18 (250*70mm, 10 um); mobile phase: [water (formic acid) in acetonitrile; 21%-51%, 25 min) to afford 1'-benzyl-6- (trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-one (2.4 g, 67%) as a white solid. LC/MS (ESI) m/z: 376.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.55 - 7.46 (m, 8H), 4.29 (s, 2H), 3.63 (J = 2.8, 12.0 Hz, 2H), 3.38 (d, J = 12.0 Hz, 2H), 3.10 (s, 2H), 2.36 (s, 2H), 2.01 (d, J = 14.4 Hz, 2H). Step 3: Preparation of 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-ol A mixture of 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-one (2.3 g, 6.13 mmol, 1 eq) and lithium aluminum hydride (465 mg, 12.25 mmol, 2 eq) in tetrahydrofuran (20 mL) was degassed and purged with nitrogen for 3 times. The reaction mixture was then stirred at 0°C for 1 h under nitrogen atmosphere. Water (0.5 mL) and 1 N sodium hydroxide (1.5 mL) were added, and the resulting mixture was stirred for 0.5 h. The mixture was filtered, and the filtrate was dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-ol (3 g, crude) as a colorless oil. LC/MS (ESI) m/z: 378.1 [M+H]+. Step 4: Preparation of 1'-benzyl-5-(trifluoromethoxy)spiro[indane-2,4'-piperidine] To 1'-benzyl-6-(trifluoromethoxy)spiro[indane-2,4'-piperidine]-1-ol (2.8 g, 7.42 mmol, 1 eq) were added triethylsilane (4.31 g, 37.10 mmol, 5.93 mL, 5 eq) and trifluoroacetic acid (5.08 g, 44.52 mmol, 3.30 mL, 6 eq), and the reaction mixture was stirred at 80 °C for 12 h. The pH was adjusted to 8 by addition of NaHCO3, and the resulting mixture was extracted with ethyl acetate (30 mL x 3). The combined organic layers were washed with brine 90 mL (30 mL x 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by flash column chromatography (petroleum ether/ethyl acetate = 5/1 to 1/1) to afford 1'-benzyl-5-(trifluoromethoxy)spiro[indane-2,4'-piperidine] (950 mg, 35%) as a colorless oil. LC/MS (ESI) m/z: 362.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.27 - 7.16 (m, 5H), 7.07 (d, J = 8.0 Hz, 1H), 6.94 (s, 1H), 6.90 (d, J = 8.4 Hz, 1H), 3.47 (s, 2H), 2.72 (d, J = 10.0 Hz, 4H), 2.39 (s, 4H), 1.59 (t, J = 5.6 Hz, 4H). Step 5: Preparation of 5-(trifluoromethoxy)spiro[indane-2,4'-piperidine] To a solution of 1'-benzyl-5-(trifluoromethoxy)spiro[indane-2,4'-piperidine] (800 mg, 2.21 mmol, 1 eq) in methanol (8 mL) was added 10% palladium on carbon (200 mg, 2.21 mmol, 1 eq) under nitrogen, and the suspension was degassed under vacuum and purged with hydrogen several times. The mixture was stirred under hydrogen (50 psi) at 25°C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to afford 5- (trifluoromethoxy)spiro[indane-2,4'-piperidine] (550 mg, 92%) as a colorless oil. LC/MS (ESI) m/z: 272.0 [M+H]+. Step 6: Preparation of 6-chloro-1-methyl-2-oxo-4-(5-(trifluoromethoxy)-1,3- dihydrospiro[indene-2,4'-piperidin]-1'-yl)-1,2-dihydro-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-4-[4-(4-chloroanilino)- 1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from 5- (trifluoromethoxy)spiro[indane-2,4'-piperidine]. (yellow solid). LC/MS (ESI) m/z: 489.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.79 (d, J = 9.2 Hz, 1H), 7.32 (d, J = 8.4 Hz, 1H), 7.20 (s, 1H), 7.10 (d, J = 8.0 Hz, 1H), 3.85 (s, 4H), 3.52 (s, 3H), 2.93 (d, J = 12.8 Hz, 4H), 1.82 (t, J = 4.8 Hz, 4H). Synthesis of Compound 243 Exemplary Synthesis of 7-[4-(4-chloroanilino)-1-piperidyl]-2,4-dimethyl-5-oxo- thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from N-(4- chlorophenyl) piperidin-4-amine. (off-white solid). LC/MS (ESI) m/z: 413.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.09 (d, J = 8.8 Hz, 2H), 6.64 (d, J = 8.8 Hz, 2H), 5.83 (d, J = 8.0 Hz, 1H), 4.37 (d, J = 13.2 Hz, 2H), 3.65 - 3.51 (m, 3H), 3.48 (s, 3H), 2.70 (s, 3H), 2.12 - 2.02 (m, 2H), 1.63 - 1.50 (m, 2H). Synthesis of Compound 244 Exemplary Synthesis of 2,4-dimethyl-7-[(3R,4S)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]-1-piperidyl]-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-7-[(3S,4R)-3- methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]-1-piperidyl]-5-oxo-thiazolo[5,4- b]pyridine-6-carbonitrile starting from tert-butyl (3R,4S)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate. (off-white solid). LC/MS (ESI) m/z: 492.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.17 (d, J = 8.8 Hz, 2H), 6.86 (d, J = 9.2 Hz, 2H), 4.52 (d, J = 12.0 Hz, 1H), 4.32 (d, J = 13.2 Hz, 1H), 4.15 - 4.07 (m, 1H), 3.79 (dd, J = 2.4, 13.2 Hz, 1H), 3.49 (s, 3H), 3.46 - 3.38 (m, 1H), 2.85 (s, 3H), 2.72 (s, 3H), 2.61 - 2.55 (m, 1H), 2.38 - 2.33 (m, 1H), 1.76 (dd, J = 2.0, 12.8 Hz, 1H), 0.84 (d, J = 6.8 Hz, 3H).
Synthesis of Compound 245 Exemplary Synthesis of 7-((3S,4R)-4-((4-chloro-2-methoxyphenyl)amino)-3- methylpiperidin-1-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3S,4R)-N-(4-chloro-2-methoxy-phenyl)-3-methyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 457.13 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.86 (d, J = 2.4 Hz, 1H), 6.8 (d, J = 2.3, 8.4 Hz, 1H), 6.6 (d, J = 8.4 Hz, 1H), 4.60 (d, J = 8.8 Hz, 1H), 4.24 - 4.10 (m, 2H), 3.82 (s, 3H), 3.82 - 3.76 (m, 1H), 3.78 (d, J = 4.0 Hz, 1H), 3.68 - 3.58 (m, 1H), 3.47 (s, 3H), 2.64 (s, 3H), 2.24(m, 1H), 2.08 - 1.96 (m, 1H), 1.82 (m ,J = 4.0, 9.0 Hz, 1H), 0.78 (d, J = 6.4 Hz, 3H).
Synthesis of Compound 246 Exemplary Synthesis of 7-[(3S,4R)-4-[(4-chloro-2-fluorophenyl)amino]-3- methylpiperidin-1-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate, tert-butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate, tert-butyl (3R,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate, and tert-butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl- piperidine-1-carboxylate To a solution of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.07 mmol, 1 eq) in methanol (25 mL) and acetic acid (25 mL) were added 4-chloro-2-fluoro-aniline (2.46 g, 16.88 mmol, 1.88 mL, 1.2 eq) and 2-methylpyridine borane (3.76 g, 35.18 mmol, 2.5 eq), and the reaction mixture was stirred at 40 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18 (250*70mm,10 um); mobile phase: [water (formic acid)- acetonitrile]; B%: 65%-95%, 19 min) and then further separated by SFC (column: DAICEL CHIRALPAK AD(250mm*30mm,10um); mobile phase: [0.1% ammonium hydroxide methanol]; B%: 15%-15%, 4.0; 480 min). LC/MS (ESI) m/z: 287.0 [M-55]+. tert-Butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (444 mg, 9%) was obtained as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 7.18 (dd, J = 2.4, 12.0 Hz, 1H), 7.00 (dd, J = 1.2, 8.4 Hz, 1H), 6.81 (t, J = 9.2 Hz, 1H), 5.22 (d, J = 7.2 Hz, 1H), 3.93 - 3.75 (m, 1H), 3.70 - 3.52 (m, 2H), 3.08 (d, J = 12.0 Hz, 1H), 3.00 - 2.77 (m, 1H), 2.04 (td, J = 3.6, 6.8 Hz, 1H), 1.74 - 1.61 (m, 1H), 1.56 - 1.46 (m, 1H), 1.39 (s, 9H), 0.75 (d, J = 6.8 Hz, 3H). tert-Butyl (3R,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (558 mg, 12%) was obtained as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 7.18 (dd, J = 2.4, 12.0 Hz, 1H), 7.00 (dd, J = 1.2, 8.4 Hz, 1H), 6.81 (t, J = 9.6 Hz, 1H), 5.22 (d, J = 7.2 Hz, 1H), 3.93 - 3.76 (m, 1H), 3.71 - 3.57 (m, 2H), 3.08 (d, J = 12.0 Hz, 1H), 2.99 - 2.76 (m, 1H), 2.11 - 1.99 (m, 1H), 1.76 - 1.61 (m, 1H), 1.57 - 1.46 (m, 1H), 1.39 (s, 9H), 0.75 (d, J = 7.2 Hz, 3H). tert-Butyl (3R,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (149 mg, 3%) was obtained as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 7.15 (dd, J = 2.4, 12.0 Hz, 1H), 6.98 (d, J = 8.4 Hz, 1H), 6.79 (t, J = 9.2 Hz, 1H), 5.34 (dd, J = 2.0, 9.2 Hz, 1H), 3.91 (d, J = 9.2 Hz, 2H), 3.18 - 3.05 (m, 1H), 2.90 - 2.76 (m, 1H), 2.42 (s, 1H), 1.83 (dd, J = 3.6, 13.2 Hz, 1H), 1.68 - 1.57 (m, 1H), 1.40 (s, 9H), 1.24 - 1.14 (m, 1H), 0.86 (d, J = 6.4 Hz, 3H). tert-Butyl (3S,4S)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1-carboxylate (136 mg, 3%) was obtained as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 7.15 (dd, J = 2.4, 12.0 Hz, 1H), 7.02 - 6.95 (m, 1H), 6.79 (t, J = 9.2 Hz, 1H), 5.35 (dd, J = 2.0, 9.2 Hz, 1H), 3.91 (d, J = 10.0 Hz, 2H), 3.14 - 3.04 (m, 1H), 2.90 - 2.77 (m, 1H), 2.49 - 2.41 (m, 1H), 1.83 (dd, J = 3.2, 13.0 Hz, 1H), 1.69 - 1.57 (m, 1H), 1.40 (s, 9H), 1.28 - 1.14 (m, 1H), 0.86 (d, J = 6.8 Hz, 3H). Step 2: Preparation of (3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4- amine To a solution of tert-butyl (3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-piperidine-1- carboxylate (70 mg, 0.20 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 66.15 eq), and the reaction the mixture was stirred at 25 C for 0.5 h. The mixture was concentrated under reduced pressure to give (3S,4R)-N-(4-chloro- 2-fluoro-phenyl)-3-methyl-piperidin-4-amine (73 mg, trifluoroacetic acid) as a colorless oil. LC/MS (ESI) m/z: 243.0 [M+H]+. Step 3: Preparation of 7-[(3S,4R)-4-(4-chloro-2-fluoro-anilino)-3-methyl-1-piperidyl]- 2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile To a solution of (3S,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4-amine (73 mg, 0.21 mmol, 1 eq, trifluoroacetic acid) in acetonitrile (2 mL) was added N,N- diisopropylethylamine (132 mg, 1.02 mmol, 0.178 mL, 5 eq) dropwise followed by (6-cyano- 2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate (72 mg, 0.205 mmol, 1 eq), and the reaction mixture was stirred at 40 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water (formic acid)- acetonitrile]; B%: 52%-82%, 9 min) to give 7-[(3S,4R)-4-(4-chloro-2-fluoro-anilino)-3- methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (70.5 mg, 77%) as a yellow solid. LC/MS (ESI) m/z: 446.0 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 7.19 (dd, J = 2.4, 11.6 Hz, 1H), 7.06 - 6.95 (m, 1H), 6.95 - 6.83 (m, 1H), 5.46 - 5.37 (m, 1H), 4.27 - 4.15 (m, 2H), 3.86 - 3.74 (m, 2H), 3.67 - 3.57 (m, 1H), 3.48 (s, 3H), 2.70 (s, 3H), 2.30 - 2.21 (m, 1H), 2.19 - 2.02 (m, 1H), 1.84 - 1.70 (m, 1H), 0.81 (d, J = 6.8 Hz, 3H). Synthesis of Compound 247 Exemplary Synthesis of 7-{4-[(4-chloro-2-fluorophenyl)amino]piperidin-1-yl}-2,4- dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of tert-butyl 4-(4-chloro-2-fluoro-anilino)piperidine-1-carboxylate A mixture of 4-chloro-2-fluoro-aniline (1 g, 6.87 mmol, 1 eq), tert-butyl 4-oxopiperidine-1- carboxylate (1.51 g, 7.56 mmol, 1.1 eq), borane;2-methylpyridine (1.84 g, 17.17 mmol, 2.5 eq), in methanol (10 mL), and acetic acid (10 mL) was degassed and purged with nitrogen (3X), then stirred at 40 °C for 12 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18250*50mm*15um; mobile phase: [water (formic acid)- acetonitrile]; B%: 35%-65%, 20 min) to afford tert-butyl 4-(4-chloro-2-fluoro-anilino)piperidine-1- carboxylate (1.5 g, 66%) as a white solid.1H NMR (400 MHz, CDCl3) δ 7.06 - 6.96 (m, 2H), 6.69 ( d, J = 5.2 Hz, 1H), 4.07 ( d, J = 10.0 Hz, 2H), 3.46 - 3.36 (m, 1H), 2.92 ( t, J = 12.4 Hz, 2H), 2.06 - 2.00 (m, 2H), 1.47 (s, 9H), 1.42 - 1.37 (m, 2H). Step 2: Preparation of N-(4-chloro-2-fluoro-phenyl)piperidin-4-amine A mixture of tert-butyl 4-(4-chloro-2-fluoro-anilino)piperidine-1-carboxylate (100 mg, 0.3 mmol, 1 eq) and trifluoroacetic acid (1.08 g, 9.45 mmol, 0.7 mL, 31.09 eq) in dichloromethane (0.7 mL) was degassed and purged with nitrogen atmosphere (3X), then stirred at 25 °C for 0.5 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure to afford N-(4-chloro-2-fluoro-phenyl)piperidin-4-amine trifluoroacetic acid (104 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 229 [M+H]+. Step 3: Preparation of 7-[4-(4-chloro-2-fluoro-anilino)-1-piperidyl]-2,4-dimethyl-5-oxo- thiazolo[5,4-b]pyridine-6-carbonitrile
The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (6- cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate. (white solid). LC/MS (ESI) m/z: 432 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.18 (dd, J = 2.4, 11.6 Hz, 1H), 7.09 - 7.00 (m, 1H), 6.88 (t, J = 9.2 Hz, 1H), 5.53 (d, J = 7.2 Hz, 1H), 4.43 (d, J = 13.2 Hz, 2H), 3.74 - 3.59 (m, 1H), 3.53 - 3.45 (m, 5H), 2.69 (s, 3H), 2.09 - 1.98 (m, 2H), 1.74 - 1.59 (m, 2H). Synthesis of Compound 248 Exemplary Synthesis of 4-[(3S,4R)-4-[(4-fluorophenyl)(methyl)amino]-3- methylpiperidin-1-yl]-1,6-dimethyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 4-(5-fluorospiro[3H-benzofuran- 2,4'-piperidine]-1'-yl)-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-N-(4-fluorophenyl)-N,3-dimethyl-piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 420.2 [M+H]+; 1H NMR (400MHz, DMSO-d6) δ 7.92 (d, J = 8.8 Hz, 1H), 7.60 (d, J = 8.8 Hz, 1H), 7.10 - 6.97 (m, 2H), 6.86 (dd, J = 4.4, 9.2 Hz, 2H), 4.58 - 4.38 (m, 1H), 4.10 - 3.97 (m, 2H), 3.86 (dd, J = 2.0, 12.8 Hz, 1H), 3.52 (s, 3H), 3.43 - 3.35 (m, 2H), 2.83 (s, 3H), 2.74 - 2.68 (m, 1H), 2.60 (s, 3H), 1.79 - 1.67 (m, 1H), 0.85 (d, J = 7.2 Hz, 3H).
Synthesis of Compound 249 Exemplary Synthesis of 1,6-dimethyl-4-[methyl({1-[4- (trifluoromethoxy)phenyl]piperidin-4-yl})amino]-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile Step 1: Preparation of tert-butyl N-methyl-N-[1-[4-(trifluoromethoxy)phenyl]-4- piperidyl]carbamate A mixture of tert-butyl N-methyl-N-(4-piperidyl)carbamate (200 mg, 0.93 mmol, 1 eq), 1- bromo-4-(trifluoromethoxy)benzene (0.34 g, 1.40 mmol, 0.2 mL, 1.5 eq), cesium carbonate (0.9 g, 2.80 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H- imidazol-1-ium-2-ide;3-chloropyridine dichloropalladium (45 mg, 0.05 mmol, 0.05 eq) in dioxane (3 mL) was degassed and purged with nitrogen (3X), then stirred at 100 °C for 4 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure, and the resulting residue was purified by column chromatography (petroleum ether:ethyl acetate = 3/1) to afford tert-butylN-methyl-N-[1-[4-(trifluoromethoxy)phenyl]-4- piperidyl]carbamate (311 mg, 89%) as a yellow solid. LC/MS (ESI) m/z: 375.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.10 (d, J = 8.8 Hz, 2H), 6.94 - 6.85 (m, 2H), 4.31 - 3.89 (m, 1H), 3.74 - 3.67 (m, 2H), 2.87 - 2.69 (m, 5H), 1.91 - 1.69 (m, 4H), 1.48 (s, 9H). Step 2: Preparation of N-methyl-1-[4-(trifluoromethoxy)phenyl]piperidin-4-amine H N N F3CO A mixture of tert-butyl N-methyl-N-[1-[4-(trifluoromethoxy)phenyl]-4-piperidyl]carbamate (100 mg, 0.28 mmol, 1 eq) and triethylamine (1.1 g, 9.5 mmol, 0.7 mL, 35.40 eq) in dichloromethane (0.7 mL) was degassed and purged with nitrogen (3X), then stirred at 25°C for 0.5 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure to give N-methyl-1-[4-(trifluoromethoxy)phenyl]piperidin-4-amine trifluoroacetic acid (103 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 275.1 [M+H]+. Step 3: Preparation of 1,6-dimethyl-4-[methyl-[1-[4-(trifluoromethoxy)phenyl]-4- piperidyl]amino]-2-oxo-1,5-naphthyridine-3-carbonitrile A mixture of N-methyl-1-[4-(trifluoromethoxy)phenyl]piperidin-4-amine trifluoroacetic acid (103 mg, 0.27 mmol, 1 eq), 4-chloro-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (62 mg, 0.27 mmol, 1 eq) and diisopropylethylamine(102 mg, 0.80 mmol, 0.14 mL, 3 eq) in acetonitrile (2 mL) was degassed and purged with nitrogen (3X), then stirred at 40 °C for 3 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex Luna C18 150*25mm*10um; mobile phase: [water (formic acid)- acetonitrile]; B%:52%-82%, 9 min) to afford 6-dimethyl-4-[methyl-[1-[4-(trifluoromethoxy)phenyl]-4-piperidyl]amino]-2-oxo- 1,5-naphthyridine-3-carbonitrile formic acid (24 mg, 17%) as a yellow solid. LC/MS (ESI) m/z: 472.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.41 (s, 1H), 7.91 (d, J = 8.8 Hz, 1H), 7.59 (d, J = 8.8 Hz, 1H), 7.18 (d, J = 8.6 Hz, 2H), 7.05 (d, J = 9.2 Hz, 2H), 4.65 - 4.50 (m, 1H), 3.86 (d, J = 12.4 Hz, 2H), 3.52 (s, 3H), 3.24 (s, 3H), 2.81 - 2.74 (m, 2H), 2.55 (s, 3H), 2.17 - 2.06 (m, 4H). Synthesis of Compound 250 Exemplary Synthesis of 7-((3R,4S)-4-((4-chloro-2-methoxyphenyl)amino)-3- methylpiperidin-1-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 7-((3S,4R)-4-((4-chloro-2- methoxyphenyl)amino)-3-methylpiperidin-1-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4- b]pyridine-6-carbonitrile starting from (3R,4S)-N-(4-chloro-2-methoxy-phenyl)-3-methyl- piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 457.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.87 (d, J = 2.0 Hz, 1H), 6.84 - 6.78 (m, 1H), 6.69 (d, J = 8.4 Hz, 1H), 4.60 (m, J = 8.4 Hz, 1H), 4.24 - 4.06 (m, 2H), 3.82 (s, 3H), 3.81 - 3.79 (m, 1H), 3.78 (m, J = 4.0 Hz, 1H), 3.62 (m, J = 3.2 Hz, 1H), 3.48 (s, 3H), 2.70 (s, 3H), 2.29 (d, J = 4.8 Hz, 1H), 2.09 - 1.98 (m, 1H), 1.88 - 1.73 (m, 1H), 0.78 (d, J = 6.8 Hz, 3H). Synthesis of Compound 251 Exemplary Synthesis of 7-[(3R,4S)-4-[(4-chloro-2-fluorophenyl)amino]-3- methylpiperidin-1-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloro-2-fluoro- anilino)-3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3R,4S)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4-amine. (off-white solid). LC/MS (ESI) m/z: 446.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.19 (dd, J = 2.4, 11.6 Hz, 1H), 7.02 (dd, J = 1.2, 8.4 Hz, 1H), 6.89 (t, J = 9.2 Hz, 1H), 5.42 (dd, J = 2.0, 8.4 Hz, 1H), 4.29 - 4.14 (m, 2H), 3.85 - 3.74 (m, 2H), 3.67 - 3.56 (m, 1H), 3.48 (s, 3H), 2.70 (s, 3H), 2.26 (dd, J = 1.2, 2.8 Hz, 1H), 2.12 (dd, J = 4.0, 9.6 Hz, 1H), 1.82 - 1.74 (m, 1H), 0.81 (d, J = 6.8 Hz, 3H). Synthesis of Compound 252 Exemplary Synthesis of 6-chloro-1-methyl-4-((3R,4S)-3-methyl-4-(methyl(4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile Step 1: Preparation of tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate and tert-butyl (3R,4S)-3-methyl-4- [N-methyl-4-(trifluoromethoxy)anilino]piperidine-1-carboxylate Racemic tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (600 mg, 1.54 mmol) was purified by SFC, then further purified by prep-SFC (column: DAICEL CHIRALPAK AD (250mm*30mm, 10um); mobile phase: [0.1% ammonium hydroxide﹒methyl alcohol]; B%: 10%-10%, 5.0;190 min). tert-Butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (200 mg, 33%) was obtained as gray oil. LC/MS (ESI) m/z: 389.5 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.15 ( d, J = 8.8 Hz, 2H), 6.79 (d, J = 9.2 Hz, 2H), 4.24 - 4.06 (m, 1H), 3.93 - 3.71 (m, 2H), 3.21 - 2.95 (m, 1H), 2.80 (s, 3H), 2.53 - 2.52 (m, 1H), 2.14 (s, 1H), 2.09 - 2.00 (m, 1H), 1.50 (d, J = 11.2 Hz, 1H), 1.40 (s, 9H), 0.82 (d, J = 6.8 Hz, 3H). tert-Butyl (3R,4S)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1- carboxylate (300 mg, 50%) was obtained as a gray oil. LC/MS (ESI) m/z: 389.4 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.11 (d, J = 8.8 Hz, 2H), 6.69 (d, J = 9.2 Hz, 2H), 4.44 - 4.18 (m, 1H), 4.09 - 3.87 (m, 1H), 3.75 (td, J = 3.6, 12.0 Hz, 1H), 3.07 - 2.91 (m, 1H), 2.86 (s, 3H), 2.82 - 2.67 (m, 1H), 2.25 (s, 1H), 2.16 - 2.06 (m, 1H), 1.60 - 1.56 (m, 1H), 1.48 (s, 9H), 0.94 (d, J = 7.2 Hz, 3H). step 2: preparation of 6-chloro-1-methyl-4-((3R,4S)-3-methyl-4-(methyl(4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile
Figure imgf000491_0001
The title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3S,4R)-3- methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3R,4S)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 492.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 7.17 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 9.2 Hz, 2H), 4.38 - 4.31 (m, 1H), 4.20 - 4.12 (m, 1H), 4.03 - 3.96 (m, 1H), 3.91 - 3.85 (m, 1H), 3.54 (s, 3H), 3.45 (t, J = 12.0 Hz, 1H), 2.87 (s, 3H), 2.78 - 2.69 (m, 1H), 2.35 (dd, J = 3.2, 5.6 Hz, 1H), 1.77 (d, J = 10.0 Hz, 1H), 0.89 (d, J = 7.2 Hz, 3H). Synthesis of Compound 253 Exemplary Synthesis of 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of 3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine A mixture of tert-butyl 3-methyl-4-[4-(trifluoromethoxy)anilino]piperidine-1-carboxylate (1.82 g, 4.87 mmol, 1 eq) and HCl in dioxane (4 M, 6.33 mL, 5.2 eq) in dichloromethane (20 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 1 h under nitrogen atmosphere. The reaction was concentrated, and the resulting residue was poured into ice and saturated sodium bicarbonate solution (10 mL). The mixture was extracted with ethyl acetate (15 mL x 2), and the combined organic layers were washed with brine (30 mL), dried over sodium sulfate, and concentrated under reduced pressure to afford 3-methyl-N-[4- (trifluoromethoxy)phenyl]piperidin-4-amine (1.45 g, crude) as a yellow oil. LC/MS (ESI) m/z: 275.2 [M+H]+. Step 2: Preparation of (3S,4R)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4- amine and (3R,4S)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine Racemic 3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (1.5 g, 5.47 mmol) was purified by SFC (column: DAICEL CHIRALPAK IG(250mm*50mm,10um); mobile phase: [0.1 % ammonium hydroxide ethyl alcohol]; B%: 35%-35%,15;180 min), then further purified by prep-HPLC (column: Phenomenex luna C18150*25 mm* 10 um; mobile phase: [water formic acid-acetonitrile]; B%: 15%-45 %,10 min), and then SFC (column: DAICEL CHIRALPAK AD(250 mm*30 mm,10 um); mobile phase: [0.1 % ammonium hydroxide isopropyl alcohol]; B%: 15%-15%,11.55;427 min). (3S,4R)-3-Methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (479 mg, 1.75 mmol, 31 % yield) was obtained as a white solid.1H NMR (400 MHz, CDCl3) δ 7.02 ( d, J = 8.4 Hz, 2H), 6.57 (d, J = 8.8 Hz, 2H), 3.75 - 3.65 (m, 1H), 3.55 ( s, 1H), 3.06 - 2.96 (m, 1H), 2.92 - 2.85 (m, 1H), 2.83 - 2.72 (m, 2H), 2.18 - 2.09 (m, 1H), 1.73 - 1.65 (m, 2H), 1.29 - 1.23 (m, 1H), 0.97 (d, J = 7.2 Hz, 3H). (3R,4S)-3-Methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (276 mg, 1.01 mmol, 18 % yield) was obtained as a white solid.1H NMR (400 MHz, CDCl3) δ 6.94 (d, J = 8.4 Hz, 2H), 6.51 - 6.47 (m, 2H), 3.63 ( s, 1H), 3.47 (s, 1H), 2.92 (J = 5.2, 12.4 Hz, 1H), 2.82 - 2.75 (m, 1H), 2.74 - 2.65 (m, 2H), 2.08 - 2.00 (m, 1H), 1.76 - 1.68 (m, 1H), 1.64 - 1.56 (m, 2H), 0.88 (d, J = 7.2 Hz, 3H). Step 3: Preparation of 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from (3S,4R)-3- methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 492.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 7.05 (d, J = 8.8 Hz, 2H), 6.81 - 6.69 (m, 2H), 5.99 (d, J = 8.4 Hz, 1H), 4.06 - 3.96 (m, 1H), 3.90 - 3.83 (m, 2H), 3.79 ( J = 3.6, 7.8 Hz, 1H), 3.72 - 3.64 (m, 1H), 3.53 (s, 3H), 2.32 - 2.24 (m, 1H), 2.14 - 2.03 (m, 1H), 1.90 - 1.81 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H). Synthesis of Compound 254 Exemplary Synthesis of 2,4-dimethyl-7-[(3S,4R)-3-methyl-4-{[4- (trifluoromethoxy)phenyl]amino}piperidin-1-yl]-5-oxo-4H,5H-[1,3]thiazolo[5,4- b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3S,4R)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 478.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.04 (d, J = 8.8 Hz, 2H), 6.73 (d, J = 9.2 Hz, 2H), 5.94 (d, J = 8.4 Hz, 1H), 4.05 (dd, J = 5.6, 13.2 Hz, 2H), 3.84 (dd, J = 2.8, 13.2 Hz, 1H), 3.79 - 3.70 (m, 2H), 3.48 (s, 3H), 2.69 (s, 3H), 2.29 - 2.21 (m, 1H), 2.04 - 1.92 (m, 1H), 1.84 - 1.73 (m, 1H), 0.83 (d, J = 6.8 Hz, 3H). Synthesis of Compound 255 Exemplary Synthesis of 7-((3S,4R)-4-((4-chlorophenyl)amino)-3-methylpiperidin-1-yl)- 2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate and tert-butyl (3R,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate Racemic tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (500 mg, 1.54 mmol) was purified by SFC, then further purified by prep- SFC (column: DAICEL CHIRALCEL OD(250mm*30mm,10um); mobile phase: [0.1% ammonium hydroxide IPA]; B%: 20%-20%, 8.0; 128 min). tert-Butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (240 mg, 48%) was obtained as colorless oil. LC/MS (ESI) m/z: 269.0 [M-55]+. tert-Butyl (3R,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (230 mg, 46%) was obtained as a colorless oil. LC/MS (ESI) m/z: 269.0 [M-55]+. Step 2: Preparation of (3S,4R)-N-(4-chlorophenyl)-3-methyl-piperidin-4-amine To a solution of tert-butyl (3S,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (100 mg, 0.31 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 21.94 eq), and the reaction mixture was stirred at 25 °C for 0.25 h. The mixture was concentrated under reduced pressure to give (3S,4R)-N-(4-chlorophenyl)-3- methyl-piperidin-4-amine trifluoroacetic acid (100 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 225.0 [M+H]+. Step 3: Preparation of 7-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile To a solution of (3S,4R)-N-(4-chlorophenyl)-3-methyl-piperidin-4-amine trifluoroacetic acid (100 mg, 0.30 mmol, 1 eq) in acetonitrile (1 mL) was added diisopropylethylamine (191 mg, 1.48 mmol, 0.26 mL, 5 eq) and (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate (104 mg, 0.30 mmol, 1 eq), and the reaction mixture was stirred at 40 °C for 12 h. The mixture was suspended in DMF/acetonitrile = 1:5 (1.8 mL) and collected by filtration to give 7-[(3S,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-2,4-dimethyl-5- oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (75 mg, 59% ) as a white solid. LC/MS (ESI) m/z: 428.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.07 (d, J = 8.8 Hz, 2H), 6.69 (d, J = 8.8 Hz, 2H), 5.84 (d, J = 8.8 Hz, 1H), 4.07 (dd, J = 5.6, 12.8 Hz, 2H), 3.83 (dd, J = 2.4, 13.2 Hz, 1H), 3.72 (dd, J = 4.0, 7.6 Hz, 2H), 3.47 (s, 3H), 2.69 (s, 3H), 2.28 - 2.20 (m, 1H), 2.03 - 1.92 (m, 1H), 1.81 - 1.79 (m, 1H), 0.82 (d, J = 6.8 Hz, 3H). Synthesis of Compound 256 Exemplary Synthesis of 6-chloro-1-methyl-4-((3S,4R)-3-methyl-4-(methyl(4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3S,4R)-3- methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3- carbonitrile starting from tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 506.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 9.2 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 7.17 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 9.2 Hz, 2H), 4.34 (d, J = 11.6 Hz, 1H), 4.20 - 4.13 (m, 1H), 4.04 - 3.96 (m, 1H), 3.92 - 3.85 (m, 1H), 3.53 (s, 3H), 3.45 (t, J = 12.0 Hz, 1H), 2.87 (s, 3H), 2.78 - 2.69 (m, 1H), 2.40 - 2.34 (m, 1H), 1.77 (d, J = 10.4 Hz, 1H), 0.89 (d, J = 6.8 Hz, 3H). Synthesis of Compound 257 Exemplary Synthesis of 6-chloro-1-methyl-4-[(3R,4S)-3-methyl-4-{[4- (trifluoromethoxy)phenyl]amino}piperidin-1-yl]-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile
The title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3S,4R)-3- methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,4S)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (yellow solid). LC/MS (ESI) m/z: 492.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.05 (d, J = 8.8 Hz, 2H), 6.75 (d, J = 9.2 Hz, 2H), 5.99 (d, J = 8.8 Hz, 1H), 4.07 - 3.94 (m, 1H), 3.86 (s, 2H), 3.81 - 3.74 (m, 1H), 3.73 - 3.63 (m, 1H), 3.53 (s, 3H), 2.32 (d, J = 1.6 Hz, 1H), 2.12 - 2.02 (m, 1H), 1.86 (J = 2.8, 7.1 Hz, 1H), 0.87 (d, J = 6.8 Hz, 3H). Synthesis of Compound 258 Exemplary Synthesis of 7-[(3R,4S)-4-[(4-chlorophenyl)amino]-3-methylpiperidin-1-yl]- 2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloroanilino)- 3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3R,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate. (white solid). LC/MS (ESI) m/z: 428.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.07 (d, J = 8.8 Hz, 2H), 6.69 (d, J = 8.8 Hz, 2H), 5.85 (d, J = 8.8 Hz, 1H), 4.07 (dd, J = 5.2, 12.8 Hz, 2H), 3.83 (dd, J = 2.4, 13.2 Hz, 1H), 3.77 - 3.67 (m, 2H), 3.47 (s, 3H), 2.69 (s, 3H), 2.28 - 2.20 (m, 1H), 2.03 - 1.92 (m, 1H), 1.82 - 1.74 (m, 1H), 0.82 (d, J = 6.8 Hz, 3H). Synthesis of Compound 259 Exemplary Synthesis of 6-chloro-1-methyl-2-oxo-4-((3aS,7aR)-1-(4- (trifluoromethoxy)phenyl)octahydro-5H-pyrrolo[3,2-c]pyridin-5-yl)-1,2-dihydro-1,5- naphthyridine-3-carbonitrile Step 1: Preparation of O5-benzyl O1-tert-butyl (3aR,7aS)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-1,5-dicarboxylate To a solution of tert-butyl (3aR,7aS)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine-1- carboxylate (1 g, 4.42 mmol, 1 eq) in dichloromethane (20 mL) was added triethylamine (894 mg, 8.84 mmol, 1.23 mL, 2 eq) followed by benzyl carbonochloridate (1.13 g, 6.63 mmol, 0.942 mL, 1.5 eq) dropwise under nitrogen atmosphere, and the reaction mixture was stirred at 25 °C for 12 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18 250×50mm×5um;mobile phase: [water(formic acid)- acetonitrile]; B%: 30%-60%, 35min) to give O5-benzyl O1-tert-butyl (3aR,7aS)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine- 1,5-dicarboxylate (1.78 g, crude) as a colorless oil. LC/MS (ESI) m/z: 304.9 [M-55] +; 1H NMR (400 MHz, DMSO-d6) δ 7.43 - 7.27 (m, 5H), 5.07 (s, 2H), 3.77 (s, 3H), 3.30 - 3.15 (m, S3H), 2.99 - 2.77 (m, 1H), 2.26 (s, 1H), 1.96 - 1.86 (m, 1H), 1.76 (d, J = 1.2 Hz, 1H), 1.71 - 1.59 (m, 1H), 1.43 - 1.40 (m, 1H), 1.38 (s, 9H). Step 2: Preparation of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2- c]pyridine-5-carboxylate To a solution of O5-benzyl O1-tert-butyl (3aR,7aS)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-1,5-dicarboxylate (400.00 mg, 1.11 mmol, 1 eq) in dichloromethane (4 mL) was added trifluoroacetic acid (6.16 g, 54.03 mmol, 4.00 mL, 48.68 eq) at 25 °C, and the reaction mixture was stirred at 25 °C for 1 h. The mixture was concentrated under reduced pressure to afford benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5-carboxylate trifluoroacetic acid (0.65 g, crude) as a brown oil. LC/MS (ESI) m/z: 261.1 [M+H]+. Step 3: Preparation of benzyl (3aR,7aS)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate and benzyl (3aS,7aR)-1-[4- (trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate To a solution of 1-bromo-4-(trifluoromethoxy)benzene (1.85 g, 7.68 mmol, 1.14 mL, 2.5 eq) in dioxane (8 mL) and benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate (800.00 mg, 3.07 mmol, 1 eq) were added cesium carbonate (3.00 g, 9.22 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (149 mg, 0.15 mmol, 0.05 eq) at 25°C, and the resulting mixture was degassed and purged with nitrogen (3X). The reaction mixture was then stirred at 100 °C for 3 h under nitrogen atmosphere. The reaction was diluted with water (200 mL) and extracted with ethyl acetate (200 mL × 3). The combined organic layers were washed by brine and dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by semi-preparative reverse phase HPLC ([water(ammonium bicarbonate)-acetonitrile];B%: 57% - 87%,8 min). The selected fraction was concentrated to remove most of the acetonitrile, then extracted with ethyl acetate (100 mL × 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated to afford the racemic product. LC/MS (ESI) m/z: 421.1 [M+H]+; 1H NMR (400MHz, DMSO- d6) δ 7.30 - 7.50 (m, 5H), 7.08 (br d, J = 8.63 Hz, 2H), 6.52 (br d, J=9.01 Hz, 2H), 5.09 - 5.23 (m, 2H), 4.13 (q, J = 7.13 Hz, 1H), 3.89 - 4.08 (m, 2H), 3.79 - 3.88 (m, 1H), 3.33 - 3.46 (m, 2H), 3.27 (q, J=8.63 Hz, 1H), 2.90 - 3.09 (m, 1H), 2.35 - 2.54 (m, 1H). The product was further purified by SFC (column: DAICEL CHIRALPAK AD (250mm×30mm,10um); mobile phase: [0.1% ammonium hydroxide isopropanol]; B%: 30%-30%,6.10; 128 min). Benzyl (3aR,7aS)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (0.35 g, 27 %) was obtained as a colorless oil. Benzyl (3aS,7aR)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (0.32 g, 24 %) was obtained as a colorless oil. Step 4: Preparation of (3aS,7aR)-1-(4-(trifluoromethoxy)phenyl)octahydro-1H- pyrrolo[3,2-c]pyridine To a solution of benzyl (3aS,7aR)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-5-carboxylate (0.32 g, 0.76 mmol, 1 eq) in tetrahydrofuran (5 mL) was added 10% palladium on carbon (0.3 g, 0.76 mmol, 1 eq) at 25 °C, and the resulting mixture was degassed and purged with hydrogen (3X). The reaction mixture was then stirred at 25°C for 2 h under hydrogen atmosphere. The reaction mixture was filtered, then concentrated under reduced pressure to afford (3aS,7aR)-1-(4- (trifluoromethoxy)phenyl)octahydro-1H-pyrrolo[3,2-c]pyridine (0.293 g, crude) as a yellow oil. LC/MS (ESI) m/z: 287.2 [M+H]+. Step 5: Preparation of 4-[(3aS,7aR)-1-[4-(trifluoromethoxy)phenyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from (3aS,7aR)- 1-(4-(trifluoromethoxy)phenyl)octahydro-1H-pyrrolo[3,2-c]pyridine. (yellow solid). LC/MS (ESI) m/z: 504.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.10 - 7.19 (m, 2H), 6.66 – 6.61 (m, 2H), 4.20 - 4.27 (m, 1H), 4.09 - 4.16 (m, 1H), 4.02 - 4.09 (m, 1H), 3.99 - 4.17 (m, 1 H), 3.93 -3.91 (m, 1H), 3.53 (s, 3H), 3.44 - 3.51 (m, 1H), 3.34 - 3.44 (m, 1H), 3.34 - 3.35 (m, 1H), 3.22 - 3.28 (m, 1H), 2.31 - 2.36 (m, 1H), 2.17 - 2.36 (m, 1H), 2.06 - 2.15 (m, 2H), 1.76 - 1.87 (m, 1H). Synthesis of Compound 260 Exemplary Synthesis of 2,4-dimethyl-5-oxo-7-((3aS,7aR)-1-(4- (trifluoromethoxy)phenyl)octahydro-5H-pyrrolo[3,2-c]pyridin-5-yl)-4,5- dihydrothiazolo[5,4-b]pyridine-6-carbonitrile
Figure imgf000501_0001
The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aS,7aR)-1-(4-(trifluoromethoxy)phenyl)octahydro-1H-pyrrolo[3,2-c]pyridine. (gray solid). LC/MS (ESI) m/z: 490.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.13 (d, J = 8.4 Hz, 2H), 6.63 (d, J = 9.2 Hz, 2H), 4.34 - 4.42 (m, 1H), 4.21 - 4.30 (m, 1H), 3.91 - 4.06 (m, 2H), 3.48 (s, 3H), 3.45 (br s, 1H), 3.17 - 3.26 (m, 1H), 2.69 (s, 3H), 2.56 - 2.65 (m, 2H), 2.16 - 2.26 (m, 1H), 2.00 - 2.09 (m, 1H), 1.90 - 2.00 (m, 1H), 1.65 - 1.77 (m, 1H).
Synthesis of Compound 261 Exemplary Synthesis of 6-chloro-4-{4-[(5-chloropyridin-2-yl)methyl]-4- hydroxypiperidin-1-yl}-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl 4-((5-chloropyridin-2-yl)methyl)-4-hydroxypiperidine- 1-carboxylate To a solution of 5-chloro-2-methyl-pyridine (100 mg, 0.78 mmol, 1.0 eq) in tetrahydrofuran (1 mL) was added n-butyllithium (2.5 M, 0.37 mL, 1.2 eq) dropwise at -78 °C for 0.5 h. tert- Butyl 4-oxopiperidine-1-carboxylate (156 mg, 0.78 mmol, 1.0 eq) in tetrahydrofuran (1 mL) was then added dropwise at -78°C, and the reaction mixture was stirred at -78°C for 0.5 h. The reaction was diluted with saturated aqueous ammonium chloride solution (15 mL) and water (15 mL), and the resulting mixture was extracted with ethyl acetate (10 mL × 3). The combined organic extracts were washed by brine and dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by pre-TLC (silica, 50% ethyl acetate in petroleum ether, Rf = 0.2) to afford tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4- hydroxy-piperidine-1-carboxylate (84 mg, 33%) as a white solid. LC/MS (ESI) m/z: 326.9 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.47 (d, J = 2.0 Hz, 1H), 7.62 (dd, J = 8.0, 2.40 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 5.24 (br s, 1H), 3.79 (br s, 2H), 3.21 (br s, 2H), 2.88 (s, 2H), 1.49 (s, 4H), 1.44 (s, 9H). Step 2: Preparation of 4-((5-chloropyridin-2-yl)methyl)piperidin-4-ol To a solution of tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4-hydroxy-piperidine-1- carboxylate (84 mg, 0.26 mmol, 1.0 eq) in dichloromethane (3 mL) was added trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 52.5 eq), and the reaction mixture was stirred at 25 °C for 1 h. The mixture was concentrated under vacuum to afford 4-[(5-chloro-2- pyridyl)methyl]piperidin-4-ol trifluoroacetic acid (189 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 227.1 [M+H]+. Step 3: Preparation of 6-chloro-4-(4-((5-chloropyridin-2-yl)methyl)-4-hydroxypiperidin- 1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS (ESI) m/z: 444.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.54 (d, J = 2.0 Hz, 1H), 8.04 (d, J = 9.2 Hz, 1H), 7.86 (dd, J = 8.4, 2.8 Hz, 1H), 7.78 (d, J = 9.2 Hz, 1H), 7.39 (d, J = 8.4 Hz, 1H), 4.86 (s, 1H), 3.95 (br d, J = 12.8 Hz, 2H), 3.69 (br t, J = 11.2 Hz, 2H), 3.51 (s, 3H), 2.95 (s, 2H), 1.93 - 1.79 (m, 2H), 1.63 (br d, J = 13.6 Hz, 2H). Synthesis of Compound 262 Exemplary Synthesis of 7-[(3aR,7aS)-1-(4-fluorophenyl)-octahydro-1H-pyrrolo[3,2- c]pyridin-5-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of benzyl (3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate and benzyl (3aS,7aR)-1-(4-fluorophenyl)- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate A mixture of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate trifluoroacetic acid (420 mg, 1.12 mmol, 1 eq), 1-bromo-4-fluoro-benzene (294 mg, 1.68 mmol, 0.185 mL, 1.5 eq), cesium carbonate (1.46 g, 4.49 mmol, 4 eq) and 1,3- bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (55 mg, 0.056 mmol, 0.05 eq) in dioxane (5 mL) was stirred at 100 °C for 3 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (petroleum ether/ethyl acetate = 100/1 to 1:1), then further separated by SFC (column: DAICEL CHIRALCEL OJ (250mm*30mm, 10 um); mobile phase: [0.1% ammonium hydroxide methanol]; B%: 40%-40%, 5.4; 65 min). Benzyl (3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate (88 mg, 21%) was obtained as colorless oil. LC/MS (ESI) m/z: 355.0 [M+H]+. Benzyl (3aS,7aR)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate (80 mg, 19%) was obtained as colorless oil. LC/MS (ESI) m/z: 355.1 [M+H]+. Step 2: Preparation of (3aR,7aS)-1-(4-fluorophenyl)octahydro-1H-pyrrolo[3,2- c]pyridine To a suspension of 10% palladium on carbon (50 mg, 0.23 mmol, 1 eq) in methanol (5 mL) was added benzyl (3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (80 mg, 0.226 mmol, 1 eq) under nitrogen atmosphere. The suspension was degassed under vacuum and purged with hydrogen several times. The mixture was then stirred under hydrogen (15 Psi) at 25 °C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give (3aR,7aS)-1-(4- fluorophenyl)octahydro-1H-pyrrolo[3,2-c]pyridine (40 mg, crude) as a white solid. LC/MS (ESI) m/z: 221.0 [M+H]+. Step 3: Preparation of 7-[(3aR,7aS)-1-(4-fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloroanilino)- 3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aR,7aS)-1-(4-fluorophenyl)octahydro-1H-pyrrolo[3,2-c]pyridine. (yellow solid). LC/MS (ESI) m/z: 423.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.99 (t, J = 8.8 Hz, 2H), 6.62 - 6.54 (m, 2H), 4.33 (dd, J = 2.4, 13.6 Hz, 1H), 4.27 - 4.18 (m, 1H), 4.00 - 3.91 (m, 2H), 3.54 - 3.43 (m, 4H), 3.38 - 3.33 (m, 1H), 3.17 (q, J = 8.8 Hz, 1H), 2.68 (s, 3H), 2.66 - 2.58 (m, 1H), 2.23 - 2.13 (m, 1H), 2.08 - 1.98 (m, 1H), 1.97 - 1.85 (m, 1H), 1.76 - 1.65 (m, 1H). Synthesis of Compound 263 Exemplary Synthesis of 6-chloro-1-methyl-4-((3S,4R)-3-methyl-4-((4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydroquinoline-3- carbonitrile Step 1: Preparation of methyl 5-chloro-2-[(2-cyanoacetyl)amino]benzoate A mixture of methyl 2-amino-5-chloro-benzoate (1 g, 5.39 mmol, 1 eq), 2-cyanoacetic acid (504.12 mg, 5.93 mmol, 1.1 eq), diisopropylethylamine (2.09 g, 16.16 mmol, 2.82 mL, 3 eq) in DMF (10 mL) and 50% propylphosphonic anhydride (6.86 g, 10.78 mmol, 6.41 mL, 2 eq) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 2 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by column chromatography (petroleum ether/ethyl acetate = 5/1 to 3/1) to afford methyl 5-chloro-2-[(2-cyanoacetyl)amino]benzoate (1.3 g, 95%) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 10.64 (s, 1H), 8.02 (d, J = 8.8 Hz, 1H), 7.86 (d, J = 2.4 Hz, 1H), 7.71 (m, J = 2.6, 8.9 Hz, 1H), 3.86 (s, 3H), 3.87 - 3.84 (m, 1H). Step 2: Preparation of ( 3S,4R)-N-(4-fluorophenyl)-3-methyl-piperidin-4-amine A mixture of methyl 5-chloro-2-[(2-cyanoacetyl)amino]benzoate (4.7 g, 18.60 mmol, 1 eq) and 1,8-diazabicyclo[5.4.0]undec-7-ene (4.25 g, 27.90 mmol, 4.21 mL, 1.5 eq) in ethanol (40 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 2 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure. The mixture was quenched by 1 N hydrogen chloride, the pH adjusted to 5, and then filtered. The filtrate cake was collected to give 6-chloro-4-hydroxy-2-oxo-1H-quinoline-3-carbonitrile (2.95 g, 72%) as a yellow solid.1H NMR (400 MHz, CDCl3) δ 11.63 (s, 1H), 8.62 (d, J = 9.2 Hz, 1H), 8.06 (d, J = 2.4 Hz, 1H), 7.55 (m, J = 2.6, 9.1 Hz, 1H), 3.99 (s, 4H). Step 3: Preparation of 6-chloro-4-hydroxy-1-methyl-2-oxo-quinoline-3-carbonitrile To a mixture of 6-chloro-4-hydroxy-2-oxo-1H-quinoline-3-carbonitrile (2.95 g, 13.37 mmol, 1 eq) in DMF (15 mL) was added 60% NaH (1.34 g, 33.43 mmol, 2.5 eq), and the resulting mixture was stirred at 0 °C for 0.5 h. Iodomethane (5.69 g, 40.12 mmol, 2.50 mL, 3 eq) was then added, and the reaction mixture was stirred at 0 °C for 0.5 h under nitrogen. The mixture was quenched by 1 N HCl and the pH adjusted to 5. The resulting mixture was filtered, and the filtrate cake was collected. The crude product was triturated with water (5 mL) and acetonitrile (15 mL). After filtration, the solid was dried in vacuum to afford 6-chloro-4- hydroxy-1-methyl-2-oxo-quinoline-3-carbonitrile (2.2 g, 70%) as a white solid. LC/MS (ESI) m/z: 234.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.02 - 7.95 (m, 1H), 7.66 (m, J = 2.4, 8.8 Hz, 1H), 7.45 (d, J = 9.2 Hz, 1H), 3.48 (s, 3H). Step 4: Preparation of 4,6-dichloro-1-methyl-2-oxo-quinoline-3-carbonitrile To a mixture of 6-chloro-4-hydroxy-1-methyl-2-oxo-quinoline-3-carbonitrile (2.2 g, 9.38 mmol, 1 eq) and benzyl(triethyl)ammonium;chloride (4.27 g, 18.75 mmol, 2 eq) in acetonitrile (30 mL) were added diisopropylethylamine (7.27 g, 56.26 mmol, 9.80 mL, 6 eq) and phosphoryl chloride (7.19 g, 46.88 mmol, 4.36 mL, 5 eq) in one portion at 20 °C under nitrogen, and the reaction mixture was stirred at 25 °C for 4 h. The mixture was concentrated, and the residue was poured into ice and saturated aqueous NaHCO3 solution (10 mL). The resulting mixture was extracted with ethyl acetate (15 mL x 2), and the combined organic extracts were washed with brine (30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was triturated with acetonitrile (25 mL) and water (30 mL) to afford 4,6-dichloro-1-methyl-2-oxo-quinoline-3-carbonitrile (2.1 g, 88%) as a white solid. LC/MS (ESI) m/z: 252.8 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 2.4 Hz, 1H), 7.96 (m, J = 2.4, 9.2 Hz, 1H), 7.77 (d, J = 9.2 Hz, 1H), 3.66 (s, 3H). Step 5: Preparation of 6-chloro-1-methyl-4-((3S,4R)-3-methyl-4-((4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydroquinoline-3- carbonitrile A mixture of (3S,4R)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (60 mg, 0.22 mmol, 1 eq), 4,6-dichloro-1-methyl-2-oxo-quinoline-3-carbonitrile (55 mg, 0.22 mmol, 1 eq), and diisopropylethylamine (85 mg, 0.66 mmol, 114.31 uL, 3 eq) in acetonitrile (1 mL) was degassed and purged with nitrogen (3X), then stirred at 40 °C for 4 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex C18150*25mm*10um; mobile phase: [water(ammonium hydrogen carbonate)-acetonitrile]; B%: 55%-85%, 8min) to afford 6-chloro-1-methyl-4-[(3S,4R)-3-methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo- quinoline-3-carbonitrile (65 mg, 60%)as a white solid. LC/MS (ESI) m/z: 491.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.81 - 7.74 (m, 2H), 7.65 - 7.55 (m, 1H), 7.06 (d, J = 8.8 Hz, 2H), 6.77 (s, 1H), 6.00 (d, J = 8.4 Hz, 1H), 3.79 - 3.74 (m, 2H), 3.61 (d, J = 7.6 Hz, 1H), 3.56 (s, 3H), 3.51 - 3.46 (m, 2H), 2.08 - 1.97 (m, 2H), 1.90 - 1.85 (m, 1H), 1.18 - 1.12 (m, 1H), 0.91 (d, J = 6.8 Hz, 3H). Synthesis of Compound 264 Exemplary Synthesis of 7-((3S,4S)-4-((4-chloro-2-fluorophenyl)amino)-3- methylpiperidin-1-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloro-2-fluoro- anilino)-3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3S,4S)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 445.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.16 (dd, J = 2.4, 11.6 Hz, 1H), 7.04 - 6.98 (m, 1H), 6.87 (t, J = 9.2 Hz, 1H), 5.54 - 5.45 (m, 1H), 4.55 - 4.40 (m, 2H), 3.52 - 3.44 (m, 4H), 3.18 - 3.05 (m, 2H), 2.69 (s, 3H), 2.06 - 1.99 (m, 2H), 1.60 - 1.48 (m, 1H), 0.94 (d, J = 6.4 Hz, 3H).
Synthesis of Compound 265 Exemplary Synthesis of 7-[(3R,4R)-4-[(4-chlorophenyl)amino]-3-methylpiperidin-1-yl]- 2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate and tert-butyl (3S,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1- carboxylate Racemic tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (400 mg, 1.23 mmol) was purified by SFC then further purified by prep-SFC (column: DAICEL CHIRALCEL OD (250mm*30mm,10um); mobile phase: [0.1% ammonium hydroxide in methyl alcohol]; B%: 25%-25%, 6.7;87 min). tert-Butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (200 mg, 49%) was obtained as a colorless oil. LC/MS (ESI) m/z: 325.1 [M+H]+. tert-Butyl (3S,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate (170 mg, 42%) was obtained as a colorless oil. LC/MS (ESI) m/z: 325.1 [M+H]+. Step 2: Preparation of 7-[(3R,4R)-4-(4-chloroanilino)-3-methyl-1-piperidyl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloroanilino)- 3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3R,4R)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate. (white solid). LC/MS (ESI) m/z: 428.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.06 (d, J = 8.8 Hz, 2H), 6.62 (d, J = 8.8 Hz, 2H), 5.74 (d, J = 8.8 Hz, 1H), 4.53 - 4.33 (m, 2H), 3.56 - 3.49 (m, 1H), 3.48 (s, 3H), 3.27 - 3.21 (m, 1H), 3.14 (dd, J = 10.8, 13.2 Hz, 1H), 2.69 (s, 3H), 2.13 - 2.06 (m, 1H), 1.92 - 1.81 (m, 1H), 1.47 - 1.35 (m, 1H), 0.96 (d, J = 6.4 Hz, 3H). Synthesis of Compound 266 Exemplary Synthesis of 2,4-dimethyl-7-[(3R,4S)-3-methyl-4-{[4- (trifluoromethoxy)phenyl]amino}piperidin-1-yl]-5-oxo-4H,5H-[1,3]thiazolo[5,4- b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3R,4S)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 478.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.04 (d, J = 8.8 Hz, 2H), 6.73 (d, J = 9.2 Hz, 2H), 5.95 (d, J = 8.4 Hz, 1H), 4.05 (dd, J = 6.0, 13.2 Hz, 2H), 3.84 (dd, J = 3.2, 13.2 Hz, 1H), 3.80 - 3.69 (m, 2H), 3.48 (s, 3H), 2.69 (s, 3H), 2.26 - 2.24 (m, 1H), 2.00 - 1.95 (m, 1H), 1.84 - 1.78 (m, 1H), 0.83 (d, J = 6.8 Hz, 3H).
