US Patent Application for HPK1 ANTAGONISTS AND THEIR USE Patent Application (Application No. 20230096641, issued March 30, 2023) (2023)

CROSS REFERENCE TO RELATED ORDERS

This application claims the benefit of US Provisional Application Serial Number 63/148,857, filed February 12, 2021, the contents of which are incorporated herein by reference.

TECHNICAL FIELD OF THE INVENTION

The present invention relates to compounds and methods useful for antagonizing precursor hematopoietic kinase 1 (HPK1). The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using these compositions in the treatment of various disorders.

BACKGROUND OF THE INVENTION

Precursor hematopoietic kinase 1 (HPK1), also known as mitogen-activated protein kinase 1 (MAP4K1), is a hematopoietic cell-restricted member of the Ste20 serine/threonine kinase superfamily. The MAP4Ks family includes MAP4K1/HPK1, MAP4K2/GCK, MAP4K3/GLK, MAP4K4/HGK, MAP4K5/KHS, and MAP4K6/MINK. HPK1 is a tissue-specific upstream activator of the MEKK/JNK/SAPK signaling pathway.

HPK1 is of particular interest as it is expressed predominantly on hematopoietic cells such as T cells, B cells, macrophages, dendritic cells, neutrophils and mast cells (Hu, M.C., et al., Genes Dev, 1996. 10(18): pp. 2251-64, Kiefer, F., et al., EMBO J, 1996. 15(24): pp. 7013-25). HPK1 kinase activity has been shown to be induced upon activation of T cell receptors (TCRs) (Liou, J., et al., Immunity, 2000. 12(4): pp. 399-408), B cell receptors (BCR ) (Liou, J., et al., Immunity, 2000. 12(4): pp. 399-408), transforming growth factor receptor (TGF-PR) (Wang, W., et al., J Biol Chem, 1997 272(36): pp 22771-5 Zhou, G., et al., J Biol Chem, 1999. 274(19): pp 13133-8), or coupled PGE2 receptors to Gs (EP2 and EP4) ( Ikegami, R. et al., J Immunol, 2001. 166(7): pp. 4689-96). As such, HPK1 regulates various functions of various immune cells. HPK1 is also an example of a negative regulator of dendritic cell activation and T and B cell responses that can be targeted to enhance antitumor immunity. HPK1 is expressed predominantly in hematopoietic cells, including early progenitor cells. In T cells, HPK1 is thought to downregulate T cell activation, preventing the persistence of signaling microclusters through phosphorylation of SLP76 at Ser376 (Di Bartolo et al. (2007) JEM 204: 681 -691) and Gads at Thr254, leading to the recruitment of 14-3-3 Proteins that bind SLP76 and phosphorylated Gads and release the SLP76-Gads-14-3-3 complex from LAT-containing microclusters (Lasserre et al. (2011) J Cell Biol 195(5): 839-853). HPK1 can also be activated in response to prostaglandin E2, which is commonly secreted by tumors and contributes to the escape of tumor cells from the immune system.

HPK1 is important in regulating the functions of various immune cells and has been implicated in autoimmune diseases and antitumor immunity (Shui, J.W., et al., Nat Immunol, 2007. 8(1): pp. 84-91; Wang, X., et al., J Biol Chem, 2012. 287(14): pp. 11037-48).

SUMMARY OF THE INVENTION

It has now been discovered that the compounds of this invention and their pharmaceutically acceptable compositions are effective as antagonists of HPK1. In certain embodiments, the invention provides compounds of the formulas shown herein.

The compounds of the present invention and their pharmaceutically acceptable compositions are useful for the treatment of a variety of diseases, disorders or conditions associated with the regulation of signaling pathways involving HPK1 kinases. Such diseases, disorders or conditions include those described herein.

The compounds provided by this invention are also useful for studying HPK1 enzymes in biological and pathological phenomena; the study of intracellular signal transduction pathways that occur in body tissues; and the comparative evaluation of new inhibitors of HPK1 or other regulators of kinases, signaling pathways and cytokine levels in vitro or in vivo.

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

In certain aspects, the present invention provides a compound of formula I:

or a pharmaceutically acceptable salt thereof, wherein each of X, Y, Z, A, L, R1, R2, R3, R4, m and n are as defined below and described in embodiments herein, both individually and in combination.

In some embodiments, the present invention provides a pharmaceutical composition comprising a compound provided herein and a pharmaceutically acceptable carrier, excipient or diluent.

In some embodiments, the present invention provides a method of treating an HPK1-mediated disease, disorder or condition comprising administering to a patient in need thereof a compound provided herein, or a pharmaceutically acceptable salt thereof.

2. Connections and settings

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

The term "aliphatic" or "aliphatic group", as used herein, means a straight (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is fully saturated or contains one or more units of unsaturation, or a monocyclic or bicyclic hydrocarbon. , which is fully saturated or contains one or more unsaturation units, but is not aromatic (also referred to herein as "carbon cycle", "cycloaliphatic" or "cycloalkyl"), which has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1 to 6 aliphatic carbon atoms. In some embodiments, the aliphatic groups contain from 1 to 5 aliphatic carbon atoms. In other embodiments, the aliphatic groups contain from 1 to 4 aliphatic carbon atoms. In still other embodiments, the aliphatic groups contain 1 to 3 aliphatic carbon atoms, and in still other embodiments, the aliphatic groups contain 1 to 2 aliphatic carbon atoms. In some embodiments, "cycloaliphatic" (or "carbon cycle" or "cycloalkyl") refers to a monocyclic C3-C6Hydrocarbon that is fully saturated or contains one or more unsaturated residues, but is not aromatic and has only one point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, substituted or unsubstituted alkyl, alkenyl, linear or branched alkynyl, and hybrids thereof, such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl, or (cycloalkyl)alkenyl.

As used herein, the term "bridged bicycle" refers to any bicyclic, ie, saturated or partially unsaturated, carbocyclic or heterocyclic H. ring system with at least one bridge. As defined by IUPAC, a "bridge" is an unbranched chain of atoms or a valence atom or a bond connecting two bridgeheads, where a "bridgehead" is any skeletal atom of the ring system attached to three or more skeletal atoms (excluding hydrogen). In some embodiments, a bridging bicyclic group has 7 to 12 ring members and 0 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridged bicyclic groups are well known in the art and include the groups listed below, where each group is attached to the remainder of the molecule at any replaceable carbon or nitrogen atom. Unless otherwise indicated, a bridging bicyclic group is optionally substituted with one or more substituents as indicated for aliphatic groups. Additionally or alternatively, each substitutable nitrogen of a bridged bicyclic group is optionally substituted. Examples of bridge bikes include:

The term "lower alkyl" refers to a C1-4linear or branched alkyl group. Examples of lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.

The term "lower haloalkyl" refers to a C1-4linear or branched alkyl group substituted by one or more halogen atoms.

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

The term "unsaturated" as used herein means that a moiety has one or more unsaturation residues.

As used here, the term "divalent C1-8(o c1-6) saturated or unsaturated, straight or branched hydrocarbon chain" refers to divalent straight or branched alkylene, alkenylene and alkynylene chains, as defined herein.

The term "alkylene" refers to a divalent alkyl group. An "alkylene chain" is a polymethylene group; H.-(CH2)norte- where n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2 or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylenes are hydrogen atoms are replaced by a substituent. Suitable substituents include those described below for a substituted aliphatic group.

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

The term "halogen" means F, Cl, Br or I.

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

The terms "heteroaryl" and "heteroar-", used alone or as part of a larger unit, e.g. B. "heteroaralkyl" or "heteroaralkoxy" refers to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; with 6, 10, or 14 shared π electrons in a cyclic arrangement; and has from one to five heteroatoms in addition to carbon atoms. The term "heteroatom" refers to nitrogen, oxygen, or sulfur and includes any oxidized form of nitrogen or sulfur and any quaternized form of basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms "heteroaryl" and "heteroar-", as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where, unless otherwise specified, the radical or the point of attachment is the same is the heteroaromatic ring or one of the rings to which the heteroaromatic ring is fused. Non-limiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolyl, isoquinolyl, cinlinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenoquinothiazinyl, isoquinozinylhydro, quinoquinophene. A heteroaryl group can be mono or bicyclic. The term "heteroaryl" can be used interchangeably with the terms "heteroaryl ring", "heteroaryl group" or "heteroaromatic", each of which includes rings that are optionally substituted. The term "heteroaralkyl" refers to an alkyl group substituted by a heteroaryl, in which the alkyl and heteroaryl moieties are optionally substituted independently.

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

A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure, and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenylpyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazapinyl, morpho-oxalinyl, 2-pyrozaoxalinyl [3.3]heptane and quinuclidinyl. The terms "heterocycle", "heterocyclyl", "heterocyclyl ring", "heterocyclic group", "heterocyclic moiety" and "heterocyclic moiety" are used interchangeably herein and also include groups in which a heterocyclyl ring is fused with other aryl, heteroaryl or cycloaliphatic rings such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl or tetrahydroquinolinyl. A heterocyclic group can be mono or bicyclic. The term "heterocyclylalkyl" refers to an alkyl group substituted by a heterocyclyl, in which the alkyl and heterocyclyl moieties are optionally substituted independently.

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

As described herein, compounds of the invention may contain "optionally substituted" moieties. In general, the term "substituted", whether or not preceded by the term "optional", means that one or more hydrogen atoms of the designated moiety are replaced by a suitable substituent. Unless otherwise specified, an "optionally substituted" group may have a suitable substituent at any substitutable position in the group, and when more than one position in a given structure can be substituted by more than one substituent selected from a specified group, the substituent at each position can be the same or different. Combinations of substituents contemplated by this invention are preferably those that result in the formation of stable or chemically viable compounds. As used herein, the term "stable" refers to compounds that are not substantially changed when subjected to conditions that allow their manufacture, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes described in this document. . .

Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halo; -(CH)2)0-4Rº; -(CH2)0-4ODERo; —O(CH2)0-4Rº, —O—(CH2)0-4C(O)ORº; -(CH2)0-4CH(O)2; -(CH2)0-4SRo; -(CH2)0-4Ph, which can be replaced by Rº; -(CH)2)0-4The only2)0-1Ph, which can be replaced by Rº; -CH=CHPh which can be replaced by R°; -(CH)2)0-4The only2)0-1-pyridyl which may be substituted by R°; -NO2; -CN; -NORTH3; -(CH2)0-4N(Rº)2; -(CH2)0-4N(Rº)C(O)Rº; —N(Rº)C(S)Rº; -(CH2)0-4N(Rº)C(O)NRº2; —N(Rº)C(S)NRº2; -(CH2)0-4N(Rº)C(O)ORº; —N(Rº)N(Rº)C(O)Rº; —N(Rº)N(Rº)C(O)NRº2; —N(Rº)N(Rº)C(O)ORº; —N(Rº)C(NRº)N(Rº)2; -(CH2)0-4C(O)Rº; —C(S)Rº; -(CH2)0-4C(O)ORº; -(CH2)0-4C(O)SRº; -(CH2)0-4C(O)OSiRº3; -(CH2)0-4OC(O)Rº; —OC(O)(CH2)0-4SRo; —SC(S)Sr; -(CH2)0-4SC(O)Rº; -(CH2)0-4C(O)NRº2; —C(S)NR°2; -C(S)Sr; —SC(S)Sr, —(CH2)0-4OC(O)NRº2; —C(O)N(ORº)Rº; —C(O)C(O)Rº; -C(O)CH2C(O)Rº; —C(NORº)Rº; -(CH2)0-4SSRº; -(CH2)0-4SO)2Rº; -(CH2)0-4SO)2ODERo; -(CH2)0-4operating system (O)2Rº; -SO)2NO2; -(CH2)0-4S(O)Rº; —N(Rº)S(O)2NO2; —N(Rº)S(O)2Rº; —N(Oº)Rº; —C(NH)NRº2; -CORREOS)2Rº; —P(O)Rº2; —OP(O)Rº2; —OP(O)(Oº)2; —Sirº3; -(C1-4straight or branched chain alkylene)ON(Rº)2; o - (C1-4straight or branched chain alkylene)C(O)O—N(Rº)2, where each R° can be substituted as defined below and is independently hydrogen, C1-6aliphatic, -CH2Ph, —O(ONLY2)0-1Ph, - SOLO2-(5-6 membered heteroaryl ring) or a 5-6 membered saturated, partially unsaturated or aryl ring containing 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or notwithstanding the above definition, two independent occurrences of Rº together form its The intermediate atom(s) is a 3-12 membered monocyclic or bicyclic saturated, partially unsaturated, or aryl ring containing 0-4 independently selected nitrogen, oxygen, or sulfur heteroatoms that they may be substituted as defined below.

Suitable monovalent substituents on R° (or the ring formed by two independent occurrences of R° together with their intermediate atoms) are, independently, halogen, -(CH2)0-2R, -(haloR), -(CH2)0-2OH, -(CH2)0-2O, -(CH2)0-2CH(U)2; —Ah (OláR), -CN, -N3, -(CH2)0-2C(O)R, -(CH2)0-2C(O)OH, -(CH2)0-2C(O)U, -(CH2)0-2RS, -(CH2)0-2SH, -(CH2)0-2N.H2, -(CH2)0-2NIÑO, -(CH2)0-2NR2, -NO2, -Herr3, —OSiR3, -C(O)SR, -(C1-4straight or branched chain alkylene)C(O)OR, o —SSRwhere each Ris unsubstituted or, where "halogen" is prepended, is substituted only by one or more halogens and is independently selected from C1-4aliphatic, -CH2Ph, —O(ONLY2)0-1Ph, or a 5-6 membered saturated, partially unsaturated or aryl ring containing 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =O and =S.

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

Suitable substituents on the aliphatic group of R* include halo, -R, -(haloR), —OH, —O, —O(haloR), -CN, -C(O)OH, -C(O)OR, —NH2, -CHAPTER, —NR2, o no2, where each Ris unsubstituted or, where "halo" is prepended, is substituted only by one or more halogens and is independently C1-4aliphatic, -CH2Ph, —O(ONLY2)0-1Ph, or a 5-6 membered saturated, partially unsaturated or aryl ring containing 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.

Suitable substituents on a replaceable nitrogen of an "optionally substituted" group include -R, -NR2, -C(O)R, -C(O)U, -C(O)C(O)R, -C(O)CH2C(O)R, -SO)2R, -SO)2NR2, -C(S)NR2, -C(NH)NR2, o -N(R)SO)2R; where each Ris independently hydrogen, C1-6aliphatic which may be substituted as defined below, -OPh unsubstituted, or a saturated, partially unsaturated, or unsubstituted 5-6 membered aryl ring containing 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or not Notwithstanding the above definition, two independent occurrences of Rtogether with its intermediate atom(s) form a saturated, partially unsaturated or mono- or bicyclic 3-12 membered unsubstituted aryl ring containing 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.

Suitable substituents on the aliphatic group of R are independently halo, -R, -(haloR), —OH, —O, —O(haloR), -CN, -C(O)OH, -C(O)OR, —NH2, -CHAPTER, —NR2, o no2, where each Ris unsubstituted or, where "halo" is prepended, is substituted only by one or more halogens and is independently C1-4aliphatic, -CH2Ph, —O(ONLY2)0-1Ph, or a 5-6 membered saturated, partially unsaturated or aryl ring containing 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.

The term "pharmaceutically acceptable salt", as used herein, refers to those salts which, within sound medical judgment, are suitable for use in contact with human and lower animal tissues without undue toxicity, irritation, allergic reaction and similar, and are in an adequate risk-benefit ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Bergé et al. pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.

Pharmaceutically acceptable salts of the compounds of this invention include those derived from appropriate inorganic and organic acids and bases. Examples of non-toxic pharmaceutically acceptable acid addition salts are salts of an amino group formed with inorganic acids, such as hydrochloric, hydrobromic, phosphoric, sulfuric, and perchloric acids, or with organic acids, such as acetic, oxalic, maleic, tartaric, citric, succinic or malonic acid, or using other appropriate methods used in the art, such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate salts of phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like.

Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium, and N+(C1-4rent)4you go out. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Other pharmaceutically acceptable salts optionally include non-toxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate.

Unless otherwise indicated, the structures depicted herein are also intended to include all isomeric (eg, enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, the Z and E double bond isomers, and the Z and E conformational isomers. Thus, both individual stereochemical isomers and enantiomeric, diastereomeric, and geometric (or conformational) mixtures ) of the present compounds are within the scope of the invention. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Furthermore, unless otherwise indicated, the structures depicted herein are intended to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds with the present structures include the replacement of hydrogen with deuterium or tritium, or the replacement of a carbon with a13Neck14C-enriched carbon is within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents according to the present invention. In certain embodiments, a warhead unit, R1, a provided compound comprises one or more deuterium atoms. In certain embodiments, the B ring of a provided compound can be replaced with one or more deuterium atoms.

As used herein, an "HPK1 antagonist" or "HPK1 inhibitor" is a molecule that mediates one or more of the biological activities of HPK1 (eg, serine/threonine kinase activity, recruitment into the TCR complex, TCR activation, interaction with a binding partner protein, such as SLP76). Antagonism using the HPK1 antagonist does not necessarily indicate complete elimination of HPK1 activity. Instead, activity may decrease by a statistically significant amount, including, for example, a decrease of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% , 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 95%, or 100% HPK1 activity compared to an appropriate control. In some embodiments, the HPK1 antagonist reduces, inhibits, or otherwise decreases HPK1 serine/threonine kinase activity. In some of these embodiments, the HPK1 antagonist reduces, inhibits, or otherwise reduces HPK1-mediated phosphorylation of SLP76 and/or Gads. The compounds described herein directly bind to HPK1 and inhibit its kinase activity.

By "specific antagonist" is meant an agent that reduces, inhibits, or otherwise reduces the activity of a defined target more than that of an unrelated target. For example, a specific HPK1 antagonist reduces at least one HPK1 biological activity by a statistically greater amount than the antagonist's inhibitory effect on any other protein (eg, other serine/threonine kinases). In some embodiments, the IC50target antagonist is about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01%, 0.001 % or less IQ50from the antagonist to a non-target. The compounds described herein may or may not be a specific antagonist of HPK1. A specific HPK1 antagonist reduces the biological activity of HPK1 by an amount that is statistically greater than the inhibitory effect of the antagonist on any other protein (eg, other serine/threonine kinases). In certain embodiments, the HPK1 antagonist specifically inhibits HPK1 serine/threonine kinase activity. In some of these modalities, the CI50of HPK1 antagonist to IPK1 is approximately 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 0.1%, 0.01%, 0.001% or less than CI50the HPK1 antagonist to another serine/threonine kinase or type of kinase (eg, tyrosine kinase).

A compound of the present invention can be linked to a detectable entity. Such compounds are understood to be useful as imaging agents. One skilled in the art will recognize that a detectable moiety can be attached to a given compound through an appropriate substituent. As used herein, the term "suitable substituent" refers to an entity capable of covalently binding a detectable entity. Such fractions are well known to those skilled in the art and include groups, e.g. B. contain a carboxylate moiety, an amino moiety, a thiol moiety, or a hydroxyl moiety, to name a few. It will be appreciated that such moieties may be linked directly to a given compound or through a linking group such as a saturated or unsaturated divalent hydrocarbon chain. In some embodiments, such units can be attached using click chemistry. In some embodiments, such moieties can be attached via a 1,3-cycloaddition of an azide with an alkyne, optionally in the presence of a copper catalyst. Methods for using click chemistry are known in the art and include those described by Rostovtsev et al., Angew. chemistry in t. ed. 2002, 41, 2596-99 and Sun et al., Bioconjugate Chem., 2006, 17, 52-57.

As used in this document, the term "detectable entity" is used interchangeably with the term "tag" and refers to any entity that can be detected, e.g. B. Primary markers and secondary markers. Primary markers such as radioisotopes (for example, tritium,32when,33when,35S, bzw14C), mass tags and fluorescent markers are signal-generating groups that can be detected without further modification. Detectable fractions also include luminescent and phosphorescent groups.

The term "secondary marker", as used herein, refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate to generate a detectable signal. For biotin, the secondary intermediate can include streptavidin-enzyme conjugates. For antigen markers, secondary intermediates can include antibody-enzyme conjugates. Some fluorescent clusters act as secondary markers in that they transfer energy to another cluster in the process of non-radiative fluorescent resonance energy transfer (FRET) and the second cluster generates the detected signal.

As used herein, the terms "fluorescent label", "fluorescent dye", and "fluorophore" refer to moieties that absorb light energy at a defined excitation wavelength and emit light energy at a different wavelength. Examples of fluorescent markers include, but are not limited to: Alexa Fluor Dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660, and Alexa Fluor 680) , AMCA, AMCA-S, BODIPY COLORS (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/ 650, BODIPY 650/665), Carboxyrhodamine 6G, Carboxy-X-rodamine (ROX), Cascade Blue, Cascade Yellow, Coumarin 343, Cyanine dyes (Cy3, Cy5, Cy3.5, Cy5.5), Dansyl, Dapoxyl, Dialkylaminocoumarin, 4',5'-Dichloro-2',7'-Dimethoxyfluorescein, DM-NERF, Eosin, Erythrosine, Fluorescein, FAM, Hydroxycocoumarin, IRDyes (IRD40, IRD 700, IRD 800), JOE, Lissamine Rhodamine B, Blue Marine, Methoxycoumarin, Naphthofluorescein, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, PyMPO, Pyrene, Rhodamine B, Rhodamine 6G, Rhodamine Green, Rhodamine Red, Rodol Green, 2′, 4 ′,5′, Fluorescein 7'-tetra-bromosulfone, tetramethyl rhodamine (TMR), carboxytetramethyl rhodamine (TAMRA), Texas Red, Texas Red-X.

The term "mass tag", as used herein, refers to any entity capable of being unequivocally identified by its mass using mass spectrometry (MS) detection techniques. Examples of mass labels include electrophoric release labels such as N-[3-[4'-[(p-methoxytetrafluorobenzyl)oxy]phenyl]-3-methylglyceronyl]isonipecotic acid, 4'-[2,3,5, 6-Tetrafluoro-4-(pentafluorophenoxyl)]methylacetophenone and its derivatives. The synthesis and utility of these bulk tags are described in US Pat. Other examples of mass labels include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of variable length and composition of bases, oligopeptides, oligosaccharides, and other synthetic polymers of variable length and composition of monomers. A wide variety of organic, neutral and charged molecules (biomolecules or synthetic compounds) of an appropriate mass range (100-2000 daltons) can also be used as mass markers.

The terms "measurable affinity" and "measurable inhibition", as used herein, mean a measurable change in HPK1 protein kinase activity between a sample containing a compound of the present invention or a composition thereof and an HPK1 protein kinase. and an equivalent includes a sample comprising an HPK1 protein kinase in the absence of the compound or composition thereof.

3. Description of the Exemplary Modalities

As described above, in certain embodiments, the present invention provides a compound of formula I:

or a pharmaceutically acceptable salt thereof, in which:

  • X is N or CR3;
  • S is N or CR2;
  • Z is N or CR2;
  • A is N or CR2;
  • L is a covalent bond, -O-, -S-, -NR-, -S(O)2-, -SO)2NR—, —S(O)—, —S(O)NR—, —C(O)—, —C(O)O—, —C(O)NR—, —C(O)N(R) O—, —OC(O)—, —OC(O)NR—, —N(R)C(O)O—, —N(R)C(O)—, —N(R)S(O)2—; or L is a C1-4saturated or unsaturated, linear or branched bivalent hydrocarbon chain, one or two methylene units of the chain being optionally and independently substituted by -C(R)-2—, —N(R)—, —N(R)C(O)—, —C(O)N(R)—, —N(R)S(O)2-, -SO)2N(R)—, —O—, —C(O)—, —OC(O)—, —C(O)O—, —S—, —S(O)— o —S(O)2—;
  • R1is selected from H; W1-6aliphatic; phenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; which are replaced by q instances of RC;
  • every R2is selected independently of H; W1-6aliphatic; phenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 6-11 membered spirobicyclic, bridged, or saturated, partially unsaturated, or fused unsaturated spirobicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; which are replaced by r instances of RD; o R2halogen, -CN, -NO2, —O, —SR, —NO2, -SO)2R, —S(O)(NR)R, —S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)R, -N(R)C(O)OR, -N(R)C(O)NR2, -N(R)C(NR)NR2, —N(R)S(O)2NR2, —N(R)S(O)2R o -P(O)R2;
  • every R3is selected independently of H; W1-6aliphatic; phenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; which are replaced by s instances of Rmi; o R3halogen, -CN, -NO2, —O, —SR, —NO2, -SO)2R, —S(O)(NR)R, —S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)R, -N(R)C(O)OR, -N(R)C(O)NR2, -N(R)C(NR)NR2, —N(R)S(O)2NR2, —N(R)S(O)2R o -P(O)R2;
  • R4is it H or C1-6aliphatic; which is replaced by t instances of RF;
  • any instance of RC, RD, Rmiy R. S.Fis independently oxo, halogen, -CN, -NO2, —O, —SR, —NO2, -SO)2R, -S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(O)NR2, -N(R)C(NR)NR2, -N(R)NR2, —N(R)S(O)2NR2, —N(R)S(O)2R, -N=S(O)R2, -S(NR)(O)R, -N(R)S(O)R, -N(R)CN, -P(O)(R)NR2, -P(O)(R)OR o -P(O)R2; or any instance of RC, RD, Rmiy R. S.Findependently, an optionally substituted group selected from C1-6aliphatic; phenyl; naphthalenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; or a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; each of which is replaced by u instances of R;
  • each R is independently hydrogen, -CN, halogen, or an optionally substituted group selected from C1-6aliphatic; phenyl; naphthalenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; either:
  • two R groups on the same nitrogen are taken together with the nitrogen to form a saturated, partially unsaturated, or optionally substituted 4-7 membered heteroaryl monocyclic ring having, in addition to nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur ; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
  • m is 1, 2, or 3;
  • n is 0, 1 or 2;
  • q, r, s, and t are each independently 0, 1, 2, 3, or 4; AND
  • u is 0, 1, 2, 3, or 4.

In one embodiment, X is CR3; S is N or CR2; Z is N or CR2; and A is N or CR2.

In certain embodiments, X is CR3; and it's CR2; Z is N; and A is CR2.

In certain embodiments, X is CR3; Y is N; Z is RC2; and A is CR2.

In certain embodiments, X is CR3; and it's CR2; Z is RC2; and A is CR2.

In certain embodiments, X is CR3; and it's CR2; Z is RC2; and A is N

In certain embodiments, X is CR3; Y is N; Z is RC2; and A is N

In another embodiment, X is N; Y is N or CR2; Z is N or CR2; and A is N or CR2.

In certain embodiments, X is N; and it's CR2; Z is N; and A is CR2.

In certain embodiments, X is N; Y is N; Z is RC2; and A is CR2.

In certain embodiments, X is N; and it's CR2; Z is RC2; and A is CR2.

In certain embodiments, X is N; and it's CR2; Z is RC2; and A is N

In certain embodiments, X is N; Y is N; Z is RC2; and A is N

In certain embodiments, L is a covalent bond, -O-, -S-, -NR-, -S(O)2-, -SO)2NR—, —S(O)—, —S(O)NR—, —C(O)—, —C(O)O—, —C(O)NR—, —C(O)N(R) O—, —OC(O)—, —OC(O)NR—, —N(R)C(O)O—, —N(R)C(O)—, —N(R)C(O) NR—, —N(R)C(NR)NR—, —N(R)NR—, —N(R)S(O)2NR— o —N(R)S(O)2—.

In some embodiments, L is a covalent bond.

In certain embodiments, L is -O-, -S-, -NR-, -S(O)2-, -SO)2NR—, —S(O)—, —S(O)NR—, —C(O)—, —C(O)O—, —C(O)NR—, —C(O)N(R) O—, —OC(O)—, —OC(O)NR—, —N(R)C(O)O—, —N(R)C(O)—, —N(R)C(O) NR—, —N(R)C(NR)NR—, —N(R)NR—, —N(R)S(O)2NR— o —N(R)S(O)2—.

In certain embodiments, L is -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(O)N(R)O-, -OC(O) - , —OC(O)NR—, —N(R)C(O)O—, —N(R)C(O)—, —N(R)C(O)NR—, —N(R) C(NR)NR- o -N(R)NR-.

In certain embodiments, L is -NR-. In certain embodiments, L is -NH-.

In some embodiments, L is selected from those shown in Table 1 below.

In some embodiments, R1it's H.

In some embodiments, R1is C1-6aliphatic, which is replaced by q instances of RC; Phenyl replaced by q instances of RC; a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring substituted by q occurrences of RC; a 5 to 6 membered monocyclic heteroaryl ring containing 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur substituted by q instances of RC; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur substituted by q occurrences of RC.

In some embodiments, R1phenyl or a 5 to 6-membered monocyclic heteroaryl ring containing 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, each of which is replaced by q occurrences of RC.

In certain embodiments, R1ist -H, -Et, -i-Pr, s-Bu, geradkettiges oder verzweigtes Pentil, Ciclopropyl, Ciclobutil, Ciclopentil, Ciclohexil, Fenil, Indanil, Tetrahidronaphtil, Acridinil, Azocinil, Benzimidazolil, Benzofuranil, Benzothiofuranil, Benzothiofenil, Benzoxazolil, Benzthiazolil .. Oxadiazolyl, 1,2,3-Oxadiazolyl, 1,2,4-Oxadiazolyl; 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, oxetanyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxatinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, purinyl, pyranyl pyrazinyl ..., thienooxazolyl, thienoimidazolyl, thiofenilo, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, oxetanil, azetidinyl or xanthenyl; die durch q Instanzen von R ersetzt werdenC.

In certain embodiments, R1Ciclopropylo, Ciclobutilo, Ciclopentilo, Ciclohexilo, Fenilo, Furanilo, Furazanilo, Imidazolidinilo, Imidazolinilo, Imidazolilo, Isothiazolilo, Isoxazolilo, Morfolinilo, Oxadiazolilo, 1,2,3-Oxadiazolilo, 1,2,4-Oxadiazolilo; 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, oxetanyl, pyrimidinyl, piperazinyl, piperidinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, Pyrrolinyl 2H-pyrrolyl, pyrrolyl, tetrahydrofuranyl, tetrahydropyranyl, thiazolyl, thienyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, oxetanyl , Azetidinilo or Xantenilo; die durch q Instanzen von R ersetzt werdenC.

In certain embodiments, R1phenyl, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl; 2H-pyrrolyl, pyrrolyl, thiazolyl, thienyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl or 1,2,5-triazolyl, 1,3,4-triazolyl; die durch q Instanzen von R ersetzt werdenC.

In certain embodiments, R1is phenyl, pyrazolyl or pyridinyl; which are replaced by q instances of RC.

In certain embodiments, R1is phenyl, pyrazolyl or pyridinyl; which are replaced by q instances of RC; where each RCindependently halogen, -CN, -OR, -S(O)2R, -C(O)NR2, or any instance of RCindependently, an optionally substituted group selected from C1-6aliphatic; a 5-10 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-12 membered saturated or partially unsaturated spirocyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or two R'sCThe groups, together with the atoms to which they are attached, form a 5-6 membered fused or bridged spiroaryl ring, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a saturated or partially unsaturated monocyclic heterocyclic ring of 3-7 membered containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur; where or every instance of RCis optionally replaced independently by R and RD.

In certain embodiments, R1Y

In certain embodiments, R1Y

where each instance of RCindependently halogen, -CN, -OR, -S(O)2R, -S(O)NO2, -C(O)NO2, an optionally substituted group selected from C1-6aliphatic; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; or a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; which are replaced by r instances of R and s instances of RD.

In certain embodiments, R1Y

where each instance of R is independently -Me, -Et, -CH2N(CH3)2, -CN, -CH2CN, -F, -OMe, -S(O)2yo, -ch2SO)2Michigan

In certain embodiments, R1Y

In some embodiments, R1is selected from those shown in Table 1 below.

In some embodiments, each R2is independent H

In some embodiments, each R2is independent R2is selected from C1-6aliphatic; phenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; which are replaced by r instances of RD.

In some embodiments, each R2is selected independently of C2-6aliphatic; phenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; which are replaced by r instances of RD.

In some embodiments, each R2is independent C2-6aliphatically replaced by r instances of RD.

In some embodiments, each R2phenyl independently substituted by r instances of RD.

In some embodiments, each R2is independently a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring substituted by r occurrences of RD.

In some embodiments, each R2independently a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; instances of R replaced by rD.

In some embodiments, each R2independently a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; instances of R replaced by rD.