Synthesis of Compound 267 Exemplary Synthesis of 7-((3S,4S)-4-((4-chlorophenyl)amino)-3-methylpiperidin-1-yl)- 2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloroanilino)- 3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3S,4S)-4-(4-chloroanilino)-3-methyl-piperidine-1-carboxylate. (white solid). LC/MS (ESI) m/z: 428.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.06 (d, J = 8.8 Hz, 2H), 6.62 (d, J = 8.8 Hz, 2H), 5.74 (d, J = 8.8 Hz, 1H), 4.50 - 4.36 (m, 2H), 3.55 - 3.49 (m, 1H), 3.48 (s, 3H), 3.28 - 3.20 (m, 1H), 3.14 (dd, J = 11.2, 13.2 Hz, 1H), 2.69 (s, 3H), 2.13 - 2.06 (m, 1H), 1.93 - 1.80 (m, 1H), 1.48 - 1.35 (m, 1H), 0.96 (d, J = 6.4 Hz, 3H). Synthesis of Compound 268 Exemplary Synthesis of 1,6-dimethyl-4-[(3S,4R)-3-methyl-4-[4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of (3-cyano-1,6-dimethyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate To a mixture of 4-hydroxy-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile (1.2 g, 5.58 mmol, 1 eq), trimethylamine (1.86 g, 18.35 mmol, 2.55 mL, 3.29 eq), and 4- dimethylaminopyridine (75 mg, 0.61 mmol, 0.11 eq) in dichloromethane (15 mL) at 0°C was added triflic anhydride (3.45 g, 12.23 mmol, 2.02 mL, 2.19 eq), and the reaction mixture was stirred at 0 °C for 1 h under nitrogen. The mixture diluted with water (60 mL) and extracted with dichloromethane (60 mL x 2). The combined organic extracts were washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate = 100/1 to 1:1) to give (3-cyano-1,6-dimethyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (650 mg, 34%) as a yellow solid. LC/MS (ESI) m/z: 347.8 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.20 (d, J = 8.8 Hz, 1H), 7.87 - 7.83 (m, 1H), 3.67 (s, 3H), 2.62 (s, 3H). Step 2: Preparation of 1,6-dimethyl-4-[(3S,4R)-3-methyl-4-[4- (trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile A mixture of (3S,4R)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine (60 mg, 0.22 mmol, 1 eq), (3-cyano-1,6-dimethyl-2-oxo-1,5-naphthyridin-4-yl) trifluoromethanesulfonate (76 mg, 0.22 mmol, 1 eq), and diisopropylethylamine (85 mg, 0.66 mmol, 0.12 mL, 3 eq) in acetonitrile (2 mL) was degassed and purged with nitrogen (3X), then stirred at 40 °C for 12 h under nitrogen atmosphere The reaction was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex luna C18 150*25 mm* 10 um; mobile phase: [water formic acid-acetonitrile]; B%: 69%-99%, 10 min). 1,6-dimethyl-4-[(3S,4R)-3-methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5- naphthyridine-3-carbonitrile (60 mg, 58 %) was obtained as a white solid. LC/MS (ESI) m/z: 472.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.90 (d, J = 8.8 Hz, 1H), 7.59 (d, J = 8.8 Hz, 1H), 7.05 (d, J = 8.4 Hz, 2H), 6.75 (d, J = 9.2Hz, 2H), 5.98 (d, J = 8.4 Hz, 1H), 4.12 - 4.01 (m, 1H), 3.94 - 3.82 (m, 2H), 3.77 ( J = 4.0, 8.1 Hz, 1H), 3.74 - 3.65 (m, 1H), 3.51 (s, 3H), 2.57 (s, 3H), 2.38 - 2.30 (m, 1H), 2.20 - 2.07 (m, 1H), 1.91 - 1.81 (m, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 269 Exemplary Synthesis of 7-[(3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl-1-piperidyl]- 2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of tert-butyl (3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl- piperidine-1-carboxylate and tert-butyl (3R,4S)-4-(4-chloro-N-methyl-anilino)-3- methyl-piperidine-1-carboxylate Racemic tert-butyl (3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl-piperidine-1-carboxylate (0.5 g, 1.48 mmol) was purified by SFC (column: DAICEL CHIRALPAK AD(250 mm * 30 mm, 10 um); mobile phase: [Neu- methylalcohol]; B%: 20%-20%, 6.5; 78 min). tert-Butyl (3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl-piperidine-1-carboxylate (220 mg, 44%) was obtained as a white oil. LC/MS (ESI) m/z: 339 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.22 - 7.13 (m, 2H), 6.78 - 6.70 (m, 2H), 4.11 (s, 1H), 3.93 - 3.66 (m, 2H), 3.20 - 2.95 (m, 1H), 2.93 - 2.78 (m, 1H), 2.77 (s, 3H), 2.12 (s, 1H), 2.01 (J = 4.4, 12.4 Hz, 1H), 1.49 (dd, J = 2.8, 12.4 Hz, 1H), 1.40 (s, 9H), 0.80 (d, J = 6.8 Hz, 3H). tert-Butyl (3R,4S)-4-(4-chloro-N-methyl-anilino)-3-methyl-piperidine-1-carboxylate (240 mg, 48 %) was obtained as a white oil. LC/MS (ESI) m/z: 339 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.22 - 7.12 (m, 2H), 6.74 (d, J = 9.2 Hz, 2H), 4.11 (d, J = 1.2 Hz, 1H), 3.91 - 3.69 (m, 2H), 3.21 - 2.95 (m, 1H), 2.93 - 2.79 (m, 1H), 2.77 (s, 3H), 2.12 (s, 1H), 2.01 (dd, J = 4.4, 12.4 Hz, 1H), 1.49 (dd, J = 2.4, 12.4 Hz, 1H), 1.40 (s, 9H), 0.80 (d, J = 7.2 Hz, 3H).
Step 2: Preparation of 7-[(3S,4R)-4-(4-chloro-N-methyl-anilino)-3-methyl-1-piperidyl]- 2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3S,4R)-N-(4-chlorophenyl)-N,3-dimethyl-piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 442.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.20 (d, J = 8.8 Hz, 2H), 6.82 (d, J = 8.8 Hz, 2H), 4.51 (d, J = 13.2 Hz, 1H), 4.31 (d, J = 13.2 Hz, 1H), 4.13 - 4.05 (m, 1H), 3.83 - 3.73 (m, 1H), 3.48 (s, 3H), 3.42 (t, J = 12.0 Hz, 1H), 2.82 (s, 3H), 2.72 (s, 3H), 2.55 (d, J = 2.4 Hz, 1H), 2.33 (s, 1H), 1.75 (d, J = 10.4 Hz, 1H), 0.82 (d, J = 6.8 Hz, 3H). Synthesis of Compound 270 Exemplary Synthesis of 4-[(3aR,6aR)-5-[4-(trifluoromethoxy)phenyl]- octahydropyrrolo[3,4-c]pyrrol-2-yl]-1,6-dimethyl-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile The title compound was prepared in an analogous manner to 4-(5-fluorospiro[3H-benzofuran- 2,4'-piperidine]-1'-yl)-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert- butyl (3aR,6aR)-2-[4-(trifluoromethoxy)phenyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate. (yellow solid). LC/MS (ESI) m/z: 470.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ7.85 (d, J = 8.8 Hz, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.16 (d, J = 8.8 Hz, 2H), 6.57 (d, J = 8.8 Hz, 2H), 4.36 - 4.28 (m, 2H), 4.27 - 4.21 (m, 2H), 3.56 - 3.50 (m, 2H), 3.48 (s, 3H), 3.21 (t, J = 9.2 Hz, 2H), 2.56 (s, 3H), 2.44 (s, 2H). Synthesis of Compound 271 Exemplary Synthesis of 7-[(3aS,7aR)-1-(4-fluorophenyl)-octahydro-1H-pyrrolo[3,2- c]pyridin-5-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3aR,7aS)-1-(4- fluorophenyl)-octahydro-1H-pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5-oxo-4H,5H- [1,3]thiazolo[5,4-b]pyridine-6-carbonitrile starting from benzyl (3aS,7aR)-1-(4- fluorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate. (yellow solid). LC/MS (ESI) m/z: 424.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.00 (t, J = 8.8 Hz, 2H), 6.58 (dd, J = 4.4, 8.8 Hz, 2H), 4.33 (dd, J = 2.8, 14.0 Hz, 1H), 4.23 (d, J = 12.8 Hz, 1H), 4.04 - 3.91 (m, 2H), 3.51 - 3.43 (m, 4H), 3.38 - 3.34 (m, 1H), 3.17 (q, J = 8.8 Hz, 1H), 2.69 (s, 3H), 2.62 (dd, J = 4.0, 7.2 Hz, 1H), 2.17 (dd, J = 5.2, 10.4 Hz, 1H), 2.09 - 1.99 (m, 1H), 1.97 - 1.86 (m, 1H), 1.77 - 1.64 (m, 1H).
Synthesis of Compound 272 Exemplary Synthesis of 7-[(3aR,7aS)-1-[4-(trifluoromethoxy)phenyl]-octahydro-1H- pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile
Figure imgf000516_0001
The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aR,7aS)-1-(4-(trifluoromethoxy)phenyl)octahydro-1H-pyrrolo[3,2-c]pyridine. (white solid). LC/MS (ESI) m/z: 490.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.14 (d, J = 8.8 Hz, 2H), 6.63 (d, J = 9.2 Hz, 2 H), 4.40 – 4.37 (m, 1 H), 4.22 - 4.31 (m, 1H), 3.92 - 4.06 (m, 2H), 3.48 (s, 3H), 3.35 - 3.47 (m, 2H), 3.23 – 3.21 (m, 1H), 2.69 (s, 3H), 2.57 - 2.65 (m, 1H), 2.17 - 2.26 (m, 1H), 1.89 - 2.09 (m, 2H), 1.66 - 1.78 (m, 1H). Synthesis of Compound 273 Exemplary Synthesis of 4-[(3aR,7aS)-1-[4-(trifluoromethoxy)phenyl]-octahydro-1H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile
The title compound was prepared in an analogous manner to 4-[(3aS,7aR)-1-[4- (trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro- 1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from benzyl (3aR,7aS)-1-[4- (trifluoromethoxy)phenyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate. (yellow solid). LC/MS (ESI) m/z: 504.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 9.20 Hz, 1H), 7.79 - 7.83 (m, 1H), 7.14 (d, J = 8.88 Hz, 2H), 6.65 – 6.63 (m, 2H), 4.20 - 4.27 (m, 1H), 4.03 - 4.16 (m, 2H), 3.90 - 3.97 (m, 1H), 3.53 (s, 3H), 3.41 - 3.50 (m, 1H), 2.67 (s, 2H), 2.30 - 2.35 (m, 2H), 2.07 - 2.13 (m, 2H), 1.78 - 1.85 (m, 1 H). Synthesis of Compound 274 Exemplary Synthesis of 1,6-dimethyl-4-[(3R,4S)-3-methyl-4-{[4- (trifluoromethoxy)phenyl]amino}piperidin-1-yl]-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile The title compound was prepared in an analogous manner to 1,6-dimethyl-4-[(3S,4R)-3- methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-1,5-naphthyridine-3-carbonitrile starting from (3R,4S)-3-methyl-N-[4-(trifluoromethoxy)phenyl]piperidin-4-amine. (white solid). LC/MS (ESI) m/z: 472.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.90 (d, J = 8.8 Hz, 1H), 7.59 (d, J = 8.8 Hz, 1H), 7.05 (d, J = 8.4 Hz, 2H), 6.77 - 6.72 (m, 2H), 5.98 (d, J = 8.4 Hz, 1H), 4.13 - 3.98 (m, 1H), 3.96 - 3.84 (m, 2H), 3.82 - 3.65 (m, 2H), 3.51 (s, 3H), 2.57 (s, 3H), 2.33 (dd, J = 2.0, 3.6 Hz, 1H), 2.21 - 2.06 (m, 1H), 1.94 - 1.78 (m, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 275 Exemplary Synthesis of 7-[(3R,4R)-4-[(4-chloro-2-fluorophenyl)amino]-3- methylpiperidin-1-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloro-2-fluoro- anilino)-3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3R,4R)-N-(4-chloro-2-fluoro-phenyl)-3-methyl-piperidin-4-amine. (off-white solid). LC/MS (ESI) m/z: 446.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.16 (dd, J = 2.4, 11.6 Hz, 1H), 7.04 - 6.97 (m, 1H), 6.92 - 6.81 (m, 1H), 5.53 - 5.45 (m, 1H), 4.56 - 4.38 (m, 2H), 3.57 - 3.41 (m, 5H), 3.11 (dd, J = 11.6, 13.2 Hz, 1H), 2.69 (s, 3H), 2.06 - 1.98 (m, 2H), 1.62 - 1.48 (m, 1H), 0.94 (d, J = 6.4 Hz, 3H). Synthesis of Compound 276 Exemplary Synthesis of 6-chloro-1-methyl-4-((3R,4S)-3-methyl-4-((4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydroquinoline-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3S,4R)-3- methyl-4-[4-(trifluoromethoxy)anilino]-1-piperidyl]-2-oxo-quinoline-3-carbonitrile starting from 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (white solid). LC/MS (ESI) m/z: 491.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.83 - 7.74 (m, 2H), 7.64 - 7.57 (m, 1H), 7.05 (d, J = 8.4 Hz, 2H), 6.75 (d, J = 9.2 Hz, 2H), 5.99 (d, J = 8.4 Hz, 1H), 3.70 (s, 3H), 3.64 - 3.57 (m, 1H), 3.56 (s, 3H), 3.52 - 3.44 (m, 1H), 2.33 – 2.31 (m, 1H), 2.07 - 1.97 (m, 1H), 1.90 – 1.87 (m, 1H), 0.91 (d, J = 6.4 Hz, 3H). Synthesis of Compound 277 Exemplary Synthesis of 7-[(3R,4S)-4-[(4-chlorophenyl)(methyl)amino]-3- methylpiperidin-1-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 7-[(3S,4R)-4-(4-chloro-N- methyl-anilino)-3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6- carbonitrile starting from tert-butyl (3R,4S)-4-(4-chloro-N-methyl-anilino)-3-methyl- piperidine-1-carboxylate. (white solid). LC/MS (ESI) m/z: 442.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.20 (d, J = 9.2 Hz, 2H), 6.83 (d, J = 9.2 Hz, 2H), 4.51 (d, J = 12.8 Hz, 1H), 4.31 (d, J = 13.2 Hz, 1H), 4.15 - 4.05 (m, 1H), 3.78 (dd, J = 2.4, 13.2 Hz, 1H), 3.48 (s, 3H), 3.42 (t, J = 11.6 Hz, 1H), 2.82 (s, 3H), 2.72 (s, 3H), 2.54 (s, 1H), 2.33 (d, J = 2.0 Hz, 1H), 1.81 - 1.67 (m, 1H), 0.82 (d, J = 6.8 Hz, 3H).
Synthesis of Compound 278 Exemplary Synthesis of 1,6-dimethyl-4-[(3R,4S)-3-methyl-4-{methyl[4- (trifluoromethoxy)phenyl]amino}piperidin-1-yl]-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile The title compound was prepared in an analogous manner to 4-(5-fluorospiro[3H-benzofuran- 2,4'-piperidine]-1'-yl)-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from tert- butyl (3S,4R)-3-methyl-4-[N-methyl-4-(trifluoromethoxy)anilino]piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 485.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J = 8.8 Hz, 1H), 7.61 (d, J = 8.8 Hz, 1H), 7.17 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 9.2 Hz, 2H), 4.52 - 4.45 (m, 1H), 4.21 - 4.12 (m, 1H), 4.05 - 3.99 (m, 1H), 3.94 - 3.87 (m, 1H), 3.52 (s, 3H), 3.41 (t, J = 11.2 Hz, 1H), 2.88 (s, 3H), 2.81 (dq, J = 3.6, 12.0 Hz, 1H), 2.61 (s, 3H), 2.34 (dd, J = 3.6, 6.0 Hz, 1H), 1.76 (d, J = 10.4 Hz, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 279 Exemplary Synthesis of 6-chloro-1-methyl-2-oxo-4-((3aR,7aS)-1-(5- (trifluoromethoxy)pyridin-2-yl)octahydro-5H-pyrrolo[3,2-c]pyridin-5-yl)-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile Step 1: Preparation of benzyl (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate A mixture of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate (620 mg, 2.38 mmol, 1 eq), 2-bromo-5-(trifluoromethoxy)pyridine (576 mg, 2.38 mmol, 1 eq), 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (116 mg, 0.12 mmol, 0.05 eq), cesium carbonate (3.88 g, 11.91 mmol, 5 eq) in dioxane (6 mL) was degassed and purged with nitrogen (3X), then stirred at 100°C for 3 h under nitrogen atmosphere. The mixture was filtered and concentrated, and the resulting residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate = 5/1) to afford benzyl (3aR,7aS)-1-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate (0.5 g, 50%) as a yellow oil. LC/MS (ESI) m/z: 422.4 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.08 (d, J = 2.4 Hz, 1H), 7.55 - 7.50 (m, 1H), 7.39 - 7.33 (m, 5H), 6.49 (d, J = 9.2 Hz, 1H), 5.13 - 5.05 (m, 2H), 4.15 ( J = 6.4, 9.5 Hz, 1H), 3.95 (d, J = 13.6 Hz, 1H), 3.85 (d, J = 13.2 Hz, 1H), 3.49 - 3.40 (m, 1H), 3.34 (s, 1H), 3.30 - 3.25 (m, 1H), 2.99 - 2.83 (m, 1H), 2.40 - 2.32 (m, 1H), 2.14 - 2.05 (m, 1H), 1.96 - 1.82 (m, 2H), 1.39 - 1.30 (m, 1H). Step 2: Preparation of (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-2,3,3a,4,5,6,7,7a- octahydropyrrolo[3,2-c]pyridine To a solution of benzyl (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate (110 mg, 0.26 mmol, 1 eq) in methanol (1 mL) was added10% palladium on activated carbon catalyst (30 mg, 0.26 mmol, 1 eq). The mixture was degassed and purged with hydrogen, then stirred at 25°C for 12 h under hydrogen. The reaction mixture was filtered and concentrated under reduced pressure to give (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2- c]pyridine (42 mg, crude) as a white solid. LC/MS (ESI) m/z: 287.28 [M+H]+. Step 3: preparation of 4-[(3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine- 3- carbonitrile
Figure imgf000522_0001
The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (White solid). LC/MS (ESI) m/z: 505.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.12 - 8.05 (m, 2H), 7.81 (d, J = 8.8 Hz, 1H), 7.56 (J = 2.0, 9.2 Hz, 1H), 6.55 (d, J = 9.2 Hz, 1H), 4.35 - 4.21 (m, 2H), 4.12 (d, J = 13.2 Hz, 1H), 3.92 (J = 4.0, 13.2 Hz, 1H), 3.53 (s, 3H), 3.50 (s, 3H), 2.59 (dd, J = 2.8, 6.8 Hz, 1H), 2.34 - 2.23 (m, 1H), 2.15 - 2.06 (m, 2H), 1.97 - 1.86 (m, 1H). Synthesis of Compound 280 Exemplary Synthesis of 7-[(3aR,7aS)-1-[5-(trifluoromethoxy)pyridin-2-yl]-octahydro- 1H-pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine- 6-carbonitrile Step 1: Preparation of benzyl (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate and benzyl (3aS,7aR)- 1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine- 5-carboxylate Racemic benzyl (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (0.3 g, 0.71 mmol) was purified by SFC, then further separated by SFC (column: DAICEL CHIRALCEL OJ(250 mm*30 mm,10 um); mobile phase: [Neu-IPA];B%: 20%-20%,9.5; 190 min). Benzyl (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (130 mg, 43%) was obtained as a white oil. LC/MS (ESI) m/z: 422.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.96 (s, 1H), 7.27 (s, 5H), 7.17 (s, 1H), 6.19 (d, J = 9.2 Hz, 1H), 5.11 - 5.01 (m, 2H), 4.28 - 3.93 (m, 3H), 3.42 - 3.23 (m, 3H), 2.87 (d, J = 12.0 Hz, 1H), 2.31 (s, 1H), 2.12 (d, J = 11.6 Hz, 1H), 1.95 (d, J = 9.6 Hz, 2H), 1.46 - 1.36 (m, 1H). benzyl (3aS,7aR)-1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (150 mg, 50%) was obtained as a white oil. LC/MS (ESI) m/z: 422.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.98 (s, 1H), 7.32 - 7.25 (m, 5H), 7.19 (s, 1H), 6.21 (d, J = 9.2 Hz, 1H), 5.15 - 5.02 (m, 2H), 4.22 - 3.93 (m, 3H), 3.34 - 3.15 (m, 3H), 2.96 - 2.75 (m, 1H), 2.32 (d, J = 3.2 Hz, 1H), 2.19 - 2.07 (m, 1H), 2.03 - 1.90 (m, 2H), 1.48 - 1.35 (m, 1H). Step 2: Preparation of 7-[(3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6- carbonitrile
Figure imgf000523_0001
The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aR,7aS)-1-[5-(trifluoromethoxy)-2-pyridyl]-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2- c]pyridine. (white solid). LC/MS (ESI) m/z: 491.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J = 2.4 Hz, 1H), 7.55 (dd, J = 2.0, 9.2 Hz, 1H), 6.55 (d, J = 9.2 Hz, 1H), 4.41 (d, J = 13.6 Hz, 1H), 4.29 - 4.23 (m, 2H), 3.93 (dd, J = 4.0, 13.6 Hz, 1H), 3.54 - 3.49 (m, 1H), 3.48 (s, 3H), 3.46 - 3.34 (m, 2H), 2.69 (s, 3H), 2.63 - 2.57 (m, 1H), 2.30 - 2.22 (m, 1H), 2.06 - 1.92 (m, 2H), 1.85 - 1.74 (m, 1H). Synthesis of Compound 281 Exemplary Synthesis of 6-chloro-4-(4-hydroxy-4-{[4-(propan-2-yloxy)pyridin-2- yl]methyl}piperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of 4-isopropoxy-2-methyl-pyridine To a solution of 2-methylpyridin-4-ol (500 mg, 4.58 mmol, 1 eq), propan-2-ol (413 mg, 6.87 mmol, 526.14 uL, 1.5 eq) and triphenylphosphine (1.80 g, 6.87 mmol, 1.5 eq) in tetrahydrofuran (3 mL) was added diisopropyl azodicarboxylate (1.39 g, 6.87 mmol, 1.34 mL, 1.5 eq), and the reaction mixture was stirred at 25 °C for 8 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure, and the crude product was purified by reversed-phase HPLC (column: YMC Triart C18250*50mm*7um; mobile phase: [water (formic acid)-acetonitrile]; B%: 5%-35%, 20min) to give 4-isopropoxy-2-methyl-pyridine (280 mg, 40%) as a yellow oil. LC/MS (ESI) m/z: 152.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.28 (d, J = 5.6 Hz, 1H), 6.63 (s, 1H), 6.60 (d, J = 5.6 Hz, 1H), 4.65 – 4.59 (m, 1H), 2.49 (s, 3H), 1.35 (d, J = 6.0 Hz, 6H). Step 2: Preparation of tert-butyl 4-hydroxy-4-[(4-isopropoxy-2- pyridyl)methyl]piperidine-1-carboxylate To a solution of 4-isopropoxy-2-methyl-pyridine (280 mg, 1.85 mmol, 1 eq) in tetrahydrofuran at -78 °C (2.5 mL) was added n-butyllithium (2.5 M, 888.86 uL, 1.2 eq) dropwise for 0.5 h followed by tert-butyl 4-oxopiperidine-1-carboxylate (369 mg, 1.85 mmol, 1 eq) in tetrahydrofuran (2.5 mL) dropwise added, and the reaction mixture was stirred at -78 °C for 0.5 h. The reaction was quenched by saturated aqueous ammonium chloride solution (20 mL), and extracted with ethyl acetate (10 mL x 3). The combined organic extractrs were washed with brine (10 mL x 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (petroleum ether/ethyl acetate=20/1 to 0/1) to afford tert-butyl 4-hydroxy-4-[(4-isopropoxy-2- pyridyl) methyl] piperidine-1-carboxylate (170 mg, 26%) as a white liquid. LC/MS (ESI) m/z: 351.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.28 (d, J = 6.0 Hz, 1H), 6.69 (dd, J = 5.6 Hz, 1H), 6.60 (d, J = 2.4 Hz, 1H), 4.66 – 4.63 (m, 1H), 3.89 - 3.71 (m, 2H), 3.24 (t, J = 9.6 Hz, 2H), 2.84 (s, 2H), 1.57 - 1.49 (m, 4H), 1.46 (s, 9H), 1.38 (d, J = 6.0 Hz, 6H). Step 3: Preparation of 4-[(4-isopropoxy-2-pyridyl)methyl]piperidin-4-ol
Figure imgf000525_0001
To tert-butyl 4-hydroxy-4-[(4-isopropoxy-2-pyridyl)methyl]piperidine-1-carboxylate (100 mg, 0.28 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 23.67 eq), and the reaction mixture was degassed and purged with N2 (3X), then stirred at 25 °C for 0.5 h. The reaction was concentrated to afford 4-[(4- isopropoxy-2-pyridyl) methyl] piperidin-4-ol trifluoroacetic acid (71 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 251.0 [M+H]+. Step 4: Preparation of 6-chloro-4-[4-hydroxy-4-[(4-isopropoxy-2-pyridyl)methyl]-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
Figure imgf000525_0002
The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (white solid). LC/MS (ESI) m/z: 468.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J = 6.0 Hz, 1H), 8.06 (d, J = 8.8 Hz, 1H), 7.79 (d, J = 9.0 Hz, 1H), 7.08 (s, 2H), 5.28 (s, 1H), 4.91 - 4.80 (m, 1H), 3.99 – 3.95 (m, 2H), 3.71 (t, J = 11.2 Hz, 2H), 3.52 (s, 3H), 2.98 (s, 2H), 1.96 - 1.80 (m, 2H), 1.63 (d, J = 12.8 Hz, 2H), 1.33 (d, J = 6.0 Hz, 6H). Synthesis of Compound 282 Exemplary Synthesis of 1,6-dimethyl-2-oxo-4-[5-(trifluoromethoxy)-1,3- dihydrospiro[indene-2,4'-piperidin]-1'-yl]-1,2-dihydro-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 4-(5-fluorospiro[3H-benzofuran- 2,4'-piperidine]-1'-yl)-1,6-dimethyl-2-oxo-1,5-naphthyridine-3-carbonitrile starting from 5- (trifluoromethoxy)spiro[indane-2,4'-piperidine]. (white solid). LC/MS (ESI) m/z: 469.5 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.90 (d, J = 8.8 Hz, 1H), 7.58 (d, J = 8.8 Hz, 1H), 7.32 (d, J = 8.4 Hz, 1H), 7.21 (s, 1H), 7.11 (d, J = 7.6 Hz, 1H), 3.90 (s, 4H), 3.51 (s, 3H), 2.95 (s, 2H), 2.92 (s, 2H), 2.55 (s, 3H), 1.84 (t, J = 5.2 Hz, 4H).
Synthesis of Compound 283 Exemplary Synthesis of 2,4-dimethyl-5-oxo-7-[5-(trifluoromethoxy)-1,3- dihydrospiro[indene-2,4'-piperidin]-1'-yl]-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7-spiro[3H- benzofuran-2,4'-piperidine]-1'-yl-thiazolo[5,4-b]pyridine-6-carbonitrile starting from 5- (trifluoromethoxy)spiro[indane-2,4'-piperidine]. (yellow solid). LC/MS (ESI) m/z: 475.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.31 (d, J = 8.4 Hz, 1H), 7.20 (s, 1H), 7.11 (d, J = 8.0 Hz, 1H), 3.96 - 3.89 (m, 4H), 3.48 (s, 3H), 2.92 (s, 2H), 2.89 (s, 2H), 2.69 (s, 3H), 1.78 - 1.73 (m, 4H). Synthesis of Compound 284 Exemplary Synthesis of 6-chloro-4-{4-hydroxy-4-[6-methyl-3-(propan-2-yloxy)pyridin- 2-yl]piperidin-1-yl}-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of 5-isopropoxy-2-methyl-pyridine To a solution of 2-bromo-5-isopropoxy-pyridine (1 g, 4.63 mmol, 1 eq) in dioxane (10 mL) were added potassium carbonate (2 g, 13.88 mmol, 3 eq) , [1,1'- bis(diphenylphosphino)ferrocene] dichloropalladium(II) (338 mg, 0.46 mmol, 0.1 eq), and 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (3 g, 13.88 mmol, 3.88 mL, 50% purity, 3 eq), and the reaction mixture was stirred at 90 °C for 12 h. The mixture was filtered and concentrated, and the resulting residue was purified by column chromatography (petroleum ether/ethyl acetate = 20/1 to 3/1) to afford 5-isopropoxy-2-methyl-pyridine (500 mg, 71%) as a yellow oil. LC/MS (ESI) m/z: 152.1 [M+H]+. Step 2: Preparation of tert-butyl 4-hydroxy-4-(3-isopropoxy-6-methyl-2- pyridyl)piperidine-1-carboxylate To a solution of 5-isopropoxy-2-methyl-pyridine (500 mg, 3.31 mmol, 1 eq) in tetrahydrofuran at -78 °C (5 mL) was added n-butyllithium (2.5 M, 1.59 mL, 1.2 eq) dropwise for 0.5 h followed by tert-butyl 4-oxopiperidine-1-carboxylate (988 mg, 4.96 mmol, 1.5 eq) in tetrahydrofuran (5 mL) dropwise, and the reaction resulting mixture was stirred at - 78 °C for 0.5 h. The reaction was quenched by addition of saturated aqueous ammonium chloride solution (30 mL). The resulting mixture was extracted with ethyl acetate (20 mL x 3), and the combined organic extracts were washed with brine (20 mL x 3), dried over sodium sulfite, filtered, and concentrated under reduced pressure. The crude product was purified by reversed-phase HPLC (column: Phenomenex C18250*50mm*10um; mobile phase: [water(ammonium hydrogen carbonate)- acetonitrile]; B%: 49%-79%, 8min) to afford tert-butyl 4-hydroxy-4-(3-isopropoxy-6-methyl-2-pyridyl)piperidine-1-carboxylate (100 mg, 9%) as a yellow oil. LC/MS (ESI) m/z: 351.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.17 - 7.10 (m, 1H), 7.08 - 7.02 (m, 1H), 6.42 - 6.27 (m, 1H), 4.67 - 4.54 (m, 1H), 4.19 - 3.96 (m, 2H), 3.41 - 3.21 (m, 2H), 2.62 - 2.45 (m, 5H), 1.50 (s, 9H), 1.49 - 1.45 (m, 2H), 1.37 (d, J = 5.6 Hz, 6H). Step 3: Preparation of 4-(3-isopropoxy-6-methyl-2-pyridyl)piperidin-4-ol To a solution of tert-butyl 4-hydroxy-4-(3-isopropoxy-6-methyl-2-pyridyl)piperidine-1- carboxylate (100 mg, 0.29 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 24 eq), and the reaction mixture was stirred at 25 °C for 0.5 h. The mixture was filtered, then concentrated to afford 4-(3-isopropoxy-6-methyl-2- pyridyl)piperidin-4-ol trifluoroacetic acid (100 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 250.8 [M+H]+. Step 4: Preparation of 6-chloro-4-[4-hydroxy-4-(3-isopropoxy-6-methyl-2-pyridyl)-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS (ESI) m/z: 468.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.63 (d, J = 8.8 Hz, 1H), 7.51 (d, J = 8.8 Hz, 1H), 7.20 - 7.00 (m, 2H), 6.63 - 6.38 (m, 1H), 4.61 (td, J = 6.0, 12.0 Hz, 1H), 4.37 (d, J = 13.2 Hz, 2H), 4.08 (t, J = 12.8 Hz, 2H), 3.64 (s, 3H), 3.07 (dt, J = 4.4, 13.2 Hz, 2H), 2.51 (s, 3H), 1.57 (s, 2H), 1.33 (d, J = 6.0 Hz, 6H). Synthesis of Compound 285 Exemplary Synthesis of 2,4-dimethyl-5-oxo-7-((3aS,7aR)-1-(5- (trifluoromethoxy)pyridin-2-yl)octahydro-5H-pyrrolo[3,2-c]pyridin-5-yl)-4,5- dihydrothiazolo[5,4-b]pyridine-6-carbonitrile
Figure imgf000530_0001