In certain embodiments, each R2independientes -H, -Et, -i-Pr, s-Bu, geradkettiges oder verzweigtes Pentil, Ciclopropil, Ciclobutil, Ciclopentil, Ciclohexil, Fenil, Indanil, Tetrahidronaftil, Acridinil, Azocinil, Benzimidazolil, Benzofuranil, Benzothiofuranil, Benzothiofenil, Benzoxazolil ist , Benztiazolil, Benztriazolil, Benztetrazolil, Benzisoxazolil, Benzisothiazolil, Benzimidazolinil, Carbazolil, NH-Carbazolil, Carbolinil, Cromanil, Chromenil, Cinolinil, Decahidroquinolinil, 2H,6H-1,5,2-Ditiazinil, Dihydrofuro[2,3-b]tetrahydrofuran, Furanil, Furazanil, Imidazolidinil, Imidazolinil, Imidazolil, 1H-Indazolil, Indolenil, Indolinil, Indolizinil, Indolil, 3H-Indolil, Isoindolinil, Isoindolenil, Isobenzofuranil, Isocromanil, Isoindazolil, Isoindolinil, Isoindolil, Isoquinolinil, Isothihydrofixazolidil, Isothiisofixazolidil, Octaquinolinil oxadiazolilo, 1,2,3-oxadiazolilo, 1,2,4-oxadiazolilo; 1,2,5-oxadiazolilo, 1,3,4-oxadiazolilo, oxazolidinilo, oxazolilo, oxazolidinilo, oxetanilo, pirimidinilo, fenantridinilo, fenantrolinilo, fenazinilo, fenotiazinilo, fenoxatiinilo, fenoxazinilo, ftalazinilo, piperazinilo, piperidinilo, pteridinilo, purinilo, piranilo pirazinilo ..., tienooxazolil, tienoimidazolil, tiofenilo, triazinil, 1,2,3-triazolil, 1,2,4-triazolil, 1,2,5-triazolil, 1,3,4-triazolil, oxetanil, azetidinil o xantenil; die durch r Instanzen von R ersetzt werdenD.

In certain embodiments, each R2unfähgäng voneinander -H, -Et, -i-Pr, s-Bu, geradkettiges oder verzweigtes Pentyl, Ciclopropyl, Ciclobutil, Ciclopentil, Ciclohexyl, Fenil, Furanil, Furazanil, Imidazolidinil, Imidazolinil, Imidazolil, Isotiazolil, Isoxazolil, Morpholinil, Oxadiazolil ist , 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl; 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, oxetanyl, pyrimidinyl, piperazinyl, piperidinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, Pyrrolinyl 2H-pyrrolyl, pyrrolyl, tetrahydrofuranyl, tetrahydropyranyl, thiazolyl, thienyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, oxetanyl , Azetidinilo or Xantenilo; detrás de jede Instance von RDis independently oxo, halogen, -CN, -NO2, —O, —SR, —NO2, -SO)2R, -S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(O)NR2, -N(R)C(NR)NR2, -N(R)NR2, —N(R)S(O)2NR2, —N(R)S(O)2R, -N=S(O)R2, -S(NR)(O)R, -N(R)S(O)R, -N(R)CN, -P(O)(R)NR2, -P(O)(R)OR o -P(O)R2; or any instance of RDindependently, an optionally substituted group selected from C1-6aliphatic; phenyl; naphthalenyl; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; and a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or two R'sDThe groups, together with the atoms to which they are attached, form a 5-6 membered fused or bridged spiroaryl ring, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a saturated or partially unsaturated monocyclic heterocyclic ring of 3-7 membered containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur; and a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; where each instance of RDis optionally replaced independently by u instances of R.

In certain embodiments, each R2is independently -H, -Et, -i-Pr, s-Bu, straight or branched chain pentyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, oxetanyl, piperazinyl, piperidinyl, pyrrolidinyl, tetrahydropyranyl, azetidinyl; which are replaced by r instances of RD.

In certain embodiments, each R2is independently -H, -Et, -i-Pr, s-Bu, straight or branched chain pentyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, oxetanyl, piperazinyl, piperidinyl, pyrrolidinyl, tetrahydropyranyl, azetidinyl; each of which is replaced by each of the r instances of RD; where each RDindependent Oxo, halogen, -CN, -OR, -C(O)R, -C(O)OR, -C(O)NR ist2, -N(R)C(O)R; or any instance of RDindependently, an optionally substituted group selected from C1-6aliphatic; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; and a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or two R'sDThe groups, together with the atoms to which they are attached, form a 5-6 membered fused or bridged spiroaryl ring, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a saturated or partially unsaturated monocyclic heterocyclic ring of 3-7 membered containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur; and a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; where each instance of RDis optionally replaced independently by u instances of R.

In some embodiments, each R2independently a 6-11 membered fused, partially unsaturated or fused unsaturated 6-11 membered spiro ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; which are replaced by r instances of RD.

In certain embodiments, each R2is independently a 7 to 10 membered fused bicyclic ring containing 1 to 3 nitrogen atoms; each of which is replaced by each of the r instances of RD.

In certain embodiments, each R2is independently a 9-membered fused bicyclic ring containing 1-3 nitrogen atoms; each of which is replaced by each of the r instances of RD; where each RDis independently halogen, -CN, -OR, -C(O)NR2, —NR2; or any instance of RDindependently, an optionally substituted group selected from C1-6aliphatic; phenyl; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; and a 6-11 membered fused, bridged or bicyclic saturated or partially unsaturated spiroheterocyclic ring containing 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; where each instance of RDis optionally replaced independently by u instances of R.

In certain embodiments, each R2is independent -H,

In certain embodiments, each R2is autonomous

In certain embodiments, each R2is autonomous

In certain embodiments, each R2is autonomous

In certain embodiments, each R2along with your rDThe substitute is independent.

In certain embodiments, each R2is independent together with its RP substituents

In some embodiments, each R2is selected independently of those shown in Table 1 below.

In certain embodiments, each R3is -H independent.

In certain embodiments, each R3is independent -C1-6aliphatic o -phenyl; which are replaced by s instances of Rmi; or any R3is independently -CN, halogen, -OR, -SR, -NR2, -SO)2R, —S(O)(NR)R, —S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)R, -N(R)C(O)OR, -N(R)C(O)NR2, -N(R)C(NR)NR2, —N(R)S(O)2NR2, o -N(R)S(O)2r

In certain embodiments, each R3is independent -H,C1-6aliphatic o -phenyl; which are replaced by s instances of Rmi; or any R3is independently -CN, halogen, -OR, -SR, -NR2, -SO)2R, —S(O)(NR)R, —S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)R, -N(R)C(O)OR, -N(R)C(O)NR2, -N(R)C(NR)NR2, —N(R)S(O)2NR2, o -N(R)S(O)2r

In certain embodiments, each R3is independent -H,C1-6aliphatic o -phenyl; which are replaced by s instances of Rmi; or any R3is independently -CN, halogen, -OR, -NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)R, -N(R)C(O)OR, -N(R)C(O)NR2, -N(R)C(NR)NR2, —N(R)S(O)2NR2, o -N(R)S(O)2r

In certain embodiments, each R3is independent -H,C1-6aliphatic o -phenyl; which are replaced by s instances of Rmi; or any R3is independently -CN, halogen, -OR, -NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)R, -N(R)C(O)OR, -N(R)C(O)NR2, o -N(R)C(NR)NR2.

In certain embodiments, each R3is independent -H,C1-6aliphatic o -phenyl; which are replaced by s instances of Rmi; or any R3is independently -CN, halogen, -C(O)R, -C(O)OR, -C(O)NR2, o -C(O)N(R)OR.

In certain embodiments, each R3is independently -H, -Me, -Et, or -phenyl; which are replaced by s instances of Rmi; or any R3is independently -CN, halogen or -C(O)OR.

In some embodiments, each R3independently -H, -Me, -Et, -CN, -Br,

In some embodiments, each R3is selected independently of those shown in Table 1 below.

In some embodiments, R4it's H.

In some embodiments, R4is C1-6aliphatic, which is replaced by t instances of R.

In some embodiments, R4is selected from those shown in Table 1 below.

In some implementations, each instance of RC, RD, Rmiy R. S.Fis independently oxo, halogen, -CN, -NO2, —O, —SR, —NO2, -SO)2R, -S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(O)NR2, -N(R)C(NR)NR2, -N(R)NR2, —N(R)S(O)2NR2, —N(R)S(O)2R, -N=S(O)R2, -S(NR)(O)R, -N(R)S(O)R, -N(R)CN, -P(O)(R)NR2, -P(O)(R)OR o -P(O)R2; or any instance of RC, RD, Rmiy R. S.F, independently an optionally substituted group selected from C1-6aliphatic; phenyl; naphthalenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; or a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.

In some implementations, each instance of RC, RD, Rmiy R. S.FOxo, halogen, -CN, -NO ist2, —O, —SR, —NO2, -SO)2R, -S(O)2NR2, -S(O)R, -S(O)NR2, -C(O)R, -C(O)OR, -C(O)NR2, -C(O)N(R)OR, -OC(O)R, -OC(O)NR2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(O)NR2, -N(R)C(NR)NR2, -N(R)NR2, —N(R)S(O)2NR2, —N(R)S(O)2R, -N=S(O)R2, -S(NR)(O)R, -N(R)S(O)R, -N(R)CN, -P(O)(R)NR2, -P(O)(R)OR o -P(O)R2.

In some implementations, each instance of RC, RD, Rmiy R. S.Fis an optionally substituted group selected from C1-6aliphatic; phenyl; naphthalenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; or a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.

In some implementations, each instance of RC, RD, Rmiy R. S.Fis selected from those shown in Table 1 below.

As generally defined above, each R is independently hydrogen or an optionally substituted group selected from C1-6aliphatic; phenyl; naphthalenyl; a 3 to 7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring containing 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a saturated or partially unsaturated 6-10 membered spirocyclic ring containing 0-3 independently selected nitrogen, oxygen and sulfur heteroatoms; a 6-11 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; either:

two R groups on the same nitrogen are taken together with the nitrogen to form a saturated, partially unsaturated, or optionally substituted 4-7 membered heteroaryl monocyclic ring having, in addition to nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur ; an 8 to 10 membered bicyclic heteroaryl ring containing 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring containing 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.

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

As generally defined above, m is 1, 2, or 3. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3.

In some embodiments, m is selected from those shown in Table 1 below.

As generally defined above, n is 0, 1, or 2. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2.

In some embodiments, n is selected from those shown in Table 1 below.

As generally defined above, q is 0, 1, 2, 3, or 4. In some embodiments, q is 0. In some embodiments, q is 1, 2, 3, or 4. In some embodiments, q is 1. In some embodiments, q 2. In some embodiments, q is 3. In some embodiments, q is 4.

In some embodiments, q is selected from those shown in Table 1 below.

As generally defined above, r is 0, 1, 2, 3, or 4. In some embodiments, r is 0. In some embodiments, r is 1, 2, 3, or 4. In some embodiments, r is 1. In some realizations, r 2. In some realizations, r is 3. In some realizations, r is 4.

In some embodiments, r is selected from those shown in Table 1 below.