The title compound was prepared in an analogous manner to 7-[(3aR,7aS)-1-[5- (trifluoromethoxy)pyridin-2-yl]-octahydro-1H-pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5- oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile starting from benzyl (3aS,7aR)-1-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate. (white solid). LC/MS (ESI) m/z: 491.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J = 2.4Hz, 1H), 7.56 (dd, J = 2.4, 8.8 Hz, 1H), 6.55 (d, J = 9.2 Hz, 1H), 4.41 (d, J = 13.2 Hz, 1H), 4.30 - 4.23 (m, 2H), 3.93 (dd, J = 4.0, 13.6 Hz, 1H), 3.56 - 3.49 (m, 1H), 3.48 (s, 3H), 3.46 - 3.34 (m, 2H), 2.69 (s, 3H), 2.59 (dd, J = 3.2, 7.2 Hz, 1H), 2.30 - 2.22 (m, 1H), 2.08 - 1.94 (m, 2H), 1.86 - 1.74 (m, 1H). Synthesis of Compound 286 Exemplary Synthesis of 1,6-dimethyl-4-((3S,4R)-3-methyl-4-(methyl(4- (trifluoromethoxy)phenyl)amino)piperidin-1-yl)-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile
The title compound was prepared in an analogous manner to 1,6-dimethyl-4-[(3R,4S)-3- methyl-4-{methyl[4-(trifluoromethoxy)phenyl]amino}piperidin-1-yl]-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile starting from tert-butyl (3S,4R)-3-methyl-4-[N-methyl-4- (trifluoromethoxy)anilino]piperidine-1-carboxylate. (yellow solid). LC/MS (ESI) m/z: 486.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.92 (d, J = 8.8 Hz, 1H), 7.61 (d, J = 8.8 Hz, 1H), 7.17 (d, J = 8.8 Hz, 2H), 6.88 (d, J = 9.2 Hz, 2H), 4.48 (d, J = 12.8 Hz, 1H), 4.16 (d, J = 11.6 Hz, 1H), 4.06 - 3.99 (m, 1H), 3.94 - 3.87 (m, 1H), 3.52 (s, 3H), 3.41 (t, J = 12.0 Hz, 1H), 2.88 (s, 3H), 2.86 - 2.74 (m, 1H), 2.61 (s, 3H), 2.38 - 2.30 (m, 1H), 1.76 (d, J = 10.0 Hz, 1H), 0.86 (d, J = 6.8 Hz, 3H). Synthesis of Compound 287 Exemplary Synthesis of 4-[(3aR,7aS)-1-[5-(propan-2-yloxy)pyridin-2-yl]-octahydro-1H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile Step 1: Preparation of benzyl (3aR,7aS)-1-(5-isopropoxy-2-pyridyl)-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate
Figure imgf000531_0001
To a solution of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate trifluoroacetic acid (200 mg, 0.53 mmol, 1 eq) and 2-bromo-5-isopropoxy- pyridine (173 mg, 0.80 mmol, 1.5 eq) in dioxane (2 mL) were added cesium carbonate (522 mg, 1.60 mmol, 3 eq) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1- ium-2-ide;3-chloropyridine dichloropalladium (52 mg, 0.053 mmol, 0.1 eq), and the reaction mixture was stirred at 100 °C for 3 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel chromatography (petroleum ether/ethyl acetate = 100/1 to 1:1) to give benzyl (3aR,7aS)-1-(5-isopropoxy-2-pyridyl)-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate (200 mg, 95%) as a yellow oil. LC/MS (ESI) m/z: 396.4 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 7.87 (d, J = 2.8 Hz, 1H), 7.41 - 7.31 (m, 5H), 7.13 (dd, J = 2.8, 8.8 Hz, 1H), 6.30 (d, J = 9.2 Hz, 1H), 5.20 - 5.11 (m, 2H), 4.35 (s, 1H), 4.22 - 4.14 (m, 1H), 4.06 - 3.87 (m, 2H), 3.52 - 3.45 (m, 1H), 3.40 - 3.33 (m, 2H), 3.08 - 2.93 (m, 1H), 2.41 (d, J = 4.4 Hz, 1H), 2.22 - 2.15 (m, 1H), 2.03 - 1.93 (m, 2H), 1.58 - 1.52 (m, 1H), 1.30 (d, J = 6.0 Hz, 6H). Step 2: Preparation of (3aR,7aS)-1-(5-isopropoxy-2-pyridyl)-2,3,3a,4,5,6,7,7a- octahydropyrrolo[3,2-c]pyridine A mixture of benzyl (3aR,7aS)-1-(5-isopropoxy-2-pyridyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (100 mg, 0.25 mmol, 1 eq) and 10% palladium on activated carbon (30 mg, 0.025 mmol, 0.1 eq) in methanol (1 mL) was degassed and purged with hydrogen (3X), then stirred at 25 °C for 12 h under hydrogen atmosphere (50 psi). The mixture was filtered, then concentrated under reduced pressure to give (3aR,7aS)-1-(5- isopropoxy-2-pyridyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine (50 mg, crude) as an off-white oil. LC/MS (ESI) m/z: 262.2 [M+H]+. Step 3: Preparation of 4-[(3aR,7aS)-1-(5-isopropoxy-2-pyridyl)-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LC/MS (ESI) m/z: 479.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 9.2 Hz, 1H), 7.83 - 7.77 (m, 2H), 7.23 (dd, J = 2.8, 8.8 Hz, 1H), 6.45 (d, J = 9.2 Hz, 1H), 4.36 (td, J = 6.0, 12.0 Hz, 1H), 4.25 - 4.07 (m, 3H), 3.96 (dd, J = 4.0, 13.2 Hz, 1H), 3.53 (s, 3H), 3.49 - 3.41 (m, 2H), 3.30 - 3.25 (m, 1H), 2.59 (d, J = 6.8 Hz, 1H), 2.30 - 2.19 (m, 1H), 2.10 - 2.02 (m, 2H), 1.99 - 1.86 (m, 1H), 1.21 (d, J = 6.0 Hz, 6H). Synthesis of Compound 288 Exemplary Synthesis of 4-[(3aS,6aS)-5-[5-(propan-2-yloxy)pyrimidin-2-yl]- octahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl (3aR,6aR)-2-(5-isopropoxypyrimidin-2-yl)- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate To a solution of tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (50 mg, 0.24 mmol, 1 eq) and 2-chloro-5-isopropoxy-pyrimidine (41 mg, 0.24 mmol, 1 eq) in dioxane (1 mL) were added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine dichloropalladium (23 mg, 0.02 mmol, 0.1 eq) and cesium carbonate (230 mg, 0.71 mmol, 3 eq), and the reaction mixture was stirred at 100 °C for 3 h. The mixture was filtered, then concentrated, and the resulting residue was purified by prep-TLC (silicon dioxide, petroleum ether: ethyl acetate = 3:1) to afford tert- butyl (3aR,6aR)-2-(5-isopropoxypyrimidin-2-yl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (50 mg, 61%) as a white solid. LC/MS (ESI) m/z: 349.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.15 (s, 1H), 8.16 - 8.13 (m, 1H), 4.37 (td, J = 6.0, 12.0 Hz, 1H), 3.80 - 3.70 (m, 2H), 3.53 (dd, J = 6.0, 8.8 Hz, 2H), 3.14 - 3.06 (m, 2H), 3.04 - 2.93 (m, 2H), 2.31 - 2.23 (m, 2H), 1.41 (s, 9H), 1.21 (d, J = 6.0 Hz, 6H). Step 2: Preparation of (3aS,6aS)-5-(5-isopropoxypyrimidin-2-yl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aR,6aR)-2-(5-isopropoxypyrimidin-2-yl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (50 mg, 0.14 mmol, 1 eq) and trifluoroacetic acid (770 mg, 6.75 mmol, 0.5 mL, 47.06 eq) in dichloromethane (1 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 0.5 h under nitrogen atmosphere. The reaction was concentrated to afford (3aS,6aS)-5-(5-isopropoxypyrimidin-2-yl)-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole trifluoroacetic acid (50 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 249.1 [M+H]+. Step 3: Preparation of 4-[(3aS,6aS)-5-(5-isopropoxypyrimidin-2-yl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (off-white solid). LC/MS (ESI) m/z: 466.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.18 (s, 2H), 8.02 (d, J = 8.8 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 4.44 - 4.35 (m, 1H), 4.26 - 4.16 (m, 4H), 3.88 (dd, J = 5.6, 9.6 Hz, 2H), 3.50 (s, 3H), 3.25 (t, J = 10.0 Hz, 2H), 2.47 - 2.40 (m, 2H), 1.22 (d, J = 6.0 Hz, 6H). Synthesis of Compound 289 Exemplary Synthesis of 4-[(3aS,6aS)-5-[5-(trifluoromethoxy)pyridin-2-yl]- octahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile Step 1: preparation of tert-butyl (3aR,6aR)-2-[5-(trifluoromethoxy)-2-pyridyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate A mixture of tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (300 mg, 1.41 mmol, 1 eq), 2-bromo-5-(trifluoromethoxy)pyridine (513 mg, 2.12 mmol, 1.5 eq), cesium carbonate (1.38 g, 4.24 mmol, 3 eq), 1,3-bis[2,6-bis(1- propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (137 mg, 0.14 mmol, 0.1 eq) in dioxane (4 mL) was stirred at 100°C for 3 h under nitrogen atmosphere. The mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate = 100/1 to 1:1) to give tert-butyl (3aR,6aR)-2-[5-(trifluoromethoxy)-2- pyridyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (385 mg, 72%) as a yellow solid. LC/MS (ESI) m/z: 374.3 [M+H]+. Step 2: preparation of (3aS,6aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-2,3,3a,4,6,6a- hexahydro-1H-pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aR,6aR)-2-[5-(trifluoromethoxy)-2-pyridyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (80 mg, 0.21 mmol, 1 eq) and trifluoroacetic acid (1.23 g, 10.80 mmol, 0.80 mL, 50.43 eq) in dichloromethane (1 mL) was stirred at 25 °C for 0.5 h under nitrogen. The reaction mixture was concentrated under reduced pressure to give (3aS,6aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4- c]pyrrole trifluoroacetic acid (85 mg, crude) as a brown oil. LC/MS (ESI) m/z: 273.8 [M+H]+. Step 3: Preparation of 4-[(3aS,6aS)-2-[5-(trifluoromethoxy)-2-pyridyl]-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (white solid). LC/MS (ESI) m/z: 491.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.13 (s, 1H), 8.02 (d, J = 8.8 Hz, 1H), 7.80 (d, J = 8.8 Hz, 1H), 7.63 - 7.54 (m, 1H), 6.53 (d, J = 9.2 Hz, 1H), 4.28 - 4.18 (m, 4H), 3.80 (d, J = 1.6 Hz, 2H), 3.50 (s, 3H), 3.25 (t, J = 9.6 Hz, 2H), 2.48 - 2.42 (m, 2H). Synthesis of Compound 290 Exemplary Synthesis of 4-[(3aS,6aS)-5-[5-(propan-2-yloxy)pyridin-2-yl]- octahydropyrrolo[3,4-c]pyrrol-2-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5- naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl (3aR,6aR)-2-(5-isopropoxy-2-pyridyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate A mixture of tert-butyl (3aR,6aR)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole-5- carboxylate (100 mg, 0.471 mmol, 1 eq), 2-bromo-5-isopropoxy-pyridine (153 mg, 0.707 mmol, 1.5 eq), cesium carbonate (460 mg, 1.41 mmol, 3 eq), 1,3-bis[2,6-bis(1- propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (46 mg, 0.047 mmol, 0.1 eq) in dioxane (2 mL) was stirred at 100 °C for 3 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate = 100/1 to 1:1) to give tert-butyl (3aR,6aR)-2-(5-isopropoxy-2- pyridyl)-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (167 mg, 89%) as a yellow solid. LC/MS (ESI) m/z: 348.0 [M+H]+. Step 2: Preparation of (3aS,6aS)-5-(5-isopropoxy-2-pyridyl)-2,3,3a,4,6,6a-hexahydro- 1H-pyrrolo[3,4-c]pyrrole A mixture of tert-butyl (3aR,6aR)-2-(5-isopropoxy-2-pyridyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (0.167 g,0.481 mmol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 28.10 eq) in dichloromethane (1 mL) was stirred at 25°C for 0.5 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure to give (3aS,6aS)-5-(5-isopropoxy-2-pyridyl)-2,3,3a,4,6,6a-hexahydro-1H-pyrrolo[3,4-c]pyrrole trifluoroacetic acid (174 mg, crude) as a brown oil. LC/MS (ESI) m/z: 248.2 [M+H]+. Step 3: Preparation of 4-[(3aS,6aS)-2-(5-isopropoxy-2-pyridyl)-1,3,3a,4,6,6a- hexahydropyrrolo[3,4-c]pyrrol-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (white solid). LCMS (ESI) m/z: 465.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.02 (d, J = 9.2 Hz, 1H), 7.84 - 7.77 (m, 2H), 7.25 (dd, J = 2.8, 8.8 Hz, 1H), 6.41 (d, J = 9.2 Hz, 1H), 4.36 (td, J = 6.0, 12.0 Hz, 1H), 4.26 - 4.16 (m, 4H), 3.73 (dd, J = 6.0, 8.8 Hz, 2H), 3.50 (s, 3H), 3.19 (t, J = 9.6 Hz, 2H), 2.46 - 2.40 (m, 2H), 1.21 (d, J = 6.0 Hz, 6H). Synthesis of Compound 291 Exemplary Synthesis of 4-[(3aR,7aS)-1-[5-(propan-2-yloxy)pyrimidin-2-yl]-octahydro- 1H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine- 3-carbonitrile Step 1: Preparation of 2-chloro-5-isopropoxy-pyrimidine To a solution of 2-chloropyrimidin-5-ol (2 g, 15.32 mmol, 1 eq) in DMF (15 mL) were added potassium carbonate (6.35 g, 45.97 mmol, 3 eq) and 2-iodopropane (5.21 g, 30.64 mmol, 3.06 mL, 2 eq), and the reaction mixture was stirred at 60 °C for 12 h. Water (50 mL) was then added, and the resulting mixture was extracted with ethyl acetate (50 mL x 3). The combined organic extracts were washed with brine (150 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=5/1) to afford 2-chloro-5- isopropoxy-pyrimidine (2.4 g, 91%) as a white solid. LC/MS (ESI) m/z: 173.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.51 (s, 2H), 4.79 (td, J = 6.0, 12.0 Hz, 1H), 1.29 (d, J = 6.0 Hz, 6H). Step 2: Preparation of benzyl (3aR,7aS)-1-(5-isopropoxypyrimidin-2-yl)-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate To a solution of 2-chloro-5-isopropoxy-pyrimidine (145 mg, 0.85 mmol, 1 eq) and benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5-carboxylate hydrogen chloride (250 mg, 0.85 m mol, 1 eq) in dioxane (3 mL) were added 1,3-bis[2,6-bis(1- propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (41 mg, 0.04 mmol, 0.05 eq) and cesium carbonate (0.8 mg, 2.53 mmol, 3 eq), and the reaction mixture was stirred at 100 °C for 3 h. The mixture was filtered, then concentrated under reduced pressure. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=3/1) to afford benzyl (3aR,7aS)-1-(5-isopropoxypyrimidin-2-yl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (170 mg, 51%) as a white solid. LC/MS (ESI) m/z: 397.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.00 - 7.96 (m, 2H), 7.27 (s, 5H), 5.11 - 5.00 (m, 2H), 4.25 - 4.08 (m, 2H), 4.07 - 3.97 (m, 2H), 3.61 - 3.39 (m, 2H), 3.33 - 3.19 (m, 1H), 2.98 - 2.77 (m, 1H), 2.43 - 2.24 (m, 1H), 2.13 (d, J = 11.6 Hz, 1H), 1.94 - 1.81 (m, 2H), 1.45 (d, J = 12.0 Hz, 1H), 1.21 (d, J = 6.0 Hz, 6H). Step 3: Preparation of (3aR,7aS)-1-(5-isopropoxypyrimidin-2-yl)-2,3,3a,4,5,6,7,7a- octahydropyrrolo[3,2-c]pyridine To a solution of benzyl (3aR,7aS)-1-(5-isopropoxypyrimidin-2-yl)-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-5-carboxylate (160 mg, 0.4 mmol, 1 eq) in methanol (5 mL) was added 10% palladium on carbon (30 mg, 0.4 mmol, 1 eq) under nitrogen, and the suspension was degassed under vacuum and purged with hydrogen several times. The reaction mixture was stirred under hydrogen (15 psi) at 25 °C for 12 hours. The mixture was filtered, then concentrated under reduced pressure to afford (3aR,7aS)-1-(5-isopropoxypyrimidin-2-yl)- 2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine (95 mg, crude) as a white oil. LC/MS (ESI) m/z: 263.2 [M+H]+. Step 4: Preparation of 4-[(3aR,7aS)-1-(5-isopropoxypyrimidin-2-yl)-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine- 3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LCMS (ESI) m/z: 480.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.17 (s, 2H), 8.07 (d, J = 9.2 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 4.39 (J = 6.0, 12.0 Hz, 1H), 4.34 - 4.26 (m, 1H), 4.20 (d, J = 13.6 Hz, 1H), 4.16 - 4.07 (m, 1H), 3.96 (dd, J = 4.4, 13.6 Hz, 1H), 3.60 - 3.55 (m, 1H), 3.54 (s, 3H), 3.50 - 3.42 (m, 2H), 2.61 (d, J = 6.0 Hz, 1H), 2.31 - 2.24 (m, 1H), 2.24 - 2.23 (m, 1H), 2.10 - 1.94 (m, 3H), 1.23 (d, J = 6.0 Hz, 6H).
Synthesis of Compound 292 Exemplary Synthesis of 2,4-dimethyl-5-oxo-7-((3aR,6aR)-5-(5- (trifluoromethoxy)pyridin-2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-4,5- dihydrothiazolo[5,4-b]pyridine-6-carbonitrile Step 1: Preparation of (3aR,6aR)-tert-butyl 5-(5-(trifluoromethoxy)pyridin-2- yl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate and (3aS, 6aS)-tert-butyl 5-(5- (trifluoromethoxy) pyridin-2-yl) hexahydropyrrolo [3, 4-c] pyrrole-2(1H)-carboxylate The racemic mixture of tert-butyl (3aR,6aR)-2-[5-(trifluoromethoxy)-2-pyridyl]- 1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate (270 mg, 0.72 mmol) was purified by prep-HPLC (column: DAICEL CHIRALPAK AD (250mm*30mm, 10um); mobile phase: 20% methanol in supercritical carbon dioxide, 20%-20%, 8; 40 min). tert-Butyl (3aR,6aR)-2-[5-(trifluoromethoxy)-2-pyridyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (135 mg,50%) was obtained as a yellow solid. LC/MS (ESI) m/z: 374.2 [M+H]+. tert-Butyl (3aS,6aS)-2-[5-(trifluoromethoxy)-2-pyridyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4- c]pyrrole-5-carboxylate (131 mg, 49$) was obtained as a yellow solid. LC/MS (ESI) m/z: 374.2 [M+H]+. Step 2: Preparation of (3aR, 6aR)-2-(5-(trifluoromethoxy) pyridin-2-yl) octahydropyrrolo [3, 4-c] pyrrole A mixture of (3aS, 6aS)-tert-butyl 5-(5-(trifluoromethoxy) pyridin-2-yl) hexahydropyrrolo [3, 4-c] pyrrole-2(1H)-carboxylate (131 mg, 0.35 mmol, 1eq) and trifluoroacetic acid (3.08 g, 27.01 mmol, 2.00 mL, 76.99 eq) in dichloromethane (2 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 1 h under nitrogen atmosphere. The mixture was concentrated to afford (3aR, 6aR)-2-(5-(trifluoromethoxy) pyridin-2-yl) octahydropyrrolo [3, 4-c] pyrrole trifluoroacetic acid (135 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 273.7 [M+H]+. Step 3: Preparation of 2,4-dimethyl-5-oxo-7-((3aR,6aR)-5-(5-(trifluoromethoxy)pyridin- 2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-4,5-dihydrothiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 7-[(3aR,7aS)-1-[5- (trifluoromethoxy)pyridin-2-yl]-octahydro-1H-pyrrolo[3,2-c]pyridin-5-yl]-2,4-dimethyl-5- oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile starting from (6-cyano-2,4-dimethyl- 5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate. (white solid). LC/MS (ESI) m/z: 476.9 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.11 (d, J = 2.4 Hz, 1H), 7.39 (d, J = 8.0 Hz, 1H), 6.36 (d, J = 8.0 Hz, 1H), 4.28-4.36 (m, 2H), 4.17-4.27 (m, 2H), 3.88 (s, 2H), 3.58 (s, 3H), 3.30 (t, J = 9.6 Hz, 2H), 2.71 (s, 3H), 2.39-2.54 (m, 2H). Synthesis of Compound 293 Exemplary Synthesis of 6-chloro-4-(4-hydroxy-4-{[4-(trifluoromethyl)pyridin-2- yl]methyl}piperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of 2-methyl-4-(trifluoromethyl) pyridine To a solution of 2-chloro-4-(trifluoromethyl) pyridine (5 g, 27.54 mmol, 1 eq) and trichloroiron (20 mg, 4.48e-3 eq) in tetrahydrofuran (20 mL) was added chloro(methyl)magnesium (3 M, 9.64 mL, 1.05 eq) over 30 min at 0 °C under nitrogen, and the reaction mixture was stirred at 25 °C for 2 h. The mixture was diluted with saturated aqueous ammonium chloride solution (50 mL) and water (40 mL), then extracted with ethyl acetate (35 mL x 3). The combined organic extracts were washed by brine and dried over sodium sulfate. HCl (1 M, 20 ml) was then added, and the resulting mixture was concentrated under reduced pressure. The residue was purified by semi-preparative reverse phase HPLC (water (hydrogen chloride) acetonitrile; B%: 1%-35%, 20 min). The selected fraction was concentrated to remove most of the acetonitrile, and the resulting suspension was lyophilized to afford 2-methyl-4-(trifluoromethyl) pyridine hydrogen chloride (80 mg, 2%) as a yellow solid. LC/MS (ESI) m/z: 162 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 9.0 (s, 1H), 7.98-7.91 (m, 2H), 3.13 (s, 3H). Step 2: Preparation of tert-butyl 4-hydroxy-4-((4-(trifluoromethyl) pyridin-2-yl) methyl) piperidine-1-carboxylate To a solution of 2-methyl-4-(trifluoromethyl) pyridine (50 mg, 0.31mmol, 1 eq) in tetrahydrofuran (1 mL) at -78 °C was added butyllithium (2.5 M, 0.273mL, 2.2 eq) dropwise for 0.5 h followed by tert-butyl 4-oxopiperidine-1-carboxylate (93 mg,0.47mmol, 1.5 eq) in tetrahydrofuran (1 mL) dropwise. And the reaction mixture was stirred at -78 °C for 0.5 h. The reaction was quenched with saturated aqueous ammonium chloride solution (5 mL), then extracted with ethyl acetate (5mL x 3). The combined organic extracts were washed with brine (5 mL x 3), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (0~50% ethyl acetate/petroleum ether gradient @ 40mL/min) to afford tert-butyl 4-hydroxy-4-((4- (trifluoromethyl) pyridin-2-yl) methyl) piperidine-1-carboxylate (51 mg, 46%) as a yellow oil. LC/MS (ESI) m/z: 305 [M-55]+; 1H NMR (400 MHz, CDCl3) δ 8.72 (d, J = 6.0 Hz, 1H), 7.46 (d, J = 4.8 Hz, 1H), 7.39 (s, 1H), 4.26-4.32 (m, 1H), 3.85 (s, 4H), 3.04 (s, 2H), 2.46-2.56 (m, 2H), 1.73-1.85 (m, 2H), 1.50 (s, 9H), 0.79-1.00 (m, 4H). Step 3: Preparation of 4-((4-(trifluoromethyl) pyridin-2-yl) methyl) piperidin-4-ol A mixture of tert-butyl 4-hydroxy-4-((4-(trifluoromethyl) pyridin-2-yl) methyl) piperidine-1- carboxylate (50 mg, 0.14mol, 1 eq) and trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 97.35 eq) in dichloromethane (1 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 1 h under nitrogen atmosphere. The mixture was concentrated to afford 4-((4- (trifluoromethyl) pyridin-2-yl) methyl) piperidin-4-ol trifluoroacetic acid (51 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 260.9 [M+H]+. Step 4: Preparation of 6-chloro-4-(4-hydroxy-4-((4-(trifluoromethyl) pyridin-2-yl) methyl) piperidin-1-yl)-1-methyl-2-oxo-1, 2-dihydro-1, 5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LCMS (ESI) m/z: 478.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.62-8.87 (m, 1H), 7.62 (d, J = 8.8 Hz, 1H), 7.46-7.54 (m, 2H), 7.38-7.45 (m, 1H), 4.16 ( d, J = 12.8 Hz, 2H), 3.89-4.04 (m, 2H), 3.62 (s, 3H), 3.13 (s, 2H), 1.90-2.01 (m, 2H), 1.81 ( d, J = 12.8 Hz, 2H). Synthesis of Compound 294 Exemplary Synthesis of 2,4-dimethyl-5-oxo-7-((3aS,6aS)-5-(5- (trifluoromethoxy)pyridin-2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-4,5- dihydrothiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7- ((3aR,6aR)-5-(5-(trifluoromethoxy)pyridin-2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)- 4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3aS,6aS)-2-[5- (trifluoromethoxy)-2-pyridyl]-1,3,3a,4,6,6a-hexahydropyrrolo[3,4-c]pyrrole-5-carboxylate. (white solid). LC/MS (ESI) m/z: 477.0 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.11 (d, J = 2.4 Hz, 1H), 7.39 (d, J = 6.8 Hz, 1H), 6.35 (d, J = 9.2 Hz, 1H), 4.27-4.34 (m, 2H), 4.17-4.25 (m, 2H), 3.87 (d, J = 5.2 Hz, 2H), 3.58 (s, 3H), 3.30 (t, J = 9.6 Hz, 2H), 2.71 (s, 3H), 2.46 (m, 2H). Synthesis of Compounds 295 amd 296 Exemplary Synthesis of 7-((3S,4R)-4-((2-methoxy-4-(trifluoromethoxy)phenyl)amino)-3- methylpiperidin-1-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6- carbonitrile and 7-[(3R,4S)-4-{[2-methoxy-4-(trifluoromethoxy)phenyl]amino}-3- methylpiperidin-1-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6- carbonitrile Step 1: Preparation of N-[2-methoxy-4-(trifluoromethoxy)phenyl]-1,1-diphenyl- methanimine
Figure imgf000547_0002
A mixture of 1-bromo-2-methoxy-4-(trifluoromethoxy)benzene (2 g, 7.38 mmol, 1eq), diphenylmethanimine (2 g, 11.07 mmol, 1.86 mL, 1.5eq), tris(dibenzylideneacetone)dipalladium (676 mg, 0.74 mmol, 0.1 eq), 2- (dicyclohexylphosphino)-2,4,6-tri-i-propyl-1,1-bipheny (704 mg, 1.48 mmol, 0.2 eq), and t- BuONa (1 M, 18.45 mL, 2.5 eq) in dioxane (20 mL) was degassed and purged with nitrogen (3X), then stirred at 100 °C for 12 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=20/1 to 10/1) to get N-[2-methoxy-4- (trifluoromethoxy)phenyl]-1,1-diphenyl-methanimine (600 mg, 23%) as a yellow solid. LC/MS (ESI) m/z: 354.2 [M+H]+. Step 2: Preparation of 2-methoxy-4-(trifluoromethoxy)aniline
Figure imgf000547_0001
A mixture of N-[2-methoxy-4-(trifluoromethoxy)phenyl]-1,1-diphenyl-methanimine (1.3 g, 3.50 mmol, 1 eq) and hydrochloride (2 M, 6.19 mL, 3.54 eq) in tetrahydrofuran (7 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 12 h under nitrogen atmosphere. The reaction was quenched with sodium carbonate and the pH adjusted to 9. Brine (30 mL) was then added, and the resulting mixture was extracted with ethyl acetate (30 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated to give compound 2-methoxy-4-(trifluoromethoxy)aniline (675 mg, 93%) as a yellow oil. LC/MS (ESI) m/z: 207.2 [M+H]+. Step 3: Preparation of tert-butyl 4-[2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl- piperidine-1-carboxylate To a solution of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (1.65 g, 7.72 mmol, 2 eq) and 2-methoxy-4-(trifluoromethoxy)aniline (800 mg, 3.86 mmol, 1.35 mL, 1 eq) in methanol (12 mL) and acetic acid (12 mL) was added 2-methylpyridine borane (1.24 g, 11.58 mmol, 3 eq), and the reaction mixture was stirred at 25 °C for 12 h . The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18 (250*70mm,10 um); mobile phase: [water(formic acid)- acetonitrile]; 43%-73%, 35min) to give tert-butyl 4-[2-methoxy-4-(trifluoromethoxy)anilino]- 3-methyl-piperidine-1-carboxylate (900 mg, 58%) as a yellow oil. LC/MS (ESI) m/z: 405.2 [M+H]+;1H NMR (400 MHz, DMSO-d6) δ 6.85 - 6.78 (m, 1H), 6.78 - 6.70 (m, 1H), 6.67 - 6.55 (m, 1H), 4.73 - 4.46 (m, 1H), 3.95 - 3.54 (m, 6H), 3.18 - 2.77 (m, 2H), 2.06 (m, J = 3.6, 6.8 Hz, 1H), 1.71 - 1.46 (m, 2H), 1.45 - 1.35 (m, 9H), 0.76 (d, J = 7.6 Hz, 3H). Step 4: Preparation of N-[2-methoxy-4-(trifluoromethoxy)phenyl]-3-methyl-piperidin-4- amine To a solution of tert-butyl 4-[2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl-piperidine-1- carboxylate (400 mg, 0.99 mmol, 1eq) in dichloromethane (5 mL) was added trifluoroacetic acid (4.62 g, 40.52 mmol, 3 mL, 40.97 eq), and the reaction mixture was stirred at 25 °C for 1h. The reaction mixture was filtered and concentrated to give N-[2-methoxy-4- (trifluoromethoxy)phenyl]-3-methyl-piperidin-4-amine trifluoroacetic acid (330 mg, crude) as a white oil. LC/MS (ESI) m/z: 304.3 [M+H]+. Step 5: Preparation of 7-[(3S,4R)-4-[2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl- 1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile and 7-[(3R,4S)-4- [2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo- thiazolo[5,4-b]pyridine-6-carbonitrile To a solution of (6-cyano-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate (278.70 mg, 0.79 mmol, 1 eq) and N-[2-methoxy-4- (trifluoromethoxy)phenyl]-3-methyl-piperidin-4-amine trifluoroacetic acid (330 mg, 0.79 mmol, 1 eq) in acetonitrile (10 mL) was added diisopropylethylamine (509.76 mg, 3.94 mmol, 687.00 uL, 5 eq), and the reaction mixture was stirred at 40 °C for 4 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by prep- HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(formic acid)- acetonitrile]; 55%-85%,10min) to afford racemic 7-[4-[2-methoxy-4- (trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-2,4-dimethyl-5-oxo-thiazolo[5,4- b]pyridine-6-carbonitrile (210 mg, 52%) as a white solid. This material was purified by SFC (column: DAICEL CHIRALCEL OJ(250mm*30mm,10um); mobile phase: 25% [0.1% ammonium hydroxide in isopropanol] in supercritical carbon dioxide: 25%-25%,5; 175min), then HPLC (column: Phenomenex Luna C18100*30mm*5um; mobile phase: [water(formic acid)- acetonitrile]; 55%-85%, 8min). The unseparated peaks were further purified by SFC (column: DAICEL CHIRALPAK AD-H(250mm*30mm,5um); mobile phase: 23% [0.1% ammonium hydroxide in ethanol] in supercritical carbon dioxid: 23%-23%,A4.5;135min) and then HPLC (column: Phenomenex Luna C18100*30mm*5um;mobile phase: water(formic acid)- acetonitrile]; 55%-85%,8min). 7-[(3S,4R)-4-[2-Methoxy-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-2,4-dimethyl- 5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (55 mg, 27%) was obtained as a white solid. LC/MS (ESI) m/z: 507.2 [M+H] +; 1H NMR (400 MHz, CDCl3) δ 6.76 (d, J = 8.8 Hz, 1H), 6.68 (s, 1H), 6.61 (d, J = 8.4 Hz, 1H), 4.25 - 4.14 (m, 1H), 4.14 - 4.05 (m, 1H), 3.96 - 3.90 (m, 1H), 3.88 (s, 3H), 3.84 - 3.78 (m, 1H), 3.77 - 3.70 (m, 1H), 3.59 (s, 3H), 2.72 (s, 3H), 2.46 - 2.36 (m, 1H), 2.23 - 2.10 (m, 1H), 2.04 - 1.93 (m, 1H), 0.97 (d, J = 6.8 Hz, 3H) 7-[(3R,4S)-4-[2-Methoxy-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-2,4-dimethyl- 5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile (55 mg, 27%) was obtained as a white solid. LC/MS (ESI) m/z: 507.2 [M+H] +; 1H NMR (400 MHz, CDCl3) δ 6.75 ( d, J = 8.4 Hz, 1H), 6.67 (s, 1H), 6.56 (d, J = 8.8 Hz, 1H), 4.25 - 4.14 (m, 1H), 4.12 - 4.04 (m, 1H), 3.97 - 3.91 (m, 1H), 3.88 (s, 3H), 3.85 - 3.78 (m, 1H), 3.77 - 3.68 (m, 1H), 3.59 (s, 3H), 2.72 (s, 3H), 2.47 - 2.34 (m, 1H), 2.21 - 2.10 (m, 1H), 2.02 - 1.92 (m, 1H), 0.96 (d, J = 6.4 Hz, 3H).
Synthesis of Compound 297 Exemplary Synthesis of 6-chloro-4-((3aS,7aR)-1-(4-chlorophenyl)octahydro-5H- pyrrolo[3,2-c]pyridin-5-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile Step 1: Preparation of (3aR,7aS)-benzyl 1-(4-chlorophenyl)hexahydro-1H-pyrrolo[3,2- c]pyridine-5(6H)-carboxylate and (3aS, 7aR)-benzyl 1-(4-chlorophenyl) hexahydro-1H- pyrrolo [3, 2-c] pyridine-5(6H)-carboxylate A mixture of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate hydrogen chloride (658 mg, 2.22 mmol, 1 eq), 1-bromo-4-chloro-benzene (1.27 g, 6.65 mmol, 1.27 mL, 3 eq), cesium carbonate (3.61 g, 11.09 mmol, 5 eq), and 1,3-bis[2,6- bis(1-propylbutyl)phenyl]-4, 5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (107 mg, 0.01 mmol, 0.05 eq) in dioxane (6 mL) was degassed and purged with nitrogen (3X), then stirred at 100 °C for 12 h under nitrogen atmosphere. The mixture was filtered and concentrated. The residue was purified by flash silica gel chromatography (0~25% ethyl acetate/petroleum ether gradient @ 40 mL/min) followed by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water (formic acid) in acetonitrile 70%-100%, 9 min) to afford benzyl (3aR,7aS)-1-(4- chlorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate (162 mg, 20%) as a white oil. LC/MS (ESI) m/z: 371.2 [M+H]+. This material was further purified by prep-HPLC (column: DAICEL CHIRALCEL OJ (250mm*30mm, 10um); mobile phase: 45% [0.1% ammonium hydroxide methanol] in supercritical carbon dioxide: 45%-45%, A5.5; 55 min). Benzyl (3aR,7aS)-1-(4-chlorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5- carboxylate (67 mg) was obtained as a yellow solid. LC/MS (ESI) m/z: 371.1 [M+H]+. Benzyl (3aS, 7aR)-1-(4-chlorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo [3, 2-c] pyridine- 5-carboxylate (63 mg) was obtained as a yellow solid. LC/MS (ESI) m/z: 371.1 [M+H]+. Step 2: Preparation of (3aS, 7aR)-1-(4-chlorophenyl) octahydro-1H-pyrrolo [3, 2-c] pyridine A mixture of benzyl (3aS,7aR)-1-(4-chlorophenyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (60 mg, 0.16 mmol, 1 eq) and hydrogen bromide (765 mg, 2.76 mmol, 0.5 mL, 30% purity, 17.07 eq) in dichloromethane (1 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 0.1 h under nitrogen atmosphere. The mixture was concentrated, and the pH adjusted to 7-8 by addition of saturated aqueous NaHCO3 solution (2 mL). The resulting mixture was extracted with ethyl acetate (15 mL), and the combined organic extracts were washed with brine (10 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford (3aS, 7aR)-1-(4-chlorophenyl)- 2,3,3a,4,5,6,7,7a-octahydropyrrolo [3, 2-c] pyridine (31 mg, crude) as a yellow solid. LC/MS (ESI) m/z: 237.0 [M+H]+. Step 3: Preparation of 6-chloro-4-((3aS, 7aR)-1-(4-chlorophenyl) hexahydro-1H-pyrrolo [3, 2-c] pyridin-5(6H)-yl)-1-methyl-2-oxo-1, 2-dihydro-1, 5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (white solid). LCMS (ESI) m/z: 453.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 9.2 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 7.19 (d, J = 8.8 Hz, 2H), 6.63 (d, J = 9.2 Hz, 2H), 4.25 (d, J = 13.2 Hz, 1H), 4.14 (d, J = 12.0 Hz, 1H), 4.02-4.10 (m, 1H), 3.95 (dd, J = 13.6, 4.4 Hz, 1H), 3.55 (s, 3H), 3.41-3.51 (m, 1H), 3.38 (s, 1H), 3.25 (m, J = 8.8 Hz, 1H), 2.62 (d, J = 3.2 Hz, 1H), 2.18-2.30 (m, 1H), 2.06-2.16 (m, 2H), 1.75-1.90 (m, 1H). Synthesis of Compounds 298 amd 299 Exemplary Synthesis of 6-chloro-4-((3R,4S)-4-((2-fluoro-4- (trifluoromethoxy)phenyl)amino)-3-methylpiperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile and 6-chloro-4-((3S,4R)-4-((2-fluoro-4- (trifluoromethoxy)phenyl)amino)-3-methylpiperidin-1-yl)-1-methyl-2-oxo-1,2-dihydro- 1,5-naphthyridine-3-carbonitrile Step 1: Preparation of N-[2-fluoro-4-(trifluoromethoxy)phenyl]-1,1-diphenyl- methanimine A mixture of diphenylmethanimine (1.05 g, 5.79 mmol, 0.97 mL, 1.5 eq), 1-bromo-2-fluoro- 4-(trifluoromethoxy)benzene (1 g, 3.86 mmol, 1 eq), tris(dibenzylideneacetone)dipalladium (353 mg, 0.386 mmol, 0.1 eq), 2-(dicyclohexylphosphino)-2,4,6-tri-i-propyl-1,1-biphenyl (368 mg, 0.77 mmol, 0.2 eq), and sodium tert-butoxide (1 M, 9.65 mL, 2.5 eq) in dioxane (10 mL) was degassed and purged with nitrogen (3X), then stirred at 100 °C for 24 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated to get N-[2-fluoro- 4-(trifluoromethoxy)phenyl]-1,1-diphenyl-methanimine (1 g, crude) as a yellow solid. LC/MS (ESI) m/z: 359.32 [M+H] +; 1H NMR (400 MHz, DMSO-d6) δ7.78 - 7.71 (m, 1H), 7.61 - 7.53 (m, 2H), 7.53 - 7.46 (m, 2H), 7.42 - 7.32 (m, 3H), 7.32 - 7.31 (m, 1H), 7.31 - 7.22 (m, 1H), 7.19 - 7.09 (m, 2H), 7.07 - 7.01 (m, 1H). Step 2: Preparation of 2-fluoro-4-(trifluoromethoxy)aniline A mixture of N-[2-fluoro-4-(trifluoromethoxy)phenyl]-1,1-diphenyl-methanimine (1 g, 2.78 mmol, 1 eq) and hydrochloride (2 M, 10.00 mL, 7.19 eq) in tetrahydrofuran (10 mL) was degassed and purged with nitrogen (3X), then stirred at 25 °C for 12 h under nitrogen atmosphere. The reaction was quenched by sodium carbonate, adjusted the pH to 9, then diluted with brine (30 mL). The resulting mixture was extracted with ethyl acetate (30 mL), and the combined organic extracts were dried over anhydrous sodium sulfate and concentrated. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=10/1 to 5/1) to get 2-fluoro-4-(trifluoromethoxy)aniline (320 mg, 59%) as a yellow oil. LC/MS (ESI) m/z: 195.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.12 (m, J = 1.6 Hz, 1H), 6.95 - 6.86 (m, 1H), 6.79 (t, J = 8.4 Hz, 1H), 5.35 (s, 2H). Step 3: Preparation of tert-butyl 4-[2-fluoro-4-(trifluoromethoxy)anilino]-3-methyl- piperidine-1-carboxylate To a solution of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (656 mg, 3.08 mmol, 2 eq) and 2-fluoro-4-(trifluoromethoxy)aniline (300 mg, 1.54 mmol, 1.35 mL, 1 eq) in methanol (3 mL) and acetic acid (2 mL) was added 2-methylpyridine borane (493 mg, 4.61 mmol, 3 eq), and the reaction mixture was stirred at 25 °C for 12 h. The reaction was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: YMC Triart C18250*50mm*7um; mobile phase: [water(formic acid)- acetonitrile];B%: 51%-91%, 25 min) to get tert-butyl 4-[2-fluoro-4- (trifluoromethoxy)anilino]-3-methyl-piperidine-1-carboxylate (410 mg, 68%) as a white solid. LC/MS (ESI) m/z: 392.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.25 - 7.13 (m, 1H), 7.03 - 6.93 (m, 1H), 6.90 - 6.79 (m, 1H), 5.49 - 5.25 (m, 1H), 3.98 - 3.64 (m, 3H), 3.13 - 2.87 (m, 2H), 2.06 (m, J = 6.4 Hz, 1H), 1.89 - 1.67 (m, 1H), 1.60 - 1.46 (m, 1H), 1.44 - 1.38 (m, 9H), 0.89 - 0.74 (m, 3H). Step 4: Preparation of N-[2-fluoro-4-(trifluoromethoxy)phenyl]-3-methyl-piperidin-4- amine. To a solution of tert-butyl 4-[2-fluoro-4-(trifluoromethoxy)anilino]-3-methyl-piperidine-1- carboxylate (50 mg, 0.13 mmol, 1 eq) in dichloromethane (0.5 mL) was added trifluoroacetic acid (462 mg, 4.05 mmol, 0.3 mL, 31.80 eq), and the reaction mixture was stirred at 25 °C for 1 h. The mixture was filtered and concentrated to give N-[2-fluoro-4- (trifluoromethoxy)phenyl]-3-methyl-piperidin-4-amine trifluoroacetic acid (50 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 293.3 [M+H] +. Step 4: Preparation of 6-chloro-4-[(3S,4R)-4-[2-fluoro-4-(trifluoromethoxy)anilino]-3- methyl-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile and 6-chloro-4- [(3S,4S)-4-[2-fluoro-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile To a solution of N-[2-fluoro-4-(trifluoromethoxy)phenyl]-3-methyl-piperidin-4-amine (50 mg, 0.12 mmol, 1 eq, trifluoroacetic acid) and 4,6-dichloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (31 mg, 0.12 mmol, 1 eq) in acetonitrile (0.5 mL) was added diisopropylethylamine (79 mg, 0.62 mmol, 0.11 mL, 5 eq), and the reaction mixture was stirred at 40 °C for 4 h. The mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(formic acid)- acetonitrile]; B%: 63%-93%, 10 min) to get 6- chloro-4-[4-[2-fluoro-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (60 mg) as a yellow solid. This material was purified by SFC (column: DAICEL CHIRALPAK AD(250mm*30mm,10um); mobile phase: 35% [0.1%ammonium hydroxide ethanol] in supercritical CO2: 35%-35%,A3.8; 60 min), then SFC (column: DAICEL CHIRALCEL OD(250mm*30mm,10um); mobile phase: 50% [0.1%ammonium hydroxide ethanol] in supercritical CO2: 50%-50%,A2.8; 33 min). 6-Chloro-4-[(3S,4R)-4-[2-fluoro-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (18 mg, 35%) was obtained as a white solid. LC/MS (ESI) m/z: 508.2 [M+H] +; HNMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8Hz, 1H), 7.25 - 7.17 (m, 1H), 7.05 - 6.92 (m, 2H), 5.58 - 5.50 (m, 1H), 4.19 - 4.08 (m, 1H), 4.04 - 3.93 (m, 1H), 3.92 - 3.79 (m, 2H), 3.61 (t, J = 9.6 Hz, 1H), 3.53 (s, 3H), 2.33 (s, 1H), 2.28 - 2.17 (m, 1H), 1.90 - 1.80 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H) 6-chloro-4-[(3S,4S)-4-[2-fluoro-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (16 mg, 32%) was obtained as a white solid. LC/MS (ESI) m/z: 508.2 [M+H] +; 1HNMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1H), 7.20 (m, J = 12.4 Hz, 1H), 7.06 - 6.89 (m, 2H), 5.54 (m, J = 7.6 Hz, 1H), 4.13 (d, J = 12.8 Hz, 1H), 4.04 - 3.93 (m, 1H), 3.91 - 3.77 (m, 2H), 3.60 (t, J = 9.6 Hz, 1H), 3.53 (s, 3H), 2.33 (s, 1H), 2.28 - 2.16 (m, 1H), 1.89 - 1.80 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H). Synthesis of Compound 300 Exemplary Synthesis of 7-{4-[(5-chloropyridin-2-yl)methyl]-4-hydroxypiperidin-1-yl}- 2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine-6-carbonitrile
Figure imgf000556_0001
The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7- ((3aR,6aR)-5-(5-(trifluoromethoxy)pyridin-2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)- 4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile starting from 4-[(5-chloro-2- pyridyl)methyl]piperidin-4-ol. (white solid). LC/MS (ESI) m/z: 430.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.54 (d, J = 2.4 Hz, 1H), 7.85 (dd, J = 2.4, 8.4 Hz, 1H), 7.38 (d, J = 8.4 Hz, 1H), 4.83 (s, 1H), 4.15 (d, J = 12.8 Hz, 2H), 3.66 (t, J = 10.8 Hz, 2H), 3.46 (s, 3H), 2.92 (s, 2H), 2.68 (s, 3H), 1.80 - 1.70 (m, 2H), 1.59 (d, J = 13.6 Hz, 2H). Synthesis of Compound 301 Exemplary Synthesis of 6-chloro-4-((3aS,7aR)-1-(5-chloropyridin-2-yl)octahydro-5H- pyrrolo[3,2-c]pyridin-5-yl)-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile Step 1: Preparation of benzyl (3aR,7aS)-1-(5-chloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-5-carboxylate and benzyl (3aS,7aR)-1-(5-chloro-2-pyridyl)- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate A mixture of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate trifluoroacetic acid (727 mg, 1.94 mmol, 1 eq), 5-chloro-2-fluoro-pyridine (383 mg, 2.91 mmol, 1.5 eq), and cesium carbonate (2.53 g, 7.77 mmol, 4 eq) in DMF (8 mL) was stirred at 80 °C for 12 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: YMC Triart C18250*50mm*7um; mobile phase: [water(formic acid) in acetonitrile]: 50%-95%, 25min) followed by SFC (column: DAICEL CHIRALCEL OJ(250mm*30mm,10um); mobile phase:40% [0.1% ammonium hydroxide in ethanol] in supercritical carbon dioxide; 40%- 40%,6.0;84min). (Benzyl (3aR,7aS)-1-(5-chloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (108 mg, 15%) was obtained as a yellow solid. LC/MS (ESI) m/z: 372.0 [M+H]+. (Benzyl (3aS,7aR)-1-(5-chloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-5-carboxylate (103 mg, 14%) was obtained as a yellow solid. LC/MS (ESI) m/z: 372.0 [M+H]+. Step 2: Preparation of (3aS,7aR)-1-(5-chloro-2-pyridyl)-2,3,3a,4,5,6,7,7a- octahydropyrrolo[3,2-c]pyridine A mixture of benzyl (3aS,7aR)-1-(5-chloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (40 mg, 0.108 mmol, 1 eq) and 33% hydrogen bromide (851 mg, 3.47 mmol, 0.57 mL, 32.28 eq) in dichloromethane (1 mL) was degassed and purged with nitrogen (3X), then stirred at 25°C for 0.5 h under nitrogen atmosphere. The reaction was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(ethyl acetate) in acetonitrile]: 0%-17%, 9min) to give (3aS,7aR)-1-(5-chloro-2-pyridyl)- 2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine (25 mg, 97%) as colorless oil. LC/MS (ESI) m/z: 238.1 [M+H]+. Step 3: Preparation of 4-[(3aS,7aR)-1-(5-chloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LCMS (ESI) m/z: 455.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.15 - 8.02 (m, 2H), 7.81 (d, J = 9.2 Hz, 1H), 7.64 - 7.47 (m, 1H), 6.53 (d, J = 9.2Hz, 1H), 4.37 - 4.18 (m, 2H), 4.18 - 4.04 (m, 1H), 3.98 - 3.87 (m, 1H), 3.53 (s, 3H), 3.51 - 3.46 (m, 1H), 3.45 - 3.36 (m, 2H), 2.61 (s, 1H), 2.30 - 2.24 (m, 1H), 2.13 - 2.06 (m, 2H), 1.95 - 1.82 (m, 1H). Synthesis of Compounds 302 amd 303 Exemplary Synthesis of 6-chloro-4-[(3R,4S)-4-[2-methoxy-4-(trifluoromethoxy)anilino]- 3-methyl-1-piperidyl]-1-methyl-2-oxo-quinoline-3-carbonitrile and 6-chloro-4-[(3S,4R)- 4-[2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1-methyl-2-oxo- quinoline-3-carbonitrile
Figure imgf000559_0001
The title compound was prepared in an analogous manner to 6-chloro-1-methyl-4-[(3R,4S)-3- methyl-4-{[4-(trifluoromethoxy)phenyl]amino}piperidin-1-yl]-2-oxo-1,2-dihydroquinoline- 3-carbonitrile starting from N-[2-methoxy-4-(trifluoromethoxy)phenyl]-3-methyl-piperidin- 4-amine. The enantiomers were separated by SFC (column: DAICEL CHIRALPAK AD (250mm*30mm,10um); mobile phase: 35% [0.1%ammonium hydroxide in ethanol] in supercritical carbon dioxide: 35%-35%,A3.8; 60min), followed by SFC (column: DAICEL CHIRALCEL OD (250mm*30mm,10um); mobile phase: 25% [0.1% ammonium hydroxide in methanol] in supercritical carbon dioxide: 25%-25%,C;10; 160min). 6-Chloro-4-[(3R,4S)-4-[2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1- methyl-2-oxo-quinoline-3-carbonitrile(26 mg, 9%) was obtained as a white solid. LC/MS (ESI) m/z: 520.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.84 - 7.76 (m, 2H), 7.61 (d, J = 8.8 Hz, 1H), 6.85 (s, 1H), 6.80 - 6.71 (m, 2H), 4.78 (d, J = 8.4 Hz, 1H), 3.85 (s, 3H), 3.84 - 3.73 (m, 3H), 3.56 (s, 4H), 3.50 - 3.42 (m, 1H), 2.38 - 2.28 (m, 1H), 2.19 - 2.04 (m, 1H), 1.92 - 1.82 (m, 1H), 0.89 (d, J = 6.8 Hz, 3H). 6-Chloro-4-[(3S,4R)-4-[2-methoxy-4-(trifluoromethoxy)anilino]-3-methyl-1-piperidyl]-1- methyl-2-oxo-quinoline-3-carbonitrile (31 mg, 10% ) was obtained as a white solid. LC/MS (ESI) m/z: 520.9 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 7.85 - 7.75 (m, 2H), 7.61 (d, J = 8.8 Hz, 1H), 6.85 (s, 1H), 6.80 - 6.67 (m, 2H), 4.78 (d, J = 8.4 Hz, 1H), 3.85 (s, 3H), 3.84 - 3.72 (m, 3H), 3.56 (s, 4H), 3.50 - 3.41 (m, 1H), 2.34 (s, 1H), 2.19 - 2.04 (m, 1H), 1.95 - 1.81 (m, 1H), 0.89 (d, J = 6.8 Hz, 3H). Synthesis of Compound 304 Exemplary Synthesis of 6-chloro-4-{4-[(4-chlorophenyl)methyl]-4-hydroxypiperidin-1- yl}-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl 4-[(4-chlorophenyl)methyl]-4-hydroxy-piperidine-1- carboxylate To a mixture of bromo-(4-chlorophenyl)magnesium (1 M, 2.81 mL, 1.2 eq) in THF (5 mL) was added bromocopper;methylsulfanylmethane (48 mg, 0.23 mmol, 0.1 eq), and the resulting mixture was stirred at 0 °C for 15 min. tert-Butyl 1-oxa-6-azaspiro[2.5]octane-6- carboxylate (0.5 g, 2.34 mmol, 1 eq) was then added, and the reaction mixture was stirred at 25 °C for 12 h. The reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (15 mL x 2). The combined organic extracts were washed with brine (15 mL x 2), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (petroleum ether: ethyl acetate=5:1) to afford tert-butyl 4-[(4-chlorophenyl)methyl]-4-hydroxy-piperidine-1-carboxylate (680 mg, 89%) as a white solid. LC/MS (ESI) m/z: 252.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.34 - 7.28 (m, 2H), 7.26 - 7.21 (m, 2H), 4.41 (s, 1H), 3.64 (d, J = 12.4 Hz, 2H), 3.02 (s, 2H), 2.67 (s, 2H), 1.42 - 1.39 (m, 2H), 1.37 (s, 9H), 1.33 (d, J = 4.4 Hz, 2H). Step 2: Preparation of 4-[(4-chlorophenyl)methyl]piperidin-4-ol To tert-butyl 4-[(4-chlorophenyl)methyl]-4-hydroxy-piperidine-1-carboxylate (100 mg, 0.31 mmol, 1 eq) in dichloromethane (1 mL) was added trifluoroacetic acid (770.00 mg, 6.75 mmol, 0.5 mL, 22.00 eq), and the reaction mixture was stirred at 25 °C for 15 min. The mixture was concentrated under vacuum to afford 4-[(4-chlorophenyl)methyl]piperidin-4-ol trifluoroacetic acid (120 mg, crude) as a red oil. LC/MS (ESI) m/z: 225.9 [M+H]+. Step 3: Preparation of 6-chloro-4-[4-[(4-chlorophenyl)methyl]-4-hydroxy-1-piperidyl]-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (off-white solid). LCMS (ESI) m/z: 433.3 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.04 (d, J = 9.2 Hz, 1H), 7.78 (d, J = 9.2 Hz, 1H), 7.37 - 7.30 (m, 2H), 7.28 - 7.25 (m, 2H), 4.64 (s, 1H), 3.97 (d, J = 13.2 Hz, 2H), 3.67 (t, J = 11.2 Hz, 2H), 3.51 (s, 3H), 2.76 (s, 2H), 1.87 - 1.75 (m, 2H), 1.56 (d, J = 13.2 Hz, 2H). Synthesis of Compound 305 Exemplary Synthesis of 2,4-dimethyl-7-[(3S,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2- pyridyl]amino]-1-piperidyl]-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrileylate Step 1: Preparation of tert-butyl 4-amino-3-methyl-piperidine-1-carboxylate A mixture of tert-butyl 3-methyl-4-oxo-piperidine-1-carboxylate (3 g, 14.07 mmol, 1 eq), ammonium acetate (10.84 g, 140.66 mmol, 10 eq) and sodium cyanoborohydride (1.33 g, 21.10 mmol, 1.5 eq) in methanol (30 mL) was degassed and purged with nitrogen (3X), then stirred at 30 °C for 12 h under nitrogen atmosphere. The mixture was concentrated under vacuum, then diluted with water (25 mL) and extracted with ethyl acetate (25 mLx3). The combined organic extracts were washed with brine (50 mL), dried with sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex C18150*25mm*10um; mobile phase: [water (ammonium bicarbonate) in acetonitrile]; 13%-43%, 8min) to give tert-butyl 4-amino-3-methyl- piperidine-1-carboxylate (1.87 g, 62%) as a colorless oil. LC/MS (ESI) m/z: 159.2 [M-55]+; 1H NMR (400 MHz, CDCl3) δ 4.06 - 2.99 (m, 3H), 2.89 - 2.71 (m, 1H), 2.55 - 2.08 (m, 1H), 2.00 (s, 1H), 1.84 - 1.52 (m, 2H), 1.45 (s, 9H), 1.09 - 0.98 (m, 3H). Step 2: Preparation of tert-butyl (3S,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2- pyridyl]amino]piperidine-1-carboxylate, tert-butyl (3S,4S)-3-methyl-4-[[5- (trifluoromethoxy)-2-pyridyl]amino]piperidine-1-carboxylate, tert-butyl (3R,4S)-3- methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1-carboxylate, tert-butyl (3R,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1-carboxylate A mixture of tert-butyl 4-amino-3-methyl-piperidine-1-carboxylate (1.9 g, 8.76 mmol, 1.41 eq), 2-bromo-5-(trifluoromethoxy)pyridine (1.5 g, 6.21 mmol, 1 eq), sodium tert-butoxide (1 M, 18.6 mL, 3 eq), and [2-(2-aminophenyl)phenyl]-methylsulfonyloxy-palladium;ditert- butyl-[2-(2,4,6-triisopropylphenyl)phenyl]phosphane (492 mg, 0.62 mmol, 0.1 eq) in 2- methyl-2-butanol (6 mL) was degassed and purged with nitrogen (3X), then stirred at 80 °C for 6 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1 to 1:1), then further purified by prep-HPLC (column: Welch Ultimate XB-CN 250*50*10um; mobile phase: [Hexane in ethanol]; 5%-30%, 30 min) to give racemic tert-butyl-3-methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1- carboxylate (1.34 g, 41%) as a brown solid. LC/MS (ESI) m/z: 376.1 [M+H]+. This material was separated by SFC (column: DAICEL CHIRALPAK AD-H(250mm*30mm, 5um); mobile phase: 7% [0.1% ammonium hydroxide ethanol] in supercritical carbon dioxide; 7%- 7%; 600min), then SFC (column: DAICEL CHIRALPAK IG (250mm*30mm, 10um); mobile phase: 25% acetonitrile in supercritical carbon dioxide; 25%-25%, 40 min). tert-Butyl (3S,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1- carboxylate (60 mg) was obtained as colorless oil. LC/MS (ESI) m/z: 376.0 [M+H]+. tert-Butyl (3S,4S)-3-methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1- carboxylate (60 mg) was obtained as colorless oil. LC/MS (ESI) m/z: 376.1 [M+H]+. tert-Butyl (3R,4S)-3-methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1- carboxylate, 63 