As generally defined above, s is 0, 1, 2, 3, or 4. In some embodiments, s is 0. In some embodiments, s is 1, 2, 3, or 4. In some embodiments, s is 1. In some realizations, s 2. In some realizations, s is 3. In some realizations, s is 4.

In some embodiments, s is selected from those shown in Table 1 below.

As generally defined above, t is 0, 1, 2, 3, or 4. In some embodiments, t is 0. In some embodiments, t is 1, 2, 3, or 4. In some embodiments, t is 1. In some embodiments, t 2. In some embodiments, t is 3. In some embodiments, t is 4.

In some embodiments, t is selected from those shown in Table 1 below.

As generally defined above, u is 0, 1, 2, 3, or 4. In some embodiments, u is 0. In some embodiments, u is 1, 2, 3, or 4. In some embodiments, u is 1. In some embodiments, u 2. In some embodiments, u is 3. In some embodiments, u is 4.

In some embodiments, u is selected from those shown in Table 1 below.

In some embodiments, the present invention provides a compound of formula II, III, IV, V, VI, VII, VIII, IX, X, or XI:

or a pharmaceutically acceptable salt thereof, wherein each of L, R1, R2, R3, R4, m and n are as defined above and described in the embodiments herein, both individually and in combination.

In some embodiments, the present invention provides a compound of formula II-a, III-a, IV-a, V-a, VI-a, VII-a, VIII-a, IX-a, X-a, or XI-a:

or a pharmaceutically acceptable salt thereof, wherein each of R1, R2, R3, R4, m and n are as defined above and described in the embodiments herein, both individually and in combination.

In some embodiments, the present invention provides a compound of formula II-b, III-b, IV-b, V-b, VI-b, VII-b, VIII-b, IX-b, X-b, or XI-b:

or a pharmaceutically acceptable salt thereof, wherein each of L, R1, R2, R3, R4ym is as defined above and described in embodiments herein, both individually and in combination.

In some embodiments, the present invention provides a compound of formula II-c, III-c, IV-c, V-c, VI-c, VII-c, VIII-c, IX-c, X-c, or XI-c:

or a pharmaceutically acceptable salt thereof, wherein each of L, R1, R2, R3, R4ym is as defined above and described in embodiments herein, both individually and in combination.

Examples of compounds of the invention are listed in Table 1 below.

TABLA 1 selected connections STRUCTURE STRUCTURE I-1 I-2 I-3 I-4 I-5 I-6 I-7 I-8 I-9 I 10 I-11 I-12 I-13 I-14 I-15 I-16 I-17 I-18 I-19 I-20 I-21 I-22 I-23 I-24 I-25 I-26 I-27 I-28 I-29 I-30 I-31 I-32 I-33 I-34 I-35 I-36 I-37 I-38 I-39 I-40 I-41 I-42 I-43 I-44 I-45 I-46 I-47 I-48 I-49 I-50 I-51 I-52 I-53 I-54 I-55 I-56 I-57 I-58 I-59 I-60 I-61 I-62 I-63 I-64 I-65 I-66 I-67 I-68 I-69 I-70 I-71 I-72 I-73 I-74 I-75 I-76 I-77 I-78 I-79 I-80 I-81 I-82 I-83 I-84 I-85 I-86 I-87 I-88 I-89 I-90 I-91 I-92 I-93 I-94 I-95 I-96 I-97 I-98 I-99 I-100 I-101 I-102 I-103 I-104 I-105 I-106 I-107 I-108 I-109 I-110 I-111 I-112 I-113 I-114 I-115 I-116 I-117 I-118 I-119 I-120 I-121 I-122 I-123 I-124 I-125 I-126 I-127 I-128 I-129 I-130 I-131 I-132 I-133 I-134 I-135 I-136 I-137 I-138 I-139 I-140 I-141 I-142 I-143 I-144 I-145 I-146 I-147 I-148 I-149 I-150 I-151 I-152 I-153 I-154 I-155 I-156 I-157 I-158 I-159 I-160 I-161 I-162 I-163 I-164 I-165 I-166 I-167 I-168 I-169 I-170 I-171 I-172 I-173 I-174 I-175 I-176 I-177 I-178 I-179 I-180 I-181 I-182 I-183 I-184 I-185 I-186 I-187 I-188 I-189 I-190 I-191 I-192 I-193 I-194 I-195 I-196 I-197 I-198 I-199 I-200 I-201 I-202 I-203 I-204 I-205 I-206 I-207 I-208 I-209 I-210 I-211 I-212 I-213 I-214 I-215 I-216 I-217 I-218 I-219 I-220 I-221 I-222 I-223 I-224 I-225 I-226 I-227 I-228 I-229 I-230 I-231 I-232 I-233 I-234 I-235 I-236 I-237 I-238 I-239 I-240 I-241 I-242 I-243 I-244 I-245 I-246 I-247 I-248 I-249 I-250 I-251 I-252 I-253 I-254 I-255 I-256 I-257 I-258 I-259 I-260 I-261 I-262 I-263 I-264 I-265 I-266 I-267 I-268 I-269 I-270 I-271 I-272 I-273 I-274 arbitrarily assigned arbitrarily assigned I-275 I-276 I-277 I-278 I-279 I-280 I-281 I-282 I-283 I-284 I-285 I-286 I-287 I-288 I-289 I-290 I-291 I-292 I-293 I-294 I-295 I-296 I-297 I-298 I-299 I-300 I-301 I-302 I-303 I-304 I-305 I-306 I-307 I-308 I-309 I-310 I-311 I-312 I-313 I-314 I-315 I-316 I-317 I-318 I-319 I-320 I-321 I-322 I-323 I-324 I-325 I-326 I-327 I-328 I-329 I-330 I-331 I-332 I-333 I-334 I-335 I-336 I-337 I-338 I-339 I-340 I-341 I-342 I-343 I-344 I-345 I-346 I-347 I-348 I-349 I-350 I-351 I-352 I-353 I-354 I-355 I-356 I-357 I-358 I-359 I-360 I-361 I-362 I-363 I-364 I-365 I-366 I-367 I-368 I-369 I-370

In some embodiments, the present invention provides a compound listed in Table 1 above, or a pharmaceutically acceptable salt thereof. In some embodiments, the present invention provides a compound shown in Table 1 above. In some embodiments, the present invention provides a pharmaceutical composition comprising a compound listed in Table 1 above, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier, excipient, or diluent.

In some embodiments, the present invention provides a compound of formula I as defined above, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula I as defined above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. B. Acceptable adjuvant or vehicle for use as a medicament.

In some embodiments, the invention also provides compounds of formula I described herein or pharmaceutical compositions described herein for use in a method for inhibiting HPK1 as described herein, in a method for enhancing an immune response in a subject who you need it as described in this document. herein, provided and/or in a method for treating an HPK1-dependent disorder as described herein.

In some embodiments, the invention also provides compounds of formula I as described herein or pharmaceutical compositions as described herein for use in a method for inhibiting HPK1 as described herein.

In some embodiments, the invention also provides compounds of formula I described herein or pharmaceutical compositions described herein for use in a method for enhancing an immune response in a subject in need thereof, as described herein.

In some embodiments, the invention also provides compounds of formula I as described herein or pharmaceutical compositions as described herein for use in a method of treating an HPK1-dependent disorder as described herein.

In some embodiments, the invention also provides the use of a compound of formula I described herein or a pharmaceutical composition described herein for the manufacture of a medicament for inhibiting HPK1, a medicament for increasing an immune response in a subject that does so. need and/or or Drug used to treat an HPK1-dependent disorder.

In some embodiments, the invention also provides the use of a compound of formula I as described herein or a pharmaceutical composition as described herein for the manufacture of a medicament for inhibiting HPK1.

In some embodiments, the invention also provides the use of a compound of formula I as described herein or a pharmaceutical composition as described herein for the manufacture of a medicament for enhancing an immune response in an individual in need thereof.

In some embodiments, the invention also provides the use of a compound of formula I as described herein or a pharmaceutical composition as described herein for the manufacture of a medicament for the treatment of an HPK1-dependent disorder.

In some embodiments, the invention also provides for the use of compounds of formula I described herein or pharmaceutical compositions described herein in a method for inhibiting HPK1 as described herein, in a method for enhancing an immune response in a subject. you need the same thing that is provided. herein and/or in a method of treating an HPK1-dependent disorder as described herein.

In some embodiments, the invention also provides the use of compounds of formula I described herein or pharmaceutical compositions described herein in a method for inhibiting HPK1 as described herein.

In some embodiments, the invention also provides the use of compounds of formula I described herein or pharmaceutical compositions described herein in a method for enhancing an immune response in a subject in need thereof, as described herein.

In some embodiments, the invention also provides the use of compounds of formula I described herein or pharmaceutical compositions described herein in a method of treating an HPK1-dependent disorder as described herein.

As generally defined above, any hydrogen attached to carbon may optionally and independently be replaced by deuterium.

In some embodiments, a hydrogen attached to carbon is replaced with deuterium.

4. General Methods for Providing Composite Gifts

The compounds of this invention can generally be prepared or isolated by synthetic and/or semi-synthetic methods known to those skilled in the art for analogous compounds and by methods described in detail in the examples presented herein.

5. Uses, Formulation and Administration

Pharmaceutically acceptable compositions.

In another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, excipient or vehicle. The amount of compound in the compositions of this invention is such that it is effective to inhibit HPK1 or a mutant thereof in a biological sample or in a patient. In certain embodiments, the amount of compound in the compositions of this invention is such that it is effective to inhibit HPK1 or a mutant thereof in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of said composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient.

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

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

A "pharmaceutically acceptable derivative" means any salt, ester, salt of an ester, or other non-toxic derivative of a compound of this invention which, upon administration to a recipient, is capable of directly or indirectly providing a compound of this invention or a inhibitor of the active metabolite or residue thereof.

As used herein, the term "active inhibitory metabolite or residue thereof" means that a metabolite or residue thereof is also an inhibitor of HPK1 or a mutant thereof.

The subject matter described herein includes prodrugs, metabolites, derivatives and pharmaceutically acceptable salts of the compounds of the invention. Metabolites include compounds produced by a process comprising contacting a compound of the invention with a mammal for a period of time sufficient to produce a metabolite therefrom. When the compound of the invention is a base, the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, by treating the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid. nitric acid, methanesulfonic acid, phosphoric acid and the like, or with an organic acid such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidylic acid such as glucuronic acid or galacturonic acid, an alpha hydroxy acid such as citric acid or tartaric acid, an amino acid such as aspartic acid or glutamic acid, an aromatic acid such as benzoic acid or cinnamic acid, a sulfonic acid such as p-toluenesulfonic acid or ethanesulfonic acid, or the like. When the compound of the invention is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, by treating the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), a hydroxide or alkali metal alkaline earth metal hydroxide or the like. Illustrative examples of suitable salts include, but are not limited to, organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary and tertiary amines and cyclic amines such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, salts derived from calcium , potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.