mg) was obtained as colorless oil. LC/MS (ESI) m/z: 376.0 [M+H]+. tert-Butyl (3R,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2-pyridyl]amino]piperidine-1- carboxylate (67 mg) was obtained as colorless oil. MS (ESI) m/z: 376.1 [M+H]+. Step 3: Preparation of N-[(3S,4R)-3-methyl-4- piperidyl]-5-(trifluoromethoxy)pyridin-2- amine A mixture of tert-butyl (3S,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2- pyridyl]amino]piperidine-1-carboxylate (20 mg, 0.05 mmol, 1 eq) and trifluoroacetic acid (770 mg, 6.75 mmol, 0.50 mL, 126.75 eq) in dichloromethane (1 mL) was degassed and purged with nitrogen for 3 times, then the mixture was stirred at 25°C for 0.5 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to give N-[(3S,4R)-3-methyl-4-piperidyl]-5-(trifluoromethoxy)pyridin-2-amine trifluoroacetic acid (21 mg, crude) as a yellow solid. LC/MS (ESI) m/z: 276.0 [M+H]+. Step 4: Preparation of 2,4-dimethyl-7-[(3S,4R)-3-methyl-4-[[5-(trifluoromethoxy)-2- pyridyl]amino]-1-piperidyl]-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrileylate The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7- ((3aR,6aR)-5-(5-(trifluoromethoxy)pyridin-2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)- 4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile starting from (6-cyano-2,4-dimethyl-5-oxo- thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate. (yellow solid). LC/MS (ESI) m/z: 479.1 [M+H]+; 1H NMR (400MHz, DMSO-d6) δ 8.00 (d, J = 2.8 Hz, 1H), 7.44 (dd, J = 2.4, 9.2 Hz, 1H), 6.98 (d, J = 8.0 Hz, 1H), 6.67 (d, J = 9.2 Hz, 1H), 4.28 (tt, J = 4.0, 8.0 Hz, 1H), 3.96-4.01(m, 2H), 3.90 - 3.80 (m, 2H), 3.48 (s, 3H), 2.70 (s, 3H), 2.25-2.27 (m, 1H), 2.03 - 1.93 (m, 1H), 1.88 - 1.77 (m, 1H), 0.85 (d, J = 6.8 Hz, 3H). Synthesis of Compound 306 Exemplary Synthesis of 4-[(3aR,7aS)-1-(3,5-dichloropyridin-2-yl)-octahydro-1H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile Step 1: Preparation of benzyl (3aR,7aS)-1-(3,5-dichloropyridin-2-yl)octahydro-5H- pyrrolo[3,2-c]pyridine-5-carboxylate A mixture of benzyl (3aR,7aS)-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine-5- carboxylate trifluoroacetic acid (51 mg, 0.14 mmol, 1 eq), 3,5-dichloro-2-fluoro-pyridine (45 mg, 0.27 mmol, 2 eq), and N-ethyl-N-isopropyl-propan-2-amine (53 mg, 0.41 mmol, 0.07 mL, 3 eq) in DMSO (1 mL) was stirred at 80 °C for 12 h. Water (10 mL) was then added, and the resulting mixture was extracted with ethyl acetate (10 mL x 2). The combined organic extracts were washed with brine (20 mL), dried with sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=0/1 to 10:1) to give benzyl (3aR,7aS)-1-(3,5-dichloro-2- pyridyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate (50 mg, 90%) as a colorless oil. LC/MS (ESI) m/z: 405.9 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.03 (d, J = 2.4 Hz, 1H), 7.52 (d, J = 2.4 Hz, 1H), 7.40 - 7.30 (m, 5H), 5.15 (d, J = 2.0 Hz, 2H), 4.44 (q, J = 5.6 Hz, 1H), 4.11 - 4.01 (m, 1H), 3.86 - 3.67 (m, 1H), 3.50 - 3.33 (m, 4H), 2.43 (s, 1H), 1.98 - 1.76 (m, 4H). Step 2: Preparation of (3aR,7aS)-1-(3,5-dichloro-2-pyridyl)-2,3,3a,4,5,6,7,7a- octahydropyrrolo[3,2-c]pyridine A mixture of benzyl (3aR,7aS)-1-(3,5-dichloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-5-carboxylate (50 mg, 0.12 mmol, 1 eq) and 33% hydrogen bromide (745 mg, 3.04 mmol, 0.5 mL, 24.69 eq) in dichloromethane (1 mL) was stirred at 20 °C for 0.5 h. The mixture was concentrate, then diluted with water (1 mL). The resulting aqueous mixture was treated with saturated NaHCO3 to adjust the pH to 7~8, then extracted with ethyl acetate (10 mL x 2). The combined organic extracts were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure to give (3aR,7aS)-1-(3,5- dichloro-2-pyridyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine (35 mg, crude) as a white solid. LC/MS (ESI) m/z: 271.9 [M+H]+. Step 3: Preparation of 4-[(3aR,7aS)-1-(3,5-dichloro-2-pyridyl)-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 6-chloro-1-methyl-2-oxo-4- spiro[indane-2,4'-piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile starting from 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile. (yellow solid). LCMS (ESI) m/z: 490.8 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.18 (d, J = 2.0 Hz, 1H), 8.07 (d, J = 9.2 Hz, 1H), 7.92 (d, J = 2.0 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 4.56 - 4.48 (m, 1H), 4.11 - 4.02 (m, 2H), 3.78 - 3.69 (m, 2H), 3.67 - 3.60 (m, 1H), 3.53 (s, 3H), 3.45 (td, J = 4.4, 9.2 Hz, 1H), 2.74 - 2.68 (m, 1H), 2.11 - 1.98 (m, 3H), 1.87 - 1.79 (m, 1H). Synthesis of Compound 307 Exemplary Synthesis of 7-[(3aS,7aR)-5-[5-(trifluoromethoxy)pyridin-2-yl]-octahydro- 1H-pyrrolo[3,2-c]pyridin-1-yl]-2,4-dimethyl-5-oxo-4H,5H-[1,3]thiazolo[5,4-b]pyridine- 6-carbonitrile Step 1: Preparation of tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate and tert-butyl (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-1-carboxylate To a solution of tert-butyl (3aR,7aS)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine-1- carboxylate (200 mg, 0.88 mmol, 1 eq) and 2-bromo-5-(trifluoromethoxy) pyridine (214 mg, 0.88 mmol, 1 eq) in dioxane (3 mL) were added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (86 mg, 0.88 mmol, 0.1 eq) and cesium carbonate (864 mg, 2.65 mmol, 3 eq), and the reaction mixture was stirred at 100 °C for 3 h. The reaction mixture was partitioned between water (5 mL) and ethyl acetate (15 mL). The organic phase was separated, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=1/0 to 5/1) to afford tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2- c]pyridine-1-carboxylate (66 mg, 19%) as a green oil. LC/MS (ESI) m/z: 387.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 8.08 (d, J = 2.0 Hz, 1H), 7.36 (d, J = 7.2 Hz, 1H), 6.55 (d, J = 9.2 Hz, 1H), 4.07 - 3.73 (m, 3H), 3.65 - 3.48 (m, 1H), 3.46 - 3.29 (m, 2H), 3.17 - 2.98 (m, 1H), 2.50 (s, 1H), 2.29 - 2.08 (m, 1H), 1.92 (dt, J = 6.8, 12.4 Hz, 1H), 1.86 - 1.70 (m, 2H), 1.48 (s, 9H). The material was separated by SFC (condition: column: REGIS(S,S)WHELK- O1(250mm*25mm,10um); mobile phase: 10% [0.1% ammonium hydroxide in ethanol] in supercritical CO2: 10%-10%,C6; 160min), then further separated by SFC (column: DAICEL CHIRALPAK AD (250mm*30mm,10um); mobile phase:10% [0.1% ammonium hydroxide in ethanol] in supercritical CO2, 10%-10%,c10; 60min). tert-Butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-1-carboxylate (66 mg, 19%) was obtained as a green oil. tert-Butyl (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-1-carboxylate (50 mg, 15%) was obtained as a green oil. Step 2: Preparation of (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a- octahydropyrrolo[3,2-c]pyridine To a solution of tert-butyl (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (80 mg, 0.21 mmol, 1 eq) in dichloromethane (1.5 mL) was added trifluoroacetic acid (1.23 g, 10.80 mmol, 0.8mL, 52.32 eq). The mixture was stirred at 25 °C for 0.5 h, then concentrated under reduced pressure to afford (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2- c]pyridine trifluoroacetic acid (83 mg, crude) as a yellow oil. LC/MS (ESI) m/z: 288.0 [M+H]+. Step 3: Preparation of 7-[(3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-2,4-dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6- carbonitrile The title compound was prepared in an analogous manner to 2,4-dimethyl-5-oxo-7- ((3aR,6aR)-5-(5-(trifluoromethoxy)pyridin-2-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)- 4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile starting from (6-cyano-2,4-dimethyl-5-oxo- thiazolo[5,4-b]pyridin-7-yl) trifluoromethanesulfonate. (white solid). LC/MS (ESI) m/z: 491.4 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 2.8 Hz, 1H), 7.57 (dd, J = 2.4, 9.2 Hz, 1H), 6.87 (d, J = 9.2 Hz, 1H), 5.54 - 5.42 (m, 1H), 4.41 - 4.25 (m, 1H), 4.06 - 3.92 (m, 2H), 3.92 - 3.83 (m, 1H), 3.56 (dd, J = 4.4, 13.6 Hz, 1H), 3.46 (s, 3H), 3.16 - 3.04 (m, 1H), 2.69 (s, 3H), 2.61 - 2.56 (m, 1H), 2.16 - 2.05 (m, 1H), 2.05 - 1.96 (m, 1H), 1.92 - 1.82 (m, 1H), 1.76 - 1.61 (m, 1H). Synthesis of Compound 308 Exemplary Synthesis of 2,4-dimethyl-5-oxo-7-((3aR,7aS)-5-(5- (trifluoromethoxy)pyridin-2-yl)octahydro-1H-pyrrolo[3,2-c]pyridin-1-yl)-4,5- dihydrothiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3aS,7aR)-5-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from tert-butyl (3aR,7aS)-5- [5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1- carboxylate. (white solid). LC/MS (ESI) m/z: 491.4 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.12 (d, J = 2.8 Hz, 1H), 7.58 (dd, J = 2.4, 8.8 Hz, 1H), 6.87 (d, J = 9.2 Hz, 1H), 5.54 - 5.43 (m, 1H), 4.38 - 4.26 (m, 1H), 4.05 - 3.93 (m, 2H), 3.92 - 3.82 (m, 1H), 3.56 (dd, J = 4.4, 13.6 Hz, 1H), 3.46 (s, 3H), 3.14 - 3.04 (m, 1H), 2.69 (s, 3H), 2.56 - 2.54 (m, 1H), 2.15 - 2.07 (m, 1H), 2.05 - 1.98 (m, 1H), 1.92 - 1.83 (m, 1H), 1.74 - 1.65 (m, 1H). Synthesis of Compound 309 Exemplary Synthesis of 7-((3aR,7aS)-1-(5-chloropyridin-2-yl)octahydro-5H-pyrrolo[3,2- c]pyridin-5-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile The title compound was prepared in an analogous manner to 7-[(3aS,7aR)-5-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aR,7aS)-1-(5-chloro-2- pyridyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine. (white solid). LC/MS (ESI) m/z: 440.9 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 7.54 (d, J = 9.2 Hz, 1H), 6.51 (d, J = 9.2 Hz, 1H), 4.40 (d, J = 13.6 Hz, 1H), 4.31 - 4.19 (m, 2H), 3.93 (dd, J = 3.5, 13.6 Hz, 1H), 3.47 (s, 4H), 3.41 (d, J = 12 Hz, 2H), 2.68 (s, 3H), 2.57 (s, 1H), 2.28 - 2.21 (m, 1H), 2.08 - 1.93 (m, 2H), 1.84 - 1.72 (m, 1H). Synthesis of Compound 310 Exemplary Synthesis of 4-[(3aR,7aS)-1-(5-chloropyridin-2-yl)-octahydro-1H- pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile The title compound was prepared in an analogous manner to 4-[(3aS,7aR)-1-(5-chloro-2- pyridyl)-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-5-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile starting from (benzyl (3aR,7aS)-1-(5-chloro-2-pyridyl)- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-5-carboxylate. (yellow solid). LC/MS (ESI) m/z: 455.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.17 - 8.01 (m, 2H), 7.81 (d, J = 8.8 Hz, 1H), 7.55 (dd, J = 2.4, 9.2 Hz, 1H), 6.53 (d, J = 9.2 Hz, 1H), 4.35 - 4.19 (m, 2H), 4.12 (d, J = 13.2 Hz, 1H), 3.92 (dd, J = 4.2, 13.6 Hz, 1H), 3.53 (s, 3H), 3.51 - 3.40 (m, 2H), 3.39 - 3.34 (m, 1H), 2.61 - 2.57 (m, 1H), 2.30 - 2.21 (m, 1H), 2.14 - 2.05 (m, 2H), 1.95 - 1.83 (m, 1H). Synthesis of Compound 311 Exemplary Synthesis of 7-((3aS,7aR)-1-(5-chloropyridin-2-yl)octahydro-5H-pyrrolo[3,2- c]pyridin-5-yl)-2,4-dimethyl-5-oxo-4,5-dihydrothiazolo[5,4-b]pyridine-6-carbonitrile
The title compound was prepared in an analogous manner to 7-[(3aS,7aR)-5-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-2,4- dimethyl-5-oxo-thiazolo[5,4-b]pyridine-6-carbonitrile starting from (3aS,7aR)-1-(5-chloro-2- pyridyl)-2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine. (white solid). LC/MS (ESI) m/z: 441.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 2.4 Hz, 1H), 7.54 (dd, J = 2.4, 9.0 Hz, 1H), 6.52 (d, J = 9.2 Hz, 1H), 4.40 (d, J = 13.9 Hz, 1H), 4.29 - 4.20 (m, 2H), 3.93 (dd, J = 4.1, 13.6 Hz, 1H), 3.52 - 3.39 (m, 5H), 3.32 - 3.27 (m, 1H), 2.69 (s, 3H), 2.62 - 2.56 (m, 1H), 2.28 - 2.20 (m, 1H), 2.08 - 1.89 (m, 2H), 1.83 - 1.71 (m, 1H). Synthesis of Compound 312 Step 1: Preparation of tert-butyl 5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate To a solution of 2-bromo-5-(trifluoromethoxy)pyridine (2.67 g, 11.05 mmol, 1 eq) and tert- butyl 2,3,3a,4,5,6,7,7a-octahydropyrrolo[3,2-c]pyridine-1-carboxylate (2.50 g, 11.05 mmol, 1 eq) in dioxane (25 mL) was added 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H- imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (1.07 g, 1.10 mmol, 0.1 eq) and cesium carbonate (10.80 g, 33.14 mmol, 3 eq). The mixture was stirred at 100 °C for 3 h. To the reaction mixture was added water (30 mL) and extracted with ethyl acetate 60 mL (30 mL x 2). The combined organic layers were washed with saturated sodium chloride (30 mL x 2), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (silicon dioxide, petroleum ether/ethyl acetate=100/1 to 5/1) to give tert-butyl 5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (3.74 g, 9.65 mmol, 87% yield) as a yellow oil. LC/MS: (ESI) m/z: 388.1 [M+1] +. Step 2: Preparation of tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate and tert-butyl (3aS,7aR)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H- pyrrolo[3,2-c]pyridine-1-carboxylate Racemic tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro- 2H-pyrrolo[3,2-c]pyridine-1-carboxylate (1.9 g, 4.90 mmol, 1 eq) was purified by SFC (column: REGIS(S,S)WHELK-O1(250mm x 25mm,10um); mobile phase: [neutral-ethyl alcohol]; 15%-15%, 200min) to afford tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2- pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (870 mg, 2.25 mmol, 45% yield) as a green oil (SFC:Rt=1.061 min) and tert-butyl (3aS,7aR)-5-[5- (trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridine-1- carboxylate (830 mg, 2.14 mmol, 43% yield)) as a green oil (SFC: Rt = 1.209 min). The absolute stereochemical configuration was tentatively assigned. Step 3: Preparation of (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 1,2,3,3a,4,6,7,7a-octahydropyrrolo[3,2-c]pyridine To a solution of tert-butyl (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-3,3a,4,6,7,7a- hexahydro-2H-pyrrolo[3,2-c]pyridine-1-carboxylate (500 mg, 1.29 mmol, 1 eq) in dichloromethane (5 mL) was added trifluoroacetic acid (7.70 g, 67.53 mmol, 5.00 mL, 52.32 eq). The mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a- octahydropyrrolo[3,2-c]pyridine (517 mg, crude, trifluoroacetic acid) as a yellow solid. LC/MS: MS (ESI) m/z: 288.3 [M+1] +. Step 4: Preparation of 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo- 1,5-naphthyridine-3-carbonitrile (Compound 312) To a solution of (3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]-1,2,3,3a,4,6,7,7a- octahydropyrrolo[3,2-c]pyridine (517 mg, 1.29 mmol, 1 eq, trifluoroacetic acid) and 4,6- dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (327 mg, 1.29 mmol, 1 eq) in acetonitrile (5 mL) was added diisopropylethylamine (833 mg, 6.44 mmol, 1.12 mL, 5 eq). The mixture was stirred at 25 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 x 25mm x 10um; mobile phase: [water (formic acid)- acetonitrile]; 58%-88%, 58min) to give 4-[(3aR,7aS)-5-[5-(trifluoromethoxy)-2-pyridyl]- 3,3a,4,6,7,7a-hexahydro-2H-pyrrolo[3,2-c]pyridin-1-yl]-6-chloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (465.7 mg, 0.89 mmol, 69% yield, 96% purity) as a yellow solid. LC/MS: (ESI) m/z: 505.2507.3, 505.3 [M+1] +.1H NMR: (400 MHz, DMSO-d6) δ: 8.13 (d, J = 2.4 Hz, 1H), 8.02 (d, J = 9.2 Hz, 1H), 7.80 (d, J = 9.2 Hz, 1H), 7.58 (dd, J = 2.4, 9.2 Hz, 1H), 6.88 (d, J = 9.6 Hz, 1H), 5.54 - 5.42 (m, 1H), 4.46 - 4.34 (m, 1H), 3.99 - 3.89 (m, 2H), 3.84 - 3.75 (m, 1H), 3.63 (dd, J = 4.4, 13.6 Hz, 1H), 3.50 (s, 3H), 3.21 - 3.13 (m, 1H), 2.57 (d, J = 6.4 Hz, 1H), 2.24 - 2.15 (m, 1H), 2.08 - 1.99 (m, 1H), 1.96 - 1.86 (m, 1H), 1.81 - 1.71 (m, 1H). Synthesis of Compound 421 Step 1: Preparation of tert-butyl 4-[(5-chloro-2- pyridyl)methylene]piperidine-1-carboxylate A mixture of 2-bromo-5-chloro-pyridine (3 g, 15.59 mmol, 1 eq), tert-butyl 4-[(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)methylene]piperidine-1-carboxylate (6.05 g, 18.71 mmol, 1.2 eq), tetrakis[triphenylphosphine]palladium (1.80 g, 1.56 mmol, 0.1 eq), sodium carbonate (2 M, 15.59 mL, 2 eq) in dioxane (20 mL) was degassed and purged with nitrogen 3 times, and then the mixture was stirred at 80 °C for 12 h under nitrogen atmosphere. The reaction mixture was added slowly to water (50ml), extracted with ethyl acetate (50 mL*3), the combined organic layers were dried over anhydrous sodium sulphate, filtered and concentrated. The resultant residue was purified by silica gel chromatography (Petroleum ether/Ethyl acetate=100/1 to 1:1) to give tert-butyl 4-[(5-chloro-2- pyridyl)methylene]piperidine-1-carboxylate (4.5 g, 14.57 mmol, 94 % yield) as yellow solid. LC/MS (ESI) m/z: 253.0[M-55]+ Step 2: Preparation of tert-butyl 2-(5-chloro-2-pyridyl)-1-oxa-6-azaspiro[2.5]octane- 6-carboxylate To a mixture of 5-methoxy-2-vinyl-pyridine tert-butyl 4-[(5-chloro-2- pyridyl)methylene]piperidine-1-carboxylate (4 g, 12.95 mmol, 1 eq) in dioxane (40 mL) and water (40 mL) was added a solution of N-bromosuccinimide (2.77 g, 15.54 mmol, 1.2 eq) in dioxane (40 mL) and water (40 mL) dropwise and stirred at 20 °C for 2 h under a nitrogen atmosphere. The reaction mixture was cooled to 0 °C and sodium hydroxide (4 M, 6.48 mL, 2 eq) was added dropwise and purged with nitrogen 3 times. The ice-bath was removed and stirred at 20 °C for 12 h. The reaction mixture was added slowly to water (100 mL), then extracted with ethyl acetate (100 mL*3). The combined organic layers were dried over anhydrous sodium sulphate, filtered and concentrated to give tert-butyl 2-(5-chloro-2- pyridyl)-1-oxa-6-azaspiro[2.5]octane-6-carboxylate (3.29 g, 10.13 mmol, 78% yield) as colorless oil. LC/MS (ESI) m/z: 268.9 [M-55]+. Step 3: Preparation of tert-butyl 4-[(1R)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy- piperidine-1-carboxylate and tert-butyl 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]-4- hydroxy-piperidine-1-carboxylate To a solution of copper iodide (2.35 g, 12.32 mmol, 2.5 eq) in diethyl ether (16 mL) was added methyl magnesium chloride (3 M, 8.21 mL, 5 eq) and then stirred at -78°C for 1 h under nitrogen atmosphere. Then the solution of tert-butyl 2-(5-chloro-2-pyridyl)-1-oxa-6- azaspiro[2.5]octane-6-carboxylate (1.6 g, 4.93 mmol, 1 eq) in diethyl ether (16 mL) was added dropwise to the mixture. The mixture was stirred at 0 °C for 3 h under nitrogen atmosphere. The mixture was diluted with saturated ammonium chloride (50 mL), extracted with ethyl acetate (3 × 50 mL), washed with brine (50mL), and dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by prep-HPLC (column: UniSil 10-120 C1850x250mm; mobile phase: [water (formic acid)-acetonitrile]: 33%-63%, 22 min), which was further purified by prep-SFC (column: DAICEL CHIRALPAK IG (250mm x 30mm, 10um); mobile phase: 25% [0.1% ammonium hydroxide - ethanol] in supercritical carbon dioxide, 25%-25%, 294 min) to give tert-butyl 4-[(1R)-1-(5-chloro-2-pyridyl)ethyl]- 4-hydroxy-piperidine-1-carboxylate (325 mg, 0.954 mmol, 19% yield) as a colorless oil and tert-butyl 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy-piperidine-1-carboxylate (384 mg, 1.13 mmol, 23% yield) as a colorless oil. tert-butyl 4-(1-(5-chloropyridin-2-yl)ethyl)-4- hydroxypiperidine-1-carboxylate LC/MS (ESI) m/z: 284.8 [M-55]+. SFC Rt: 1.328 min for tert-butyl 4-[(1R)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy-piperidine-1-carboxylate. SFC Rt: 1.432 min for tert-butyl 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy-piperidine-1- carboxylate. The absolute stereochemical configuration was tentatively assigned. Step 4: Preparation of 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]piperidin-4-ol A mixture of tert-butyl 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy-piperidine-1- carboxylate (280 mg, 0.821 mmol, 1 eq), trifluoroacetic acid (4.68 g, 41.07 mmol, 3.04 mL, 50 eq) in dichloromethane (4 mL) was degassed and purged with nitrogen 3 times, then the mixture was stirred at 25°C for 0.5 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]piperidin-4-ol (290 mg, 0.817 mmol, 99% yield, trifluoroacetic acid) was obtained as colorless oil. LC/MS (ESI) m/z: 241.0 [M+1]+ Step 5: Preparation of ethyl 3-amino-6-chloro-pyridine-2-carboxylate To a solution of 3-amino-6-chloro-pyridine-2-carboxylic acid (5 g, 28.97 mmol, 1 eq) in ethanol (80 mL) was added sulfuric acid (5.68 g, 57.95 mmol, 3.09 mL, 2 eq) at 25°C. The mixture was stirred at 90°C for 12 h. The reaction mixture was adjusted pH to 7- 8 and was diluted with water (100 mL) was extracted with ethyl acetate (50 mL × 2). The combined organic layers were dried over anhydrous sodium sulfate and were concentrated under reduced pressure to give the product. The crude product was used into the next step without further purification. Compound ethyl 3-amino-6-chloro-pyridine-2-carboxylate (6 g, crude) was obtained as a yellow oil, confirmed by HNMR.1H NMR: (400 MHz, CDCl3) δ: 7.23 (d, J = 8.8 Hz, 1H), 7.05 (d, J = 8.8 Hz, 1H), 4.5 – 4.40 (m, 2H), 3.52 - 3.81 (m, 2H), 1.44 (t, J = 7.2 Hz, 3H) LC/MS:(ESI) m/z: 201.1 [M+1]+ Step 6: Preparation of ethyl 6-chloro-3-[(2-cyanoacetyl)amino]pyridine-2- carboxylate
Figure imgf000577_0001
To a solution of ethyl 3-amino-6-chloro-pyridine-2-carboxylate (6 g, 29.91 mmol, 1 eq) and 2- cyanoacetic acid (5.09 g, 59.81 mmol, 2 eq) in dimethylformamide (50 mL) was added triethylamine (9.08 g, 89.72 mmol, 12.49 mL, 3 eq) and propylphosphonic anhydride (38.06 g, 59.81 mmol, 35.57 mL, 50% purity, 2 eq) at 25°C. The mixture was stirred at 25°C for 1 h . The reaction mixture was diluted with water (200 mL) and extracted with ethyl acetate (50 mL × 3). The organic layer was dried over sodium sulfate, filtered and filtrate was concentrated in vacuum. The crude product was triturated with (petroleum ether: ethyl acetate=3:1) at 25oC for 5 min. Ethyl 6-chloro-3-[(2-cyanoacetyl)amino]pyridine-2-carboxylate (6 g, 22.42 mmol, 74% yield) was obtained as a yellow solid, confirmed by HNMR. 1H NMR: (400 MHz, CDCl3) δ: 11.70 (s, 1H) , 9.03 (d, J = 8.8 Hz, 1 H), 7.54 (d, J = 8.8 Hz, 1H), 4.44 - 4.64 (m, 2 H), 3.64 (s, 2H), 1.48 (t, J = 7.2 Hz, 3H) LC/MS: (ESI) m/z: 268.1 [M+1]+. Step 7: Preparation of 6-chloro-4-hydroxy-2-oxo-1H-1,5-naphthyridine-3-carbonitrile To a solution of ethyl 6-chloro-3-[(2-cyanoacetyl)amino]pyridine-2-carboxylate (5.9 g, 22.04 mmol, 1 eq) in ethanol (100 mL) was added 1,8-diazabicyclo[5.4.0]undec-7-ene (5.03 g, 33.06 mmol, 4.98 mL, 1.5 eq) at 25°C. The mixture was degassed and purged with nitrogen 3 times, then the mixture was stirred at 25°C for 2 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to provide a residue. The mixture was quenched by 1 M hydrochloric acid adjust pH = 2, filtered and filtrate cake was collected. Compound 6-chloro-4-hydroxy-2-oxo-1H-1,5-naphthyridine-3-carbonitrile (3 g, 13.54 mmol, 61% yield) was obtained as a yellow solid, confirmed by HNMR.1H NMR: (400MHz, DMSO-d6) δ: 11.94 (s, 1H), 7.76 – 7.71 (m, 2H) LC/MS (ESI) m/z: 220.0 [M+1]+. Step 8: Preparation of 4,6-dichloro-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3- carbonitrile To a mixture of 6-chloro-4-hydroxy-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (3 g, 12.73 mmol, 1 eq) and diisopropylethylamine (9.87 g, 76.39 mmol, 13.31 mL, 6 eq) in acetonitrile (40 mL) was added phosphoryl chloride (9.76 g, 63.66 mmol, 5.92 mL, 5 eq) and benzyl(triethyl)ammonium chloride (5.80 g, 25.46 mmol, 2 eq) in one portion at 25 °C under nitrogen. The mixture was stirred at 25 °C for 3 h. The mixture was concentrated. The resultant residue was poured into water (200ml) and saturated sodium hydrogen carbonate solution (200 mL). The aqueous portion was extracted with dichloromethane (200 mL× 3). The combined organic extracts were washed with brine (200mL), dried with anhydrous sodium sulfate, filtered, and concentrated to give a residue. The crude product was triturated with dichloromethane : methanol = 30:1 at 25 oC for 5 min to give the provide 4,6-dichloro-1- methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (1 g, 3.94 mmol, 30% yield) as a yellow solid. 1H NMR: (400 MHz, DMSO-d6) δ: 8.28 (d, J = 9.2 Hz, 1H), 8.00 (d, J = 8.8 Hz, 1H), 3.65 (s, 3H) LC/MS: (ESI) m/z: 253.9 [M+1]+. Step 9: Preparation of 6-chloro-4-[4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (Compound 421)
A mixture of 4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]piperidin-4-ol (290 mg, 0.817 mmol, 1 eq, trifluoroacetic acid), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (208 mg, 0.817 mmol, 1 eq), N,N-diisopropylethylamine (528 mg, 4.09 mmol, 0.712 mL, 5 eq) in acetonitrile (4 mL) was degassed and purged with nitrogen 3 times, then the mixture was stirred at 40 °C for 12 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to provide a residue. The residue was dispersed with dimethylformamide (2mL), then was filtered and the filter cake washed with a small amount of acetonitrile to afford 6-chloro-4-[4-[(1S)-1-(5-chloro-2-pyridyl)ethyl]-4-hydroxy-1-piperidyl]-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (258 mg, 0.562 mmol, 69% yield, 100% purity) as a yellow solid. 1H NMR: (400 MHz, DMSO-d6). δ: 8.54 (d, J = 2.0 Hz, 1H), 8.03 (d, J = 8.8 Hz, 1H), 7.86 (dd, J = 2.4, 8.4 Hz, 1H), 7.77 (d, J = 8.8 Hz, 1H), 7.39 (d, J = 8.4 Hz, 1H), 4.79 (s, 1H), 3.97 (s, 2H), 3.67 - 3.59 (m, 2H), 3.50 (s, 3H), 3.00 (q, J = 7.2 Hz, 1H), 1.85 (dt, J = 4.4, 12.8 Hz, 1H), 1.79 - 1.74 (m, 2H), 1.37 (d, J = 13.6 Hz, 1H), 1.28 (d, J = 6.8 Hz, 3H) LC/MS: (ESI) m/z: 460.4, 458.2[M+1]+. Synthesis of Compound 397 Step 1: Preparation of (5-chloro-2-pyridyl)-phenyl-methanol To a solution of copper iodide (10 g, 52.98 mmol, 1.5 eq) in diethyl ether (50 mL) was added bromo(phenyl)magnesium (3 M, 17.66 mL, 1.5 eq) at -78 °C under nitrogen atmosphere and stirred for 0.5 h. Then 5-chloropyridine-2-carbaldehyde (5 g, 35.32 mmol, 1 eq) in diethyl ether (50 mL) was added dropwise at -78°C under nitrogen atmosphere. Then the mixture was stirred at 0°C for another 1h. The mixture was diluted with saturated ammonium chloride (100 mL), extracted with ethyl acetate (3 × 100 mL), washed with brine (3 ×100 mL), and dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1 to 1:1) to give (5-chloro-2- pyridyl)-phenyl-methanol (7.56 g, 31.66 mmol, 90% yield, 92% purity) as a yellow oil.LC/MS (ESI) m/z: 202.2 [M-17]+ Step 2: Preparation of 2-[bromo(phenyl)methyl]-5-chloro-pyridine A mixture of (5-chloro-2-pyridyl)-phenyl-methanol (500 mg, 2.28 mmol, 1 eq) in thionyl bromide (5 mL) was degassed and purged with nitrogen 3 times, then the mixture was stirred at 25 °C for 12 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure. To the resultant residue was added dichloromethane (10 mL) and was washed with saturated sodium bicarbonate (10 mL x 2). The mixture was concentrated under reduced pressure to give 2-[bromo(phenyl)methyl]-5-chloro-pyridine (460 mg, crude) as a purple oil. LC/MS: (ESI) m/z: 283.8 [M+1]+ Step 3: Preparation of 4-[(5-chloro-2-pyridyl)-phenyl-methyl]-4-hydroxy-piperidine-1- carboxylate To a solution of 2-[bromo(phenyl)methyl]-5-chloro-pyridine (460 mg, 1.63 mmol, 1 eq) and tert-butyl 4-oxopiperidine-1-carboxylate (324 mg, 1.63 mmol, 1 eq) in tetrahydrofuran (4 mL) was added n-butyllithium (2.5 M, 0.98 mL, 1.5 eq) under nitrogen atmosphere at -78°C. Then the mixture was stirred at -78°C for 1 h. The mixture was diluted with saturated ammonium chloride (20 mL), extracted with ethyl acetate (3 × 30 mL), washed with brine (3 × 30 mL). The combined organic portions were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1 to 10:1) to give tert-butyl 4-[(5-chloro-2-pyridyl)-phenyl-methyl]-4-hydroxy- piperidine-1-carboxylate (510 mg, 1.20 mmol, 74% yield, 95% purity) as a colorless oil.LC/MS: (ESI) m/z: 403.1 [M+1]+ Step 4: Preparation of tert-butyl 4-[(S)-(5-chloro-2-pyridyl)-phenyl-methyl]-4- hydroxy-piperidine-1-carboxylate and tert-butyl 4-[(R)-(5-chloro-2-pyridyl)-phenyl- methyl]-4-hydroxy-piperidine-1-carboxylate The product was purified by SFC (column: DAICEL CHIRALPAK AD (250 mm x 30 mm, 10um); mobile phase: [0.1% ammonium hydroxide ethanol], 50min) to give tert-butyl 4-[(S)- (5-chloro-2-pyridyl)-phenyl-methyl]-4-hydroxy-piperidine-1-carboxylate (250 mg, 0.62 mmol, 52% yield) as a colorless oil (LC/MS: (ESI) m/z: 403.1 [M+1]+ SFC: Rt: 1.165 min) and tert-butyl 4-[(R)-(5-chloro-2-pyridyl)-phenyl-methyl]-4-hydroxy-piperidine-1- carboxylate (150 mg, 0.37 mmol, 31% yield) as a colorless oil (LC/MS: (ESI) m/z: 403.1 [M+1]+ SFC: Rt: 1.342 min.) The absolute stereochemical configuration was tentatively assigned. Step 5: Preparation of 4-[(R)-(5-chloro-2-pyridyl)-phenyl-methyl]piperidin-4-ol A mixture of tert-butyl 4-[(R)-(5-chloro-2-pyridyl)-phenyl-methyl]-4-hydroxy-piperidine-1- carboxylate (100 mg, 0.25 mmol, 1 eq) and trifluoroacetic acid (743 mg, 0.006 mmol, 0.48 mL, 26.26 eq) in dichloromethane (0.5 mL) was degassed and purged with nitrogen 3 times, then the mixture was stirred at 25 °C for 0.5 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to give 4-[(R)-(5-chloro-2-pyridyl)-phenyl- methyl]piperidin-4-ol (103 mg, 0.25 mmol, 99% yield, trifluoroacetic acid) as a brown oil. LC/MS: (ESI) m/z: 303.0 [M+1]+ Step 6: Preparation of 6-chloro-4-[4-[(R)-(5-chloro-2-pyridyl)-phenyl-methyl]-4- hydroxy-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile Compound 433 To a solution of 4-[(R)-(5-chloro-2-pyridyl)-phenyl-methyl]piperidin-4-ol (103 mg, 0.25 mmol, 1 eq, trifluoroacetic acid) in acetonitrile (1 mL) was added N,N-diisopropylethylamine (160 mg, 1.24 mmol, 0.22 mL, 5 eq) followed by 4,6-dichloro-1-methyl-2-oxo-1,5- naphthyridine-3-carbonitrile (63 mg, 0.25 mmol, 1 eq). The mixture was stirred at 40 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 x25 mm x 10 um; mobile phase: [water (formic acid )-acetonitrile]; 10min) to give 6-chloro-4-[4-[(R)-(5-chloro-2- pyridyl)-phenyl-methyl]-4-hydroxy-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3- carbonitrile (73.0 mg, 0.14 mmol, 57% yield, 99% purity) as a yellow solid.LC/MS: (ESI) m/z: 522.3, 520.3 [M+1]+1H NMR: (400MHz, DMSO-d6) δ: 8.64 (d, J = 2.4 Hz, 1H), 8.04 (d, J = 9.2 Hz, 1H), 7.92 - 7.87 (m, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.57 (dd, J = 3.6, 8.0 Hz, 3H), 7.31 - 7.26 (m, 2H), 7.23 - 7.18 (m, 1H), 5.57 (s, 1H), 4.29 (s, 1H), 4.02 - 3.94 (m, 2H), 3.73 - 3.59 (m, 2H), 3.51 (s, 3H), 1.97 - 1.89 (m, 1H), 1.86 - 1.78 (m, 1H), 1.58 - 1.46 (m, 2H). Synthesis of Compound 416 Exemplary Synthesis of 6-chloro-4-{4-[(5-chloropyridin-2-yl)methyl]-4- methoxypiperidin-1-yl}-1-methyl-2-oxo-1,2-dihydro-1,5-naphthyridine-3-carbonitrile (
Figure imgf000583_0001
Step 1: Preparation of tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4-methoxy-piperidine-1- carboxylate
Figure imgf000583_0002
To a mixture of tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4-hydroxy-piperidine-1-carboxylate (200 mg, 0.61 mmol, 1 eq) in tetrahydrofuran (2 mL) was added sodium hydride (122 mg, 3.06 mmol, 60% purity, 5 eq) then was degassed and purged with nitrogen 3 times, then stirred at 0°C for 0.5 h. To the mixture was added iodomethane (261 mg, 1.84 mmol, 0.11 mL, 3 eq) and stirred at 0°C for 0.5 h under nitrogen. The mixture was diluted with saturated ammonium chloride (5 mL), extracted with ethyl acetate (3 x 5 mL), washed with brine (3 × 5 mL), and dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 x 25mm x 10um; mobile phase: [water(formic acid)- acetonitrile]; 50%-80%, 9 min) to give tert-butyl 4-[(5-chloro-2- pyridyl)methyl]-4-methoxy-piperidine-1-carboxylate (40 mg, 0.11 mmol, 18% yield, 95% purity) as a brown oil. LCMS: (ESI) m/z: 341.1 [M+1] + Step 2: Preparation of tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4-methoxy-piperidine-1- carboxylate
Figure imgf000584_0001
A mixture of tert-butyl 4-[(5-chloro-2-pyridyl)methyl]-4-methoxy-piperidine-1-carboxylate (40 mg, 0.12 mmol, 1 eq), and trifluoroacetic acid (274 mg, 2.40 mmol, 0.18 mL, 20 eq) in dichloromethane (0.5 mL) was degassed and purged with nitrogen 3 times, then the mixture was stirred at 25°C for 0.5 h under nitrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give 5-chloro-2-[(4-methoxy-4- piperidyl)methyl]pyridine (42 mg, crude, trifluoroacetic acid) as a brown oil. LCMS: (ESI) m/z: 241.0 [M+1] + Step 3: Preparation of 6-chloro-4-[4-[(5-chloro-2-pyridyl)methyl]-4-methoxy-1- piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile
Figure imgf000584_0002
To a solution of 5-chloro-2-[(4-methoxy-4-piperidyl)methyl]pyridine (42 mg, 0.12 mmol, 1 eq, trifluoroacetic acid) in acetonitrile (1 mL) was added N,N-diisopropylethylamine (76 mg, 0.60 nmol, 0.10 mL, 5 eq) and then 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (30 mg, 0.12 mmol, 1 eq) was added. The mixture was stirred at 40°C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The resultant residue was purified by prep-HPLC (column: Phenomenex Luna C18150 x 25mm x 10um; mobile phase: [water (formic acid) in acetonitrile]; 45%-75%, 9 min) to afford 6-chloro-4-[4-[(5-chloro-2- pyridyl)methyl]-4-methoxy-1-piperidyl]-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (35.3 mg, 0.074 mmol, 62% yield, 96% purity) as a yellow solid. LCMS: MS (ESI) m/z: 458.2 [M+1] + 1H NMR: (400MHz, DMSO-d6) δ: 8.54 (d, J = 2.8 Hz, 1H), 8.04 (d, J = 9.2 Hz, 1H), 7.85 (dd, J = 2.4, 8.4 Hz, 1H), 7.78 (d, J = 9.2 Hz, 1H), 7.34 (d, J = 8.4 Hz, 1H), 3.96 (d, J = 12.8 Hz, 2H), 3.59-3.53 (m, 2H), 3.53 - 3.49 (m, 3H), 3.33 (s, 3H), 3.04 (s, 2H), 1.92 - 1.80 (m, 4H) Synthesis of Compound 429 Exemplary Synthesis of 6-chloro-1-methyl-2-oxo-4-{3H-spiro[furo[3,2-b]pyridine-2,4'- piperidin]-1'-yl}-1,2-dihydro-1,5-naphthyridine-3-carbonitrile Step 1: Preparation of tert-butyl 4-[(3-bromo-2-pyridyl)methyl]-4-hydroxy-piperidine-1- carboxylate To a solution of 3-bromo-2-methyl-pyridine (2.7 g, 15.70 mmol, 1 eq) in tetrahydrofuran (10 mL) was added lithium diisopropylamide (2 M, 11.77 mL, 1.5 eq) in -78°C. The reaction mixture was stirred at -78°C for 1 h. Then, tert-butyl 4-oxopiperidine-1-carboxylate (3.13 g, 15.70 mmol, 1 eq) in tetrahydrofuran (10 mL) was added dropwise at -78°C and warmed to 0 °C, and stirred for 30 min at 0 °C. The reaction mixture was diluted with ammonium chloride (20 mL) and water (20 mL). Then the mixture was extracted with ethyl acetate (30 mL x 3). The combined organic layers were washed by brine and dried over sodium sulfate. Then the mixture was concentrated under reduced pressure to afford a residue. The resultant residue was purified by prep-HPLC (column: Phenomenex Luna C18 (250 x 70mm, 10 um); mobile phase: [water (formic acid) in acetonitrile]: 20%-55%, 23 min) to give the product tert-butyl 4-[(3-bromo-2-pyridyl)methyl]-4-hydroxy-piperidine-1-carboxylate (3.1 g, 8.35 mmol, 53% yield) as a yellow oil.LCMS: (ESI) m/z: 315 [M-55] +1H NMR: (400 MHz, CDCl3)δ: 8.51 (d, J = 3.2 Hz, 1H), 8.14 - 7.97 (m, 1H), 7.21 (dd, J = 4.8, 8.0 Hz, 1H), 4.95 (s, 1H), 3.66 - 3.63 (m, 2H), 3.09 (s, 2H), 3.04 (s, 2H), 1.50 (s, 2H), 1.41 - 1.38 (m 2H), 1.35 (s, 9H) Step 2: Preparation of tert-butyl spiro[3H-furo[3,2-b]pyridine-2,4'-piperidine]-1'- carboxylate. To a solution of tert-butyl 4-[(3-bromo-2-pyridyl)methyl]-4-hydroxy-piperidine-1- carboxylate (300 mg, 0.80 mmol, 1 eq) in N-methyl pyrrolidone (3 mL) was added bis[(Z)-1- methyl-3-oxo-but-1-enoxy]copper (423 mg, 1.62 mmol, 2 eq). The mixture was stirred at 100 °C for 16 hr. The reaction mixture was diluted with water 20 mL and extracted with ethyl acetate (20 mL x 2). The combined organic layers were washed with brine (40 mL x 2), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: UniSil 10-120 C1850x250mm; mobile phase: [water (formic acid) in acetonitrile]: 12%-43%, 23 min) to give the product tert-butyl spiro[3H-furo[3,2-b]pyridine-2,4'-piperidine]-1'-carboxylate (220 mg, 0.75 mmol, 93% yield) as a yellow solid. LCMS: (ESI) m/z: 291.2 [M+1]+ Step 3: Preparation of spiro[3H-furo[3,2-b]pyridine-2,4'-piperidine] To a solution of tert-butyl spiro[3H-furo[3,2-b]pyridine-2,4'-piperidine]-1'-carboxylate (130 mg, 0.44 mmol, 1 eq) in dichloromethane (0.5 mL) was added trifluoroacetic acid (1.00 g, 8.78 mmol, 19.61 eq). The mixture was stirred at 25 °C for 1 hr. The reaction mixture was concentrated under reduced pressure to remove solvent to give the product spiro[3H-furo[3,2- b]pyridine-2,4'-piperidine] (136 mg, 0.44 mmol, crude, trifluoroacetic acid) as a yellow oil. Step 4: Preparation of spiro[3H-furo[3,2-b]pyridine-2,4'-piperidine] A mixture of spiro[3H-furo[3,2-b]pyridine-2,4'-piperidine] (136 mg, 0.44 mmol, 1 eq, trifluoroacetic acid), 4,6-dichloro-1-methyl-2-oxo-1,5-naphthyridine-3-carbonitrile (114 mg, 0.44 mmol, 1 eq), and N-ethyl-N-isopropyl-propan-2-amine (288 mg, 2.23 mmol, 5 eq) in acetonitrile (1 mL) was stirred at 40 °C for 12 h. The reaction mixture was concentrated under reduced pressure to remove solvent. The mixture was triturated with acetonitrile (5 mL) to give the product 6-chloro-1-methyl-2-oxo-4-spiro[3H-furo[3,2-b]pyridine-2,4'- piperidine]-1'-yl-1,5-naphthyridine-3-carbonitrile (155 mg, 0.37 mmol, 83% yield, 98% purity) as a yellow solid. LCMS:(ESI) m/z: 408.0[M+1]+ 1H NMR: (400 MHz, CDCl3) δ: 8.09 (d, J = 9.2 Hz, 1H), 8.00 (d, J = 4.0 Hz, 1H), 7.82 (d, J = 8.8 Hz, 1H), 7.23 - 7.17 (m, 1H), 7.15 - 7.06 (m, 1H), 3.93 (d, J = 3.2 Hz, 4H), 3.61 - 3.60 (m, 1H), 3.54 (s, 3H), 3.25 (s, 2H), 2.12 (t, J = 5.2 Hz, 4H). Human DGKα kinase activity inhibition assay Human DGKα inhibitory activity of compounds of the present invention was quantified employing the human DGKα kinase activity assay as described in the following paragraphs. In essence, the enzyme activity was measured by quantification of the adenosine-di- phosphate (ADP) generated as a co-product of the enzyme reaction via the “ADP-Glo™ Kinase Assay” kit from the company Promega. This detection system works as follows: In a first step the ATP not consumed in the kinase reaction is quantitatively converted to cAMP employing an adenylate cyclase (“ADP-Glo-reagent”), then the adenylate cyclase is stopped, and the ADP generated in the kinase reaction converted to ATP, which subsequently generates in a luciferase-based reaction a glow-luminescence signal (“Kinase Detection Reagent”). N-terminal His-tagged, recombinant full-length human DGKα (expressed in baculovirus infected insect cells, purified using anti-His pulldown and size exclusion chromatography as described below, DGKα _hu_1) was used as enzyme. As substrate for the kinase 1,2- dioleoyl- sn-glycerol, reconstituted in octyl-β-D-glucopyranoside micelles, was used. For the preparation of the micelles, 1 volume of a 16.1 mM solution of 1 ,2-dioleoyl-sn-glycerol (Avanti, Cat. # 08001- 25G) in chloroform was slowly evaporated using a nitrogen stream. Subsequently, 22.55 volumes of a 510 mM solution of octyl-β-D-glucopyranoside (Sigma- Aldrich, Cat. # 08001 -10G) in 50 mM MOPS buffer (pH 7.4) were added, and the mixture was sonicated in an ultrasonic bath for 20 s. Then 35 volumes of 50 mM MOPS buffer (pH 7.4) were added to yield a solution of 0.28 mM 1 ,2 dioleoyl-sn-glycerol and 200 mM octyl- β-D-glucopyranoside, which was aliquoted, flash-frozen in liquid nitrogen, and stored at - 20Ό until use. For each experiment, a fresh aliquot was quickly thawed and diluted 24-fold with aqueous assay buffer (described below) containing 95.7 mM adenosine triphosphate (Promega) to yield a 1.67-fold concentrated substrate solution. For the assay 1000 nL of a 100-fold concentrated solution of the test compound in dimethyl sulfoxide (DMSO, Sigma) was pipetted into a white low-volume 384-well microtiter plate (both Greiner Bio-One, Frickenhausen, Germany). Subsequently, 2 µL.I of a solution of human DGKα in aqueous assay buffer [50 mM (3-(N-morpholino)propanesulfonic acid (MOPS, pH 7.4, Sigma-Aldrich), 1 mM dithiothreitol (DTT, Sigma-Aldrich), 100 mM NaCI (Sigma-Aldrich), 10 mM MgCh (Sigma-Aldrich), 0.1 % (w/v) bovine gamma globulin (BGG, Sigma-Aldrich), 1 mM CaCh (Sigma-Aldrich)] were added to the wells, and the mixture was incubated for 10 min at 22 °C to allow pre-binding of the test compounds to the enzyme. The reaction was initiated by the addition of 2 µL.L of substrate solution [preparation described above; 11.7 mM 1 ,2-dioleoyl-sn-glycerol (=> final cone in the 5 mL assay volume is 7 mM), 8.33 mM octyl-β-D-glucopyranoside (=> final conc. in 5 mL assay volume is 5 mM), and 91.67 mM adenosine triphosphate (=> final conc. in 5 mL assay volume is 55 mM) in assay buffer] and the resulting mixture was incubated for a reaction time of 60 min at 22 °C. The concentration of DGK a was adjusted depending on the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, a typical concentration is about 0.1 nM. The reaction was stopped by the addition of 5µL. of “ADP- Glo-reagent” (1 to1.5 diluted with water) and the resulting mixture was incubated at 22 for 40 minutes to convert the ATP not consumed in the kinase reaction completely to cAMP. Subsequently 10µL. of the “kinase detection reagent” (1.2-fold more concentrated than recommended by the producer) were added, the resulting mixture was incubated at 22 for 1 h and then the luminescence measured with a suitable measurement instrument (e.g., Viewlux™ from Perkin-Elmer). The amount of emitted light was taken as a measure for the amount of ADP generated and thereby for the activity of the DGKα. The data were normalized (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 11 different concentrations in the range of 20 mM to 0.07 nM (20 mM, 5.7 mM, 1.6 mM, 0.47 mM, 0.13 mM, 38 hM, 11 hM, 3.1 hM, 0.9 hM, 0.25 nM and 0.07 nM, the dilution series prepared separately before the assay on the level of the 100-fold concentrated solutions in DMSO by serial dilutions, exact concentrations may vary depending pipettors used) in duplicate values for each concentration and IC5o values were calculated using Genedata Screener™ software. Table 5. IC50 values of certain compounds of the invention in in vitro human DGKα inhibition activity assays.
Figure imgf000589_0001
Figure imgf000590_0001
Figure imgf000591_0001
Figure imgf000592_0001
Figure imgf000593_0001
Human DGKα human kinase degradation assay in Jurkat cells Human DGKα degradation activity of compounds of the present invention was quantified employing treatment of the Jurkat (human T cell leukemia) cell line for a period of 24 hours. Cells were read by flow cytometry to determine the amount of protein loss intracellularly. Jurkat cells were plated at a density of 1x106 cells/mL in 200µL. of complete cell growth media in a flat bottom 96 well tissue culture treated microtiter plate. Compounds were diluted in 100% DMSO such that the treatment range at the highest dose was 1µM continuing for a 10-point dose titration ending with the lowest dose at 0.0001µM. Compound treatments were added to the Jurkat cells as a 0.1% DMSO solution and allowed to incubate in a 37°C incubator containing 5% CO2 for 24 hours. The following day treated Jurkat cells were fixed with 2% paraformaldehyde for 15 minutes at 22°C. Cells were spun down at 300 X G for 5 minutes, supernatant decanted and washed in PBS containing 4% FBS. Cells were spun down at 300 X G for 5 minutes, supernatant decanted and permeabilized in a saponin solution for 15 minutes at 22°C. Following permeabilization, cells were spun down at 300 X G for 5 minutes and the supernatant decanted. Human DGKα-alexa fluor 488 conjugated antibody (Novus biologicals catalogue #NBP2-70547AF488) as well as a matched isotype control were diluted in a saponin solution at 1:500 and added to the Jurkat cells and allowed to incubate at 22°C for 60 minutes protected from light. Following antibody incubation, cells were washed 3 times in PBS containing 4% FBS and resuspended in 200µL. of PBS containing 4% FBS. Samples were subsequently read on a BD Symphony A3 flow cytometer. The data were normalized (staining condition with isotype control = 0% degradation, DMSO treated cells stained with DGKα conjugated to alexa fluor 488 = 100% degradation). DC50 values as well as Dmax was calculated using Genedata ScreenerTM software. Human IL-2/Interferon-γ/TNF-α cytokine secretion in normal human PBMC’s Human DGKα protein loss/inhibition downstream functional effect of compounds of the present invention was quantified employing the treatment of normal human PBMC’s in the presence of a sub-optimal T cell receptor stimulation and subsequently looking at IL- 2/Interferon γ/TNF-α cytokine secretion using Meso scale platform technology. Normal human PBMC’s were plated on a 96 well tissue culture treated microtiter plate coated with 40ng/mL of human CD3 (clone OKT3) at a density of 1x106 cells/mL. in 200µL. of complete cell growth media containing 1µg/mL. of CD28 (clone CD28.2). Compounds were diluted in 100% DMSO such that the highest treatment concentration was 1µM. Compound treatments were titrated for a 7-point dose curve ending with the lowest dose of 0.0005µM. Compounds were added to the PBMC’s as a 0.1% DMSO solution and allowed to incubate in a 37°C incubator containing 5% CO2 for 24 hours. The following day, PBMC’s containing compound treatment were spun down at 500 X G for 10 minutes. Supernatant was carefully transferred to a 96-well u-bottom microtiter plate and placed on ice. A calibrator standard curve was diluted according to the manufacturers’ instructions and added to columns 1 and 2 of each MSD plate used. PBMC supernatants were diluted 1:8 in buffer and added to a pre-coated MSD plate and allowed to shake for 60 minutes at 22°C. Detection reagent was diluted according to manufacturers’ instructions and added to the MSD plate containing PBMC supernatant samples and allowed to shake for an additional 60 minutes at 22°C. Following the final incubation with detection reagent, the MSD plate was washed 3X with PBS-Tween wash buffer.150µL of MSD read buffer was added to each well and read on the MSD reader. The data were quantified according to the calibrator standard curve and normalized (sub- optimal simulation without compound = 100% cytokine secretion). Enhancement of cytokine secretion above the sub-optimal stimulation without compound is reported as an EC50 using Genedata ScreenerTM software. Table 6. DC50 values of certain compounds of the invention in in vitro Jurkat degradation assay.
Figure imgf000595_0001
Figure imgf000596_0001
Figure imgf000597_0001
HiBiT DGKα human kinase degradation assay in Jurkat cells HiBiT-DGKAKI Jurkat CPM Cells (Promega), and cell media consisted of RPMI 1640 with 10% FBS and 1% pen/strep. The cells were counted, measured for viability, and 35 µL of the cell solution was seeded into 384 well flat bottom plates at a ratio of 20,000 cells per 35 µL, after which the plates were incubated for ~24 hours at 37° C and 5% CO2. Compounds were ordered from Evotec in a 384 well plate at a 1 mM top concentration in DMSO and serially diluted 1:3 in DMSO for an 11-point dose response. Using the Bravo instrument, 1µL of compound solution or DMSO was transferred to 125 µL of media. Each well was treated with 5 µL of compound plus media mixture or DMSO and subsequently incubated for at least 6-24 hours at 37° C and 5% CO2. After incubation, 30 µL of either NanoGlo media was added to each cell, and the plates read on the Synergy Neo2 machine. Table 7. DC50 values of certain compounds of the invention in HiBit Jurkat degradation assay.
Figure imgf000597_0002
Figure imgf000598_0001
Figure imgf000599_0001
Figure imgf000600_0001
Figure imgf000601_0001