A compound of the invention may be in "prodrug" form, which includes compounds with moieties that can be metabolized in vivo. In general, prodrugs are metabolized in vivo to active drugs via esterases or other mechanisms. Examples of prodrugs and their uses are well known in the art (see, eg, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66:1-19). Prodrugs can be prepared in situ during final isolation and purification of the compounds or by separately reacting the purified compound in its free acid or hydroxyl form with a suitable esterifying agent. Hydroxyl groups can be converted to esters by treatment with a carboxylic acid. Examples of prodrug moieties include branched or unsubstituted lower alkyl ester moieties (eg, propionic acid esters), lower alkenyl esters, di-lower alkylamino lower alkyl ester (eg, ester dimethylaminoethyl), acylamino ester, lower alkyl (e.g. acetyloxymethyl ester), acyloxy lower alkyl esters (e.g. pivaloyloxymethyl esters), aryl esters (phenyl esters), aryl lower alkyl esters ( eg, benzyl esters), aryl esters, and substituted aryl and lower alkyl (eg, with methyl, halogen, or methoxy substituents), amides, lower alkyl amides, di-lower alkyl amides, and hydroxy amides. Also included are prodrugs that are converted to active forms in vivo by other mechanisms. In some aspects, the compounds of the invention are prodrugs of any of the formulas set forth herein.

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

Any bland fixed oil can be used for this purpose, including synthetic monoglycerides or diglycerides. Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are pharmaceutically acceptable natural oils such as olive oil or castor oil, particularly in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcoholic diluent or dispersant, such as carboxymethyl cellulose or similar dispersants commonly used in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants such as Tweens, Spans, and other emulsifiers or bioavailability enhancers commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for formulation purposes.

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

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

The pharmaceutically acceptable compositions of this invention may also be administered topically, particularly when the treatment objective involves areas or organs readily accessible by topical application, including eye, skin, or lower intestinal tract disorders. Appropriate topical formulations are easily prepared for each of these areas or organs.

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

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

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

The pharmaceutically acceptable compositions of this invention may also be administered by nasal spray or inhalation. Said compositions are prepared according to techniques well known in the field of pharmaceutical formulation and may be prepared as saline solutions using benzyl alcohol or other suitable preservatives, absorption enhancers to improve bioavailability, fluorocarbons and/or other conventional solubilizing or dispersing agents.

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

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

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

Uses of pharmaceutically acceptable compounds and compositions

The compounds and compositions described herein are generally useful for inhibiting the kinase activity of one or more enzymes. In some embodiments, the kinase inhibited by the compounds and methods of the invention is HPK1.

The compounds described herein find use for inhibiting the activity of the HPK1 enzyme. HPK1 is a member of the Ste20-related serine/threonine kinase subfamily of germinal center kinases. HPK1 functions like MAP4K by phosphorylating and activating MAP3K proteins, including MEKK1, MLK3, and TAK1, resulting in activation of MAPK Jnk.

In one embodiment, the subject matter described herein relates to a method of inhibiting HPK1, the method comprising contacting HPK1 with an effective amount of a compound of the invention or a pharmaceutical composition as described herein.

In certain embodiments, the subject matter described herein relates to a method of enhancing an immune response in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of the invention or a pharmaceutical composition described in the subject present document. In certain aspects of this embodiment, the T cells in the subject have at least one of increased initiation, increased activation, increased migration, increased proliferation, increased survival, and increased cytolytic activity relative to prior to administration of the compound or pharmaceutical composition. In certain aspects of this embodiment, T cell activation is evidenced by a higher frequency of γ-IFN+CD8 T cells or higher levels of IL-2 or granzyme B production by T cells relative to before. administration of the Compound or composition. pharmacist. In certain aspects of this embodiment, the number of T cells is increased relative to prior to administration of the compound or pharmaceutical composition. In certain aspects of this embodiment, the T cell is an antigen-specific CD8 T cell. In certain aspects of this embodiment, the antigen-presenting cells in the subject exhibit enhanced maturation and activation relative to prior to administration of the compound or pharmaceutical composition. In certain aspects of this embodiment, the antigen presenting cells are dendritic cells. In certain aspects of this modality, antigen-presenting cell maturation is characterized by an increased abundance of CD83+ dendritic cells. In certain aspects of this modality, activation of antigen presenting cells is characterized by increased expression of CD80 and CD86 on dendritic cells.

The compounds described herein directly bind to HPK1 and inhibit its kinase activity. In some embodiments, the compounds described herein reduce, inhibit, or otherwise reduce HPK1-mediated phosphorylation of SLP76 and/or Gads.

The compounds described herein may or may not be a specific antagonist of HPK1. A specific HPK1 antagonist reduces the biological activity of HPK1 by an amount that is statistically greater than the inhibitory effect of the antagonist on any other protein (eg, other serine/threonine kinases). In certain embodiments, the compounds described herein specifically inhibit the serine/threonine kinase activity of HPK1. In some of these modalities, the CI50of HPK1 antagonist to HPK1 is approximately 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 0.1%, 0.01%, 0.001% or less than CI50the HPK1 antagonist to another serine/threonine kinase or type of kinase (eg, tyrosine kinase).

The compounds described herein can be used in a method for inhibiting HPK1. Such methods include contacting HPK1 with an effective amount of a compound described herein. By "contact" is meant bringing the compound close enough to an isolated HPK1 cell or enzyme that expresses HPK1 (eg, T cell, B cell, dendritic cell) so that the compound can bind to HPK1 and inhibit its activity. The compound can be contacted with HPK1 in vitro or in vivo by administering the compound to a subject.

Any method known in the art to measure HPK1 kinase activity can be used to determine if HPK1 has been inhibited, including in vitro kinase assays, immunoblotting with antibodies specific for phosphorylated HPK1 targets, such as SLP76 and Gads , or the measurement of a subsequent biological effect of HPK1. Kinase activity, such as recruitment of 14-3-3 proteins to SLP7 and phosphorylated Gads, release of the SLP76-Gads-14-3-3 complex from LAT-containing microclusters, or activation of T or B cells.

The compounds described herein can be used to treat an HPK1-dependent disorder. As used herein, an "HPK1-dependent disorder" is a pathological condition in which HPK1 activity is required to initiate or maintain the pathological condition. In some embodiments, the HPK1-dependent disease is cancer.

The compounds described herein also find use in enhancing an immune response in a subject in need thereof. Such methods include the administration of an effective amount of a compound of the invention.

As used herein, "increased immune response" refers to an increase in any immunogenic response to an antigen. Non-limiting examples of improvements in an immunogenic response to an antigen include increased dendritic cell maturation or migration, increased activation of T cells (eg, CD4 T cells, CD8 T cells), increased T cells (eg. , CD4 T cells, CD8 T cells cell proliferation), increased B cell proliferation, increased T cell and/or B cell survival, enhanced antigen presentation by antigen presenting cells (p. a la prostaglandin E2-induced immunosuppression and increased activation and/or cytolytic activity of CD8 T cells.

In some embodiments, CD8 T cells in the subject have enhanced initiating, activating, proliferative, and/or cytolytic activity compared to prior to administration of the compound of the invention or a pharmaceutically acceptable salt, prodrug, metabolite, or derivative thereof. In some embodiments, priming of CD8 T cells is characterized by increased CD44 expression and/or increased cytolytic activity on CD8 T cells. In some embodiments, CD8 T cell activation is characterized by increased abundance of γ-IFN+CD8 T cells. In some embodiments, the CD8 T cell is an antigen-specific T cell.

In some embodiments, antigen-presenting cells in the subject exhibit enhanced maturation and activation compared to prior to administration of the compound of the invention or a pharmaceutically acceptable salt, prodrug, metabolite, or derivative thereof. In some embodiments, the antigen presenting cells are dendritic cells. In some embodiments, antigen-presenting cell maturation is characterized by an increased abundance of CD83.+dendritic cells. In some embodiments, activation of antigen presenting cells is characterized by increased expression of CD80 and CD86 on dendritic cells.

In some embodiments, the serum levels of the cytokine IL-10 and/or chemokine IL-8, a human homologue of murine KC, in the subject are reduced as compared to prior to administration of the compound of formula I or Ia or a salt pharmaceutically acceptable, prodrug, metabolite or derivative thereof.

TCR activation leads to activation of HPK1, which acts as a negative regulator of the TCR-induced AP-1 pathway. HPK1 is thought to downregulate T cell activation by reducing the persistence of signaling microclusters through phosphorylation of SLP76 at Ser376 (Di Bartolo et al. (2007) JEM 204: 681-691) and Gads at Thr254, leading to the recruitment of 14-3-3 protein results that bind to SLP76 and phosphorylated Gads and release the SLP76-Gads-14-3-3 complex from LAT-containing microclusters, resulting in dysfunction of T cells, including anergy and fatigue (Lasserre et al (2011) J Cell Biol. 195(5):839-853).

In some embodiments, administration of a compound of the invention, or a pharmaceutically acceptable salt, prodrug, metabolite, or derivative thereof to a subject results in an increase in T cell function.

Accordingly, the compounds of the invention described herein, or their pharmaceutically acceptable salts, prodrugs, metabolites, or derivatives, are useful in the treatment of T-cell dysfunction disorders. A "T-cell dysfunction" is a disorder or condition of T cells characterized by a decreased response to antigenic stimulation. In a particular embodiment, a T-cell dysfunction disorder is a disorder specifically associated with increased HPK1 kinase activity. In another embodiment, a T cell dysfunction is one in which the T cells are anergic or have a reduced ability to secrete cytokines, proliferate, or perform cytolytic activity. In a specific aspect, reduced responsiveness results in ineffective control of a pathogen or tumor expressing an immunogen. Examples of T cell dysfunction disorders characterized by T cell dysfunction include unresolved acute infections, chronic infections, and tumor immunity.

Therefore, the compounds described herein can be used to treat conditions where increased immunogenicity is desired, such as. B. an increase in the immunogenicity of the tumor for the treatment of cancer.

The term "dysfunction" in the context of immune dysfunction refers to a condition of reduced immune response to antigenic stimulation. The term encompasses the common elements of exhaustion and/or anergy, where antigen recognition may occur, but the subsequent immune response is ineffective in controlling infection or tumor growth.

The term "dysfunctional", as used herein, also includes refractory or unresponsive to antigen recognition, specifically an impaired ability to translate antigen recognition into downstream T cell effector functions, eg, B proliferation. ., production of cytokines (eg IFN) and/or target cells. murder.

The term "anergy" refers to the state of unresponsiveness to antigenic stimulation resulting from incomplete or inappropriate signals emitted by the T-cell receptor (for example, increased intracellular Ca+2in the absence of ras activation). T cell anergy may also be due to antigen stimulation without costimulation, rendering the cell itself recalcitrant in the context of costimulation to subsequent antigen activation. The unresponsive state can often be reversed in the presence of interleukin-2. Anergic T cells do not undergo clonal expansion or acquire effector functions.

The term "fatigue" refers to T cell depletion as a state of T cell dysfunction arising from sustained TCR signaling that occurs during many chronic infections and cancers. It differs from anergy in that it does not arise from incomplete or faulty signaling, but from sustained signalling. It is defined by impaired effector function, persistent expression of inhibitory receptors, and a transcriptional state distinct from that of functional or memory effector T cells. Exhaustion prevents optimal control of infections and tumors. Fatigue can result from both extrinsic negative regulatory pathways (eg, immunoregulatory cytokines) and cellular (costimulatory) negative regulatory pathways (PD-1, B7-H3, B7-H4, etc.).