Claims

CLAIMS 1. A compound having the structure of Formula (A): (A) or a pharmaceutically acceptable salt thereof, wherein: Ring A is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaromatic ring fused to ring B; Ring C is a heterocyclic ring, which may be further substituted in addition to R3 and R4; R1 is C1–C4 alkyl; R2 is H or an electron withdrawing functional group; each R3 is independently NR5R6; each R4 is independently C1–C4 alkyl or C1–C4 alkoxy, or one R3 and one R4, together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring; R5 and R6 are each independently H, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or R5 and R6, together with the nitrogen to which they are attached, form a substituted or unsubstituted ring; m and n are each independently 0, 1, 2, 3, or 4.
2. The compound of claim 0, or a pharmaceutically acceptable salt thereof, wherein Ring A is a substituted or unsubstituted heteroaromatic ring fused to ring B.
3. The compound of claim 0 or 0, having the structure of Formula (II): (II), or a pharmaceutically acceptable salt thereof, wherein R9 is H or straight or branched C1–C4 alkyl.
4. The compound of claim 3 wherein R9 is methyl.
5. The compound of claim 1 or 2, having the structure of Formula (III): (III) or a pharmaceutically acceptable salt thereof, wherein R10 is straight or branched C1–C4 alkyl C1–C4 alkyl or an electron withdrawing functional group.
6. The compound of any one of claims 1, 2, and 5, having the structure of Formula (IIIa): (IIIa) or a pharmaceutically acceptable salt thereof.
7. The compound of claims 5 or 6 wherein R10 is selected from halogen, -CH3, -SO2CF3, -N(R11)2, -NO2, -SO2R12, -C≡N, - C(R13)3, -COR14, -CO2R16, -CON(R17)2, and -N=O, wherein: each of R11, R12, R14, R16, or R17 is independently H or straight or branched C1–C4 alkyl; and each R13 is halogen.
8. The compound of any one of claims 5-7 wherein R10 is -Cl.
9. The compound of any one of claims 5-7 wherein R10 is -C≡N.
10. The compound of any one of claims 1-9 wherein R1 is straight or branched chain C1– C4 alkyl.
11. The compound of any one of claims 1-10 wherein R1 is methyl.
12. The compound of any one of claims 1-11wherein each R4 is independently straight or brached C1–C4 alkyl or C1–C4 alkoxy, or one R3 and one R4, together with the atoms of ring C to which they are attached, form a substituted or unsubstituted ring.
13. The compound of any one of claims 0-12 wherein R2 is selected from H, halogen, - SO2CF3, -N(R32)2, -NO2, -SO2R26, -C≡N, -C(R33)3, -COR28, -CHO, -COR29, -CO2R30, - CON(R31)2, and -N=O, wherein: each of R32, R26, R28, R29, R30, or R31 is independently H or C1–C4 alkyl; and each R33 is halogen.
14. The compound of any one of claims 0-12 or wherein R2 is selected from H, halogen, - SO2CF3, -N(R32)2, -NO2, -SO2R26, -C≡N, -C(R33)3, -COR28, -CHO, -COR29, -CO2R30, - CON(R31)2, and -N=O, wherein: each of R32, R26, R28, R29, R30, or R31 is independently H or straight or branched C1– C4 alkyl; and each R33 is halogen.
15. The compound of any one of claims 0-14 wherein R2 is H.
16. The compound of any one of claims 0-14 wherein R2 is -C≡N.
17. The compound of any one of claims 0-16 wherein ring C is further substituted by one or more substituents selected from R15, R18, R19, R20, R21, and R22, wherein: R15 is C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R18 and R19 are each, independently H, OH, C1–C4 alkoxy, C1–C4 alkyl, or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R20 is H, C1–C4 alkyl, or an electron withdrawing group, or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring; R21 is H, C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R22 is C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and R23 an R24 are each, independently H, C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
18. The compound of any one of claims 0-16 wherein ring C is further substituted by one or more substituents selected from R15, R18, R19, R20, R21, and R22, wherein: R15 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R18 and R19 are each, independently H, OH, C1–C4 alkoxy, straight or branched C1–C4 alkyl, or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R20 is H, straight or branched C1–C4 alkyl, or an electron withdrawing group, or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring; R21 is H, straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R22 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and R23 an R24 are each, independently H, straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
19. The compound of any one of claims 0-16 wherein ring C is further substituted by one or more substituents selected from R15, R18, R19, R20, R21, and R22, wherein: R15 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R18 and R19 are each, independently C1–C4 alkoxy, C1–C4 alkyl, or -NR23R24, or R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted ring; R20 is straight or branched C1–C4 alkyl, or an electron withdrawing group, or R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached, form a substituted or unsubstituted ring; R21 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl; R22 is straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and R23 and R24 are each, independently straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
20. The compound of any one of claims 1 to 19 wherein ring C is selected from ; ; ; and , wherein: indicates the point of attachment of ring C to ring B.
21. The compound of any one of claims 17-20, wherein R21 is straight or branched C1–C4 alkyl.
22. The compound of any one of claims 17-21, wherein R22 is straight or branched C1–C4 alkyl.
23. The compound of any one of claims 17-22, wherein R15 is straight or branched C1–C4 alkyl.
24. The compound of any one of claims 1 to 16 wherein ring C is , wherein R18 and R19, together with the carbon atom to which they are attached, form a substituted or unsubstituted 5- or 6-membered carbocyclic ring or a substituted or unsubstituted 5- or 6-membered heterocyclic ring.
25. The compound of any one of claims 1 to 16 wherein ring C is , wherein R18 and R20, or R19 and R20, together with the carbon atoms to which they are attached form a substituted or unsubstituted 5-, 6-, or 7-membered carbocyclic ring or a substituted or unsubstituted 5-, 6-, or 7-membered heterocyclic ring.
26. The compound of any one of claims 17-25 wherein R20 is straight or brached C1–C4 alkyl.
27. The compound of any one of claims 0-16 wherein ring C is selected from
Figure imgf000608_0001
, , , , , , ,
Figure imgf000608_0002
, and
Figure imgf000608_0003
.
28. The compound of any one of claims 1-16 wherein ring C is , wherein R21 is , wherein each R25 is independently selected from OH, halogen, C1–C4 alkyl, C1–C4 alkoxy, C1– C4 haloalkyl, C1–C4 haloalkoxy, substituted and unsubstituted aryl, and substituted and unsubstituted heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R21 to N.
29. The compound of any one of claims 1-16 wherein ring C is , wherein R21 is , wherein each R25 is independently selected from OH, halogen, straight or branched C1–C4 alkyl, C1–C4 alkoxy, straight or branched C1–C4 haloalkyl, C1–C4 haloalkoxy, substituted and unsubstituted aryl, and substituted and unsubstituted heteroaryl; t is 0, 1, or 2; and indicates the point of attachment of R21 to N.
30. The compound of claim 28 or 29 wherein each R25 is independently selected from OH, halogen, C1–C4 alkyl, C1–C4 alkoxy, C1–C4 haloalkyl, C1–C4 haloalkoxy, unsubstituted aryl, and unsubstituted heteroaryl.
31. The compound of claim 28 or 29 wherein each R25 is independently selected from OH, halogen, straight or branched C1–C4 alkyl, C1–C4 alkoxy, straight or branched C1–C4 haloalkyl, C1–C4 haloalkoxy, unsubstituted aryl, and unsubstituted heteroaryl. N N
32. The compound of claim 29 wherein ring C is R21.
33. The compound of any one of claims 017-20, and 27-32 wherein R21 is selected from ; ; ; ; ; ; and .
34. The compound of any one of claims 17-19 and 22 wherein ring C is selected from ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; and .
35. The compound of any one of claims 17-19, 27, and 34 wherein R22 is C1–C4 alkyl, unsubstituted aryl, or unsubstituted heteroaryl.
36. The compound of claim 35 wherein R22 is straight or branched C1–C4 alkyl.
37. The compound of any one of claims 1 to 16 wherein ring C is , wherein R18 and R19 are each independently selected from -H, -OH, alkyl, and , wherein: R26 is H, C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, C1–C4 alkyl, halogen, C1–C4 alkoxy, or C1–C4 haloalkoxy, or two adjacent R27 groups, and the carbon atoms two which they are attached, form a substituted or unsubstituted 5- or 6-membered ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R18 and R19 to ring C.
38. The compound of any one of claims 1 to 16 wherein ring C is , wherein R18 and R19 are each independently selected from -H, -OH, C1–C4 alkyl, and , wherein: R26 is H, C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, C1–C4 alkyl, halogen, C1–C4 alkoxy, or C1–C4 haloalkoxy, or two adjacent R27 groups, and the carbon atoms two which they are attached, form a substituted or unsubstituted 5- or 6-membered ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R18 and R19 to ring C.
39. The compound of any one of claims 1 to 16 wherein ring C is , or a pharmaceutically acceptable salt thereof, wherein R18 and R19 are each independently selected from -H, -OH, straight or branched C1–C4 alkyl, and , wherein: R26 is H, straight or branched C1–C4 alkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; each R27 is -OH, straight or branched C1–C4 alkyl, halogen, C1–C4 alkoxy, or C1–C4 haloalkoxy, or two adjacent R27 groups, and the carbon atoms two which they are attached, form a substituted or unsubstituted 5- or 6-membered ring; q is 0, 1, or 2; and indicates the point of attachment of one or both of R18 and R19 to ring C.
40. The compound of any one of claims 37-39 wherein R26 is straight or branched C1–C4 alkyl.
41. The compound of any one of claims 1 to 19 wherein ring C is selected from
Figure imgf000612_0001
;
Figure imgf000612_0002
42. The compound of any one of claims 27 and 34-41 wherein R18 and R19, together with the carbon atom to which they are attached, form , wherein: X and Y are each, independently CH2, NR29, O, or C=O; each R28 is independently selected from -OH, halogen, C1–C4 alkyl, C1–C4 haloalkyl, C1–C4 alkoxy, and C1–C4 haloalkoxy; R29 is H or C1–C4 alkyl; and w is 0, 1, or 2.
43. The compound of any one of claims 27 and 34-41 wherein R18 and R19, together with the carbon atom to which they are attached, form , wherein: X and Y are each, independently CH2, NR29, O, or C=O; each R28 is independently selected from -OH, halogen, straight or branched C1–C4 alkyl, straight or branched C1–C4 haloalkyl, C1–C4 alkoxy, and C1–C4 haloalkoxy; R29 is H or C1–C4 alkyl; and w is 0, 1, or 2.
44. The compound of any one of claims 0–16 wherein ring C is selected from ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ;
Figure imgf000615_0001
Figure imgf000616_0001
Figure imgf000617_0001
Figure imgf000618_0001
; ; ; ; ; ;
Figure imgf000618_0002
; and
Figure imgf000618_0003
45. The compound of any one of claims 0–16 wherein ring C is selected from
Figure imgf000618_0004
; ; ;
Figure imgf000618_0005
; ; and
Figure imgf000618_0006
.
46. The compound of any one of claims 1-16 wherein ring C is selected from ; ; ; ; ; ; ; and .
47. The compound of any one of claims 1-16 wherein ring C is selected from ; ; ; and , wherein R34 is C1–C4 alkyl.
48. The compound of any one of claims 1-16 wherein ring C is selected from ; ; ; and , wherein R34 is straight or branched C1–C4 alkyl.
49. The compound of any one of claims 0-16, or a pharmaceutically acceptable salt thereof, wherein ring C is , wherein R15 is ; ; ; or .
50. The compound of any one of claims 0-16 wherein ring C is selected from ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ;
Figure imgf000621_0001
; ; ; ;
Figure imgf000621_0002
; and
Figure imgf000621_0003
.
51. The compound of any one of claims 0-16 wherein ring C is or .
52. A compound selected from: ; ; ; ; ; ; ;
Figure imgf000623_0001
Figure imgf000624_0001
Figure imgf000625_0001
Figure imgf000626_0001
Figure imgf000627_0001
Figure imgf000628_0001
Figure imgf000629_0001
Figure imgf000630_0001
Figure imgf000631_0001
Figure imgf000632_0001
;
Figure imgf000633_0001
Figure imgf000634_0001
Figure imgf000635_0001
Figure imgf000636_0001
Figure imgf000637_0001
Figure imgf000638_0001
Figure imgf000639_0001
Figure imgf000640_0001
Figure imgf000641_0001
Figure imgf000642_0001
Figure imgf000643_0001
Figure imgf000644_0001
Figure imgf000645_0001
Figure imgf000646_0001
Figure imgf000647_0001
Figure imgf000648_0001
Figure imgf000649_0001
Figure imgf000650_0001
Figure imgf000651_0001
;
Figure imgf000652_0001
Figure imgf000653_0001
Figure imgf000654_0001
; and and
Figure imgf000654_0003
Figure imgf000654_0002
Figure imgf000655_0001
; or a pharmaceutically acceptable salt thereof.
53. A compound selected from:
Figure imgf000655_0002
; ; ;
Figure imgf000655_0003
Figure imgf000656_0001
Figure imgf000657_0001
Figure imgf000658_0001
Figure imgf000659_0001
Figure imgf000660_0001
Figure imgf000661_0001
Figure imgf000662_0001
Figure imgf000663_0001
Figure imgf000664_0001
; ; ; ;
Figure imgf000664_0003
Figure imgf000664_0002
; and , or a pharmaceutically acceptable salt thereof, wherein: X is CH2, O, or NR29; and R29 is H or C1–C4 alkyl.
54. The compound of claim 53, wherein R29 is straight or branched C1–C4 alkyl.
55. A compound, wherein the compound is selected from:
Figure imgf000664_0004
Figure imgf000665_0001
Figure imgf000666_0001
Figure imgf000667_0001
Figure imgf000668_0001
Figure imgf000669_0001
Figure imgf000670_0001
Figure imgf000671_0001
Figure imgf000672_0001
Figure imgf000673_0001
Figure imgf000674_0001
Figure imgf000675_0001
or a pharmaceutically acceptable salt thereof.
56. A pharmaceutical composition comprising a compound any one of claims 0–55 and a pharmaceutically acceptable excipient.
57. The pharmaceutical composition of claim 56, further comprising one or more additional anti-cancer agents.
58. The pharmaceutical composition of claim 57, wherein at least one of the additional anti-cancer agents is an immune checkpoint inhibitor.
59. A method of treating cancer, comprising administering a therapeutically effective amount of the compound of any one of claims 1-55 or the pharmaceutical composition of any one of claims 56-58 to a subject in need thereof.
60. The method of claim 59, wherein the cancer is selected from colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, head and/or neck cancer, lymphoma, lymphoma, leukemia, and melanoma.
61. A method of inhibiting the activity of diacylglycerol kinase alpha (DGKα), comprising administering a therapeutically effective amount of the compound of any one of claims 1-55 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of any one of claims 56-58 to a subject in need thereof.
PCT/US2023/012145 2022-02-01 2023-02-01 Dgk targeting compounds and uses thereof WO2023150186A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263305668P 2022-02-01 2022-02-01
US63/305,668 2022-02-01

Publications (1)

Publication Number Publication Date
WO2023150186A1 true WO2023150186A1 (en) 2023-08-10

Family

ID=85476265

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/012145 WO2023150186A1 (en) 2022-02-01 2023-02-01 Dgk targeting compounds and uses thereof

Country Status (1)

Country Link
WO (1) WO2023150186A1 (en)

Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006105021A2 (en) 2005-03-25 2006-10-05 Tolerrx, Inc. Gitr binding molecules and uses therefor
WO2006122150A1 (en) 2005-05-10 2006-11-16 Incyte Corporation Modulators of indoleamine 2,3-dioxygenase and methods of using the same
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007075598A2 (en) 2005-12-20 2007-07-05 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2008036653A2 (en) 2006-09-19 2008-03-27 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2008036642A2 (en) 2006-09-19 2008-03-27 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2008132601A1 (en) 2007-04-30 2008-11-06 Immutep Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2009009116A2 (en) 2007-07-12 2009-01-15 Tolerx, Inc. Combination therapies employing gitr binding molecules
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2009073620A2 (en) 2007-11-30 2009-06-11 Newlink Genetics Ido inhibitors
WO2009156652A1 (en) 2008-05-29 2009-12-30 Saint-Gobain Centre De Recherches Et D'etudes Europeen Cellular structure containing aluminium titanate
WO2010019570A2 (en) 2008-08-11 2010-02-18 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011028683A1 (en) 2009-09-03 2011-03-10 Schering Corporation Anti-gitr antibodies
WO2011056652A1 (en) 2009-10-28 2011-05-12 Newlink Genetics Imidazole derivatives as ido inhibitors
WO2011070024A1 (en) 2009-12-10 2011-06-16 F. Hoffmann-La Roche Ag Antibodies binding preferentially human csf1r extracellular domain 4 and their use
WO2011109400A2 (en) 2010-03-04 2011-09-09 Macrogenics,Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
WO2011107553A1 (en) 2010-03-05 2011-09-09 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011131407A1 (en) 2010-03-05 2011-10-27 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
WO2012032433A1 (en) 2010-09-09 2012-03-15 Pfizer Inc. 4-1bb binding molecules
WO2012142237A1 (en) 2011-04-15 2012-10-18 Newlink Geneticks Corporation Fused imidazole derivatives useful as ido inhibitors
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013087699A1 (en) 2011-12-15 2013-06-20 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2013119716A1 (en) 2012-02-06 2013-08-15 Genentech, Inc. Compositions and methods for using csf1r inhibitors
WO2013132044A1 (en) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2013169264A1 (en) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014036357A1 (en) 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2020006016A1 (en) * 2018-06-27 2020-01-02 Bristol-Myers Squibb Company Naphthyridinone compounds useful as t cell activators
WO2020006018A1 (en) * 2018-06-27 2020-01-02 Bristol-Myers Squibb Company Substituted naphthyridinone compounds useful as t cell activators
WO2021133750A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted bicyclic piperidine derivatives useful as t cell activators
WO2021133749A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted piperazine derivatives useful as t cell activators
WO2021133752A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted heteroaryl compounds useful as t cell activators
WO2021133748A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted quinolinonyl piperazine compounds useful as t cell activators
WO2021258010A1 (en) * 2020-06-19 2021-12-23 Gossamer Bio Services, Inc. Oxime compounds useful as t cell activators

Patent Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006105021A2 (en) 2005-03-25 2006-10-05 Tolerrx, Inc. Gitr binding molecules and uses therefor
WO2006122150A1 (en) 2005-05-10 2006-11-16 Incyte Corporation Modulators of indoleamine 2,3-dioxygenase and methods of using the same
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007075598A2 (en) 2005-12-20 2007-07-05 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2008036653A2 (en) 2006-09-19 2008-03-27 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2008036642A2 (en) 2006-09-19 2008-03-27 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
WO2008132601A1 (en) 2007-04-30 2008-11-06 Immutep Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2009009116A2 (en) 2007-07-12 2009-01-15 Tolerx, Inc. Combination therapies employing gitr binding molecules
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2009073620A2 (en) 2007-11-30 2009-06-11 Newlink Genetics Ido inhibitors
WO2009156652A1 (en) 2008-05-29 2009-12-30 Saint-Gobain Centre De Recherches Et D'etudes Europeen Cellular structure containing aluminium titanate
WO2010019570A2 (en) 2008-08-11 2010-02-18 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011028683A1 (en) 2009-09-03 2011-03-10 Schering Corporation Anti-gitr antibodies
WO2011056652A1 (en) 2009-10-28 2011-05-12 Newlink Genetics Imidazole derivatives as ido inhibitors
WO2011070024A1 (en) 2009-12-10 2011-06-16 F. Hoffmann-La Roche Ag Antibodies binding preferentially human csf1r extracellular domain 4 and their use
WO2011109400A2 (en) 2010-03-04 2011-09-09 Macrogenics,Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
WO2011107553A1 (en) 2010-03-05 2011-09-09 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011131407A1 (en) 2010-03-05 2011-10-27 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
WO2012032433A1 (en) 2010-09-09 2012-03-15 Pfizer Inc. 4-1bb binding molecules
WO2012142237A1 (en) 2011-04-15 2012-10-18 Newlink Geneticks Corporation Fused imidazole derivatives useful as ido inhibitors
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013087699A1 (en) 2011-12-15 2013-06-20 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2013119716A1 (en) 2012-02-06 2013-08-15 Genentech, Inc. Compositions and methods for using csf1r inhibitors
WO2013132044A1 (en) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2013169264A1 (en) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014036357A1 (en) 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2020006016A1 (en) * 2018-06-27 2020-01-02 Bristol-Myers Squibb Company Naphthyridinone compounds useful as t cell activators
WO2020006018A1 (en) * 2018-06-27 2020-01-02 Bristol-Myers Squibb Company Substituted naphthyridinone compounds useful as t cell activators
WO2021133750A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted bicyclic piperidine derivatives useful as t cell activators
WO2021133749A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted piperazine derivatives useful as t cell activators
WO2021133752A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted heteroaryl compounds useful as t cell activators
WO2021133748A1 (en) * 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted quinolinonyl piperazine compounds useful as t cell activators
WO2021258010A1 (en) * 2020-06-19 2021-12-23 Gossamer Bio Services, Inc. Oxime compounds useful as t cell activators

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
"The McGraw-Hill Dictionary of Chemical Terms", 1985, MCGRAW-HILL
AVILA-FLORES, A. ET AL., IMMUNOLOGY AND CELL BIOLOGY, vol. 95, 2017, pages 549 - 563
CHEN, S. S. ET AL., FRONT CELL DEV BIOL, vol. 4, 2016, pages 130
FACCIABENE ET AL., CANCER RES, vol. 72, 2012, pages 2162 - 71
GOLDSTEIN ET AL., CLIN. CANCER RES., vol. 1, 1995, pages 1311 - 18
GRIFFITHS ET AL.: "Introduction to Genetic Analysis", 1999, W. H. FREEMAN & CO.
JING, W. ET AL., CANCER RESEARCH, vol. 77, 2017, pages 5676 - 86
KOHL ET AL., NAT. MED., vol. 1, no. 8, 1995, pages 792 - 97
KRISHNA, S. ET AL., FRONT IMMUNOLOGY, vol. 4, 2013, pages 178
MELLMAN ET AL., NATURE, vol. 480, 2011, pages 480 - 89
MIZOGUCHI ET AL., SCIENCE, vol. 258, 1992, pages 1795 - 98
NOESSNER, E, FRONT CELL DEV, vol. 5, 2017, pages 16
OLENCHOCK B. A. ET AL., NATURE, vol. 11, 2006, pages 1174 - 81
PRINZ, P. U. ET AL., J IMMUNOLOGY, vol. 12, 2012, pages 5990 - 6000
SAUSVILLE, CURR. MED. CHEM. ANTI-CANC. AGENTS, vol. 3, 2003, pages 47 - 56
SEKULIC ET AL., CANCER RES., vol. 60, 2000, pages 3504 - 13
SJOBLOM ET AL., SCIENCE, vol. 314, 2006, pages 268 - 74
TOPALIAN ET AL., CURR. OPIN. IMMUNOL., vol. 24, 2012, pages 1 - 6
VLAHOS ET AL., J. BIOL., vol. 269, 1994, pages 5241 - 48
WUTSGREENE: "Greene's Protective Groups in Organic Synthesis", 2007, WILEY AND SONS
ZHA Y ET AL., NATURE IMMUNOLOGY, vol. 12, 2006, pages 1343

Similar Documents

Publication Publication Date Title
EP3322711B1 (en) Hpk1 inhibitors and methods of using same
JP6532474B2 (en) 6-heteroaryloxy- and 6-aryloxy-quinoline-2-carboxamides and their use
KR101961500B1 (en) Serine/threonine kinase inhibitors
KR20210018282A (en) Tetrahydro-imidazo[4,5-c]pyridine derivatives as PD-L1 immunomodulators
JP5978317B2 (en) Novel indolizine compounds, methods for their preparation and pharmaceutical compositions containing them
CN113646049A (en) Pyrimidone derivatives as SHP2 antagonists
TW201828940A (en) Immunomodulator compounds and methods of use
JP2020504737A (en) Heterocyclic compounds as immunomodulators
CN113795483A (en) Carboxamide-pyrimidine derivatives as SHP2 antagonists
CN110831600A (en) Indole AHR inhibitors and uses thereof
WO2021258010A1 (en) Oxime compounds useful as t cell activators
JP2022552324A (en) Bicyclic heterocycles as FGFR inhibitors
CA3163003A1 (en) Substituted heteroaryl compounds useful as t cell activators
US11964973B2 (en) Substituted bicyclic compounds useful as T cell activators
JP7384535B2 (en) Quinazoline compounds and their preparation, use and pharmaceutical compositions
EP4090663A1 (en) Map4k1 inhibitors
TW202115025A (en) Aminopyrazine compounds as hpk1 inhibitor and the use thereof
TW202302589A (en) Spirocyclic lactams as jak2 v617f inhibitors
CN111205310A (en) Heterocyclic fused pyrimidine derivative, and pharmaceutical composition and application thereof
JP2024510176A (en) MAP4K1 inhibitor
CN115151534A (en) Inhibitors of interleukin-1 receptor-related kinase (IRAK)/FMS-like receptor tyrosine kinase (FLT 3), pharmaceutical products thereof, and methods thereof
WO2023150186A1 (en) Dgk targeting compounds and uses thereof
CA3214240A1 (en) Pyridinyl substituted oxoisoindoline compounds for the treatment of cancer
CN117396477A (en) Pyridyl-substituted oxoisoindoline compounds for the treatment of cancer
WO2023122772A1 (en) Oxime derivatives useful as t cell activators

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: 23708960

Country of ref document: EP

Kind code of ref document: A1