"Immunogenicity" refers to the ability of a particular substance to induce an immune response. Tumors are immunogenic and increasing the immunogenicity of the tumor helps the immune response to kill the tumor cells.

"Enhancing T cell function" means inducing, causing, or stimulating a T cell to have sustained or increased biological function, or to regenerate or reactivate depleted or inactive T cells. Examples of improvement in T cell function include: increased secretion of cytokines (eg, interferon γ, IL-2, IL-12, and TNFα), increased proliferation, increased antigen responsiveness (eg, clearance of viruses, pathogens, or tumors) compared to such pre-procedure levels and increased production of effector granules by CD8 T cells, such as granzyme B. In one embodiment, the level of increase is at least 50%, alternately 60%, 70%, 80%, 90%, 100%. , 120%, 150%, 200%. The way to measure this improvement is known to those skilled in the art.

"Tumor immunity" refers to the process by which tumors evade immune recognition and elimination. As a therapeutic concept, tumor immunity is "treated" when this bias is weakened and tumors are recognized and attacked by the immune system. Examples of tumor recognition include tumor fixation, tumor reduction, and tumor removal.

The present disclosure provides methods for modulating (eg, inhibiting) HPK1 activity, the method comprising administering to a patient a compound provided herein or a pharmaceutically acceptable salt thereof.

In one aspect, there is provided herein a method of treating cancer in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention or a pharmaceutically acceptable salt, prodrug, metabolite or derivative thereof. .

In the methods described herein, a compound of the invention or a pharmaceutical composition thereof is administered to a subject having cancer.

In certain embodiments, the subject matter described herein relates to a method of treating an HPK1-dependent disorder, the method comprising administering to a patient in need thereof an effective amount of a compound of the invention or a pharmaceutical composition as described in the present document. In certain aspects of this modality, the HPK1-dependent disorder is a cancer. In certain aspects of this embodiment, the cancer comprises at least one cancer selected from the group consisting of colon cancer, melanoma, non-small cell lung cancer, ovarian cancer, breast cancer, pancreatic cancer, hematologic malignancy, and carcinoma. of small cells. . In certain aspects of this embodiment, the cancer has increased levels of T-cell infiltration. In certain aspects of this embodiment, cancer cells in the subject selectively display increased expression of MHC class I antigen relative to prior to administration of the compound or composition. .

In some embodiments, the invention provides a pharmaceutical composition comprising an effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof and a pharmaceutically acceptable carrier.

In certain aspects, the invention provides a method of treating cell proliferative disorders including cancer, benign papillomatosis, gestational trophoblastic diseases, and benign neoplastic diseases such as skin tags (warts) and genital tags.

In one aspect, the invention provides a method of treating a cell proliferative disorder in an individual comprising administering to the individual a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof.

In certain embodiments, the cell proliferative disorder is cancer.

Examples of cancers treatable using the compounds of the present disclosure include, but are not limited to, bone cancer, pancreatic cancer, skin cancer, head or neck cancer, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, cancer rectal cancer, anal region cancer, gastric cancer, testicular cancer, uterine cancer, fallopian tube carcinoma, endometrial carcinoma, endometrial cancer, cervical carcinoma, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, non-Hodgkin's lymphoma, colon cancer, esophagus, small intestine cancer, cancer of the endocrine system, thyroid cancer, parathyroid cancer, adrenal gland cancer, soft tissue sarcoma, urethral cancer, penile cancer, chronic or acute leukemias, including leukemia acute myeloid, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia leukemia, childhood solid tumors, lymphocytic lymphoma, bladder cancer, renal or urethral cancer, renal pelvis cancer, central nervous system (CNS) neoplasm, primary CNS lymphoma , tumor angiogenesis, spinal axis tumor, brain stem glioma, pituitary adenoma, Kaposi's sarcoma, squamous cell carcinoma, squamous cell carcinoma, T-cell lymphoma, environmentally induced cancers, including those induced by asbestos, and combinations of these cancers.

In some embodiments, cancers treatable using the compounds of the present disclosure include solid tumors (eg, but not limited to, prostate cancer, colon cancer, esophageal cancer, endometrial cancer, ovarian cancer, uterine cancer). , kidney cancer, liver cancer). cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, head and neck cancer, thyroid cancer, glioblastoma, sarcoma, bladder cancer, etc.), blood cancers (eg, lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), DLBCL, mantle cell lymphoma, non-Hodgkin lymphoma (including recurrent follicular NHL or refractory and recurrent), Hodgkin lymphoma or multiple myeloma) and combinations of these cancers.

In certain embodiments, the cancer is brain cancer, leukemia, skin cancer, prostate cancer, thyroid cancer, colon cancer, lung cancer, or sarcoma. In another embodiment, the cancer is selected from the group consisting of glioma, glioblastoma multiforme, paraganglioma, supratentorial primordial neuroectodermal tumors, acute myeloid leukemia, myelodysplastic syndrome, chronic myeloid leukemia, melanoma, breast, prostate, thyroid, colon, lung, central chondrosarcoma. , central and periosteal chondromas, fibrosarcoma and cholangiocarcinoma.

In certain embodiments, the cancer is selected from brain and spine cancer, head and neck cancer, leukemia and blood cancer, skin cancer, reproductive system cancer, gastrointestinal system cancer, liver and bile duct cancer, cancer kidney and bladder cancer, bone cancer, lung cancer, malignant mesothelioma, sarcoma, lymphoma, glandular cancer, thyroid cancer, heart tumor, germ cell tumor, malignant neuroendocrine tumor (carcinoid), midline cancer, and cancer of origin unknown primary (cancer in which metastatic cancer is found but the original site of the cancer is unknown). In certain embodiments, the cancer is present in an adult patient; in additional embodiments, the cancer is present in a pediatric patient. In certain embodiments, the cancer is related to AIDS.

In another embodiment, the cancer is selected from brain and spinal cord cancer. In certain embodiments, the cancer is selected from the group consisting of anaplastic astrocytoma, glioblastoma, astrocytoma, and steosion euroblastoma (olfactory blastoma). In certain embodiments, the brain tumor is selected from the group consisting of astrocytic tumor (eg, pilocytic astrocytoma, subependymal giant cell astrocytoma, diffuse astrocytoma, pleomorphic xanthoastrocytoma, anaplastic astrocytoma, astrocytoma, giant cell glioblastoma, glioblastoma, secondary glioblastoma, primary adult glioblastoma and primary pediatric glioblastoma), oligodendroglial tumor (eg, oligodendroglioma and anaplastic oligodendroglioma), oligoastrocytic tumor (eg, oligoastrocytoma and anaplastic oligoastrocytoma), ependymoma (eg, myxopapillary ependymoma and anaplastic ependymoma) ; Medulloblastoma, primitive neuroectodermal tumor, schwannoma, meningioma, atypical meningioma, anaplastic meningioma, pituitary adenoma, brain stem glioma, cerebellar astrocytoma, cerebral astorcytoma/malignant glioma, hypothalamic and optic pathway glioma, and primary central nervous system lymphoma. In certain instances of these modalities, the brain tumor is selected from the group consisting of glioma, glioblastoma multiforme, paraganglioma, and supratentorial primordial neuroectodermal tumors (sPNET).

In specific embodiments, the cancer is selected from head and neck cancer, including nasopharyngeal cancer, nasal cavity and paranasal sinus cancer, hypopharyngeal cancer, oral cavity cancer (eg, squamous cell carcinoma, lymphoma, and sarcoma), lip cancer, oropharyngeal cancer, salivary gland tumors, laryngeal cancer (eg, squamous cell carcinoma of the larynx, rhabdomyosarcoma), and eye cancer or eye cancer. In certain embodiments, the ocular cancer is selected from the group consisting of intraocular melanoma and retinoblastoma.

In specific embodiments, the cancer is selected from leukemia and blood cancer. In certain embodiments, the cancer is selected from the group consisting of myeloproliferative neoplasms, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, acute myelogenous leukemia (AML), myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CMIL), myeloproliferative neoplasm (MPN), post-MPN-AML, post-MDS-AML, MDS or AML associated with del(5q) high-risk, blast phase chronic myeloid leukemia, angioimmunoblastic lymphoma, acute lymphoblastic leukemia, Langerans cell histiocytosis, hairy cell leukemia, and plasma cell neoplasms, including plasmacytomas and multiple myelomas. The leukemias mentioned here can be acute or chronic.

In specific embodiments, the cancer is selected from skin cancer. In certain embodiments, the skin cancer is selected from the group consisting of melanoma, squamous cell carcinoma, and basal cell carcinoma.

In specific embodiments, the cancer is selected from cancers of the reproductive system. In certain embodiments, the cancer is selected from the group consisting of breast cancer, cervical cancer, vaginal cancer, ovarian cancer, prostate cancer, penile cancer, and testicular cancer. In certain instances of these modalities, the cancer is a breast cancer selected from the group consisting of ductal carcinomas and phyllodes tumors. In certain instances of these modalities, the breast cancer may be male breast cancer or female breast cancer. In certain instances of these modalities, the cancer is cervical cancer selected from the group consisting of squamous cell carcinoma and adenocarcinoma. In certain instances of these embodiments, the cancer is ovarian cancer selected from the group consisting of epithelial cancer.

In specific embodiments, the cancer is selected from cancers of the gastrointestinal system. In certain embodiments, the cancer is selected from the group consisting of esophageal cancer, gastric cancer (also known as gastric cancer), gastrointestinal carcinoid tumors, pancreatic cancer, gallbladder cancer, colorectal cancer, and anal cancer. In cases of these modalities, the cancer is selected from the group consisting of esophageal squamous cell carcinoma, esophageal adenocarcinoma, gastric adenocarcinoma, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, gastric lymphoma, gastrointestinal lymphoma, pancreatic pseudopapillary solid tumor, pancreatoblastoma, islet cell tumor carcinoma, pancreatic carcinoma including acinar cell carcinoma and ductal adenocarcinoma, gallbladder adenocarcinoma, colorectal adenocarcinoma and anal squamous cell carcinoma.

In specific embodiments, the cancer is selected from liver and bile duct cancer. In certain embodiments, the cancer is liver cancer (hepatocellular carcinoma). In certain embodiments, the cancer is bile duct cancer (cholangiocarcinoma); in cases of these modalities, the bile duct cancer is selected from the group consisting of intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma.

In specific embodiments, the cancer is selected from kidney and bladder cancer. In certain embodiments, the cancer is renal cancer selected from the group consisting of renal cell carcinoma, Wilms tumors, and transitional cell carcinoma. In certain embodiments, the cancer is a bladder cancer selected from the group consisting of urethral carcinoma (a transitional cell carcinoma), squamous cell carcinoma, and adenocarcinoma.

In specific embodiments, the cancer is selected from bone cancer. In certain embodiments, the bone cancer is selected from the group consisting of osteosarcoma, malignant fibrous histiocytoma of bone, Ewing's sarcoma, and chordoma.

In specific embodiments, the cancer is selected from lung cancer. In certain embodiments, the lung cancer is selected from the group consisting of non-small cell lung cancer, small cell lung cancer, bronchial tumors, and pleuropulmonary blastoma.

In specific embodiments, the cancer is selected from malignant mesothelioma. In certain embodiments, the cancer is selected from the group consisting of epithelial and sarcomatoid mesothelioma.

In specific embodiments, the cancer is selected from sarcomas. In certain embodiments, the sarcoma is selected from the group consisting of central chondrosarcoma, central and periosteal chondroma, fibrosarcoma, clear cell tendon sheath sarcoma, and Kaposi's sarcoma.

In specific embodiments, the cancer is selected from lymphoma. In certain embodiments, the cancer is selected from the group consisting of Hodgkin lymphoma (eg, Reed-Sternberg cells), non-Hodgkin lymphoma (eg, diffuse large B-cell lymphoma, follicular lymphoma, mycosis fungoides , Sezary system). central nervous system lymphoma), cutaneous T-cell lymphoma, and primary central nervous system lymphoma.

In specific embodiments, the cancer is selected from glandular cancer. In certain embodiments, the cancer is selected from the group consisting of adrenocortical cancer, pheochromocytoma, paraganglioma, pituitary tumor, thymoma, and thymic carcinoma.

In specific embodiments, the cancer is selected from thyroid cancer. In particular embodiments, the thyroid cancer is selected from the group consisting of medullary thyroid carcinoma, papillary thyroid carcinoma, and follicular thyroid carcinoma.

In specific embodiments, the cancer is selected from germ cell tumors. In certain embodiments, the cancer is selected from the group consisting of malignant extracranial germ cell tumors and malignant extragonadal germ cell tumors. In certain instances of these modalities, the malignant extragonadal germ cell tumors are selected from the group consisting of nonseminoma and seminoma.

In specific embodiments, the cancer is selected from cardiac tumors. In certain embodiments, the cardiac tumor is selected from the group consisting of malignant teratoma, lymphoma, rhabdomyosarcoma, angiosarcoma, chondrosarcoma, infantile fibrosarcoma, and synovial sarcoma.

In specific embodiments, the cell proliferative disorder is selected from benign papillomatosis, benign neoplastic disease, and gestational trophoblastic disease. In certain embodiments, the benign neoplastic disease is selected from skin tags (warts) and genital tags. In certain embodiments, the trophoblastic disease of pregnancy is selected from the group consisting of hydatidiform moles and trophoblastic neoplasms of pregnancy (eg, invasive birthmarks, choriocarcinomas, trophoblastic tumors of the placenta, and epithelioid trophoblastic tumors).

In some embodiments, the subject has melanoma. Melanoma can be in an early or late stage. In some embodiments, the subject has colon cancer. Colorectal cancer can be in early stages or in advanced stages. In some embodiments, the subject has non-small cell lung cancer. Non-small cell lung cancer can be early or late stage. In some embodiments, the subject has pancreatic cancer. Pancreatic cancer can be at an early or late stage. In some embodiments, the subject has a hematologic malignancy. Hematologic malignancy may be at an early or late stage. In some embodiments, the subject has ovarian cancer. Ovarian cancer can be at an early or late stage. In some embodiments, the subject has breast cancer. Breast cancer can be at an early or late stage. In some embodiments, the subject has renal cell carcinoma. Renal cell carcinoma can be early or late stage. In some embodiments, the cancer has increased levels of T cell infiltration.

In some embodiments, cancers treatable with compounds of the present disclosure include melanoma (eg, metastatic malignant melanoma), renal cancer (eg, clear cell carcinoma), prostate cancer (eg, adenocarcinoma hormone refractory prostate cancer), breast cancer, triple negative breast cancer, colon cancer, and lung cancer (for example, non-small cell lung cancer and small cell lung cancer). In addition, the disclosure includes refractory or recurrent malignancies the growth of which can be inhibited using the compounds of the disclosure.

In some embodiments, diseases and indications treatable using the compounds of the present disclosure include, but are not limited to, hematological cancers, sarcomas, lung cancer, gastrointestinal cancer, genitourinary tract cancer, liver cancer, bone cancer, nervous system cancer. , gynecological diseases limited to cancer and skin cancer.

Examples of hematologic cancers include lymphomas and leukemias such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular disease), Hodgkin lymphoma, myeloproliferative disorders (eg, primary myelofibrosis (PMF), polycythemia vera (PV), essential thrombocytosis (ET)), myelodysplasia syndrome (MDS), T-cell acute lymphoblastic lymphoma (T-ALL), multiple myeloma, cutaneous T-cell lymphoma, Waldenstrom macroglubulinemia, hairy cell lymphoma, chronic myeloid lymphoma and Burkitt lymphoma.

Beispielhafte Sarkoma ompassen Condrossarcoma, Sarcoma de Ewing, Osteosarcoma, Rabdomiossarcoma, Angiossarcoma, Fibrossarcoma, Lipossarcoma, Mixoma, Rabdomioma, Rabdossarcoma, Fibroma, Lipoma, Harmatoma y Teratoma.

Examples of lung cancers include non-small cell lung cancer (NSCLC), small cell lung cancer, bronchogenic carcinoma (squamous cell carcinoma, small undifferentiated cell adenocarcinoma, large cell undifferentiated adenocarcinoma), alveolar (bronchiolar) carcinoma , bronchial adenoma, chondromatous hamartoma and mesothelioma.

Examples of gastrointestinal cancers include cancer of the esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small intestine ( adenocarcinoma B. lymphoma). , carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), colon (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), and colorectal cancer.

Examples of cancer of the genitourinary tract include cancer of the kidney (adenocarcinoma, Wilm's tumor [nephroblastoma]), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), and testis (seminoma , teratoma , carcinoma). , teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma).

Examples of liver cancer include hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.

Examples of bone cancers include, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticular cell sarcoma), multiple myeloma, malignant giant cell chordoma, osteochronophroma (osteocartilaginous exostosis) , benign chondroma, chondroblastoma, chondromyxofibroma, osteoidofibroma and giant cell tumor osteoma

Examples of cancers of the nervous system include cancers of the skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meningeal cancer (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, meduoblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma , glioblastoma multiforme, oligodendroglioma, schwannoma, retinoblastoma, congenital) and spinal cord tumors (neurofibroma, meningioma, glioma, sarcoma), as well as neuroblastoma and Lhermitte-Duclos disease.

Examples of gynecologic cancers include cancers of the uterus (endometrial cancer), cervix (cervical cancer, anterior cervical dysplasia), ovary (ovarian cancer (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosathecal cell tumors, Sertoli-Leydig, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), and fallopian tube ( carcinoma).

Examples of skin cancers include melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, Merkel cell skin cancer, dysplastic nevi, lipoma, angioma, dermatofibroma, and keloids. In some embodiments, diseases and indications that can be treated using the compounds of the present disclosure include, but are not limited to, sickle cell disease (eg, sickle cell anemia), triple negative breast cancer (TNBC ), myelodysplastic syndromes, testicular cancer, bile ductal cancer, esophageal cancer, and urothelial carcinoma.

Examples of head and neck cancers include glioblastoma, melanoma, rhabdosarcoma, lymphosarcoma, osteosarcoma, squamous cell carcinoma, adenocarcinoma, oral cancer, laryngeal cancer, nasopharyngeal cancer, sinus and paranasal cancer, thyroid and parathyroid cancer. .

In some embodiments, HPK1 inhibitors can be used to treat tumors that produce PGE2 (eg, Cox-2 overexpressing tumors) and/or adenosine (CD73 and CD39 overexpressing tumors). Cox-2 overexpression has been shown in a variety of tumors, including colon, breast, pancreatic, and lung cancer, where it is correlated with a poor prognosis. COX-2 overexpression has been reported in hematological cancer models such as RAJI (Burkitt's lymphoma) and U937 (acute promonocytic leukemia) and in blast cells from patients. CD73 is upregulated in several human carcinomas, including those of the colon, lung, pancreas, and ovary. It is important to emphasize that higher levels of CD73 expression are associated with neovascularization, tumor invasiveness, and metastasis, and with decreased survival in patients with breast cancer.

In some embodiments, the compounds of the invention are useful for preventing or reducing the risk of developing any of the diseases described herein; B. prevent or reduce the risk of developing a disease, condition, or disorder in a person who may be predisposed to the disease, condition, or disorder but who does not yet have the pathology or symptomatology of the disease.

The compounds described herein can be administered in any suitable manner known in the art. In some embodiments, the compound of the invention or a pharmaceutically acceptable salt, prodrug, metabolite, or derivative thereof is administered intravenously, intramuscularly, subcutaneously, topically, orally, transdermally, intraperitoneally, intraorbitally, by implantation, by inhalation, intrathecally, intraventricular. intratumoral or intranasal.

In some embodiments, the HPK1 antagonist is administered continuously. In other embodiments, the HPK1 antagonist is administered intermittently. Furthermore, treating a subject with an effective amount of an HPK1 antagonist may involve a single treatment or may involve a series of treatments.

It will be appreciated that appropriate doses of the active ingredient will depend on a number of factors well known to the commonly qualified physician or veterinarian. The dose(s) of the active ingredient will vary depending on, for example, the subject's age, body weight, general health, gender and diet, timing of administration, route of administration, clearance rate and any combination of active ingredients.

It is also to be understood that the effective dosage of a compound of the invention or a pharmaceutically acceptable salt, prodrug, metabolite or derivative thereof used in treatment may increase or decrease over the course of a particular treatment. Dose changes may arise and be evident from the results of diagnostic tests.

In some embodiments, the HPK1 antagonist is administered to the subject at a dose of between about 0.001 μg/kg and about 1000 mg/kg, including, among others, about 0.001 μg/kg, 0.01 μg/kg, 0.05 μg /kg, 0.1 µg/kg, 0.5 µg/kg, 1 µg/kg, 10 µg/kg, 25 µg/kg, 50 µg/kg, 100 µg/kg, 250 µg/kg, 500 µg/ kg, 1 mg /kg, 5 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg, 100 mg/kg y 200 mg/kg.

In the methods described herein, the method may further comprise administering a chemotherapeutic agent to the subject. In certain aspects of this embodiment, the chemotherapeutic agent is administered to the subject simultaneously with the compound or composition. In certain aspects of this embodiment, the chemotherapeutic agent is administered to the subject prior to administration of the compound or composition. In certain aspects of this embodiment, the chemotherapeutic agent is administered to the subject after administration of the compound or composition.

As used herein, the terms "treat," "treat," and "treat" refer to reversing, alleviating, delaying the onset, or inhibiting the progression of a disease or disorder, or one or more of its symptoms, such as described in this document. .document In some embodiments, treatment may be administered after the development of one or more symptoms. In other embodiments, the treatment can be administered in the absence of symptoms. For example, treatment can be administered to a susceptible individual prior to the onset of symptoms (eg, given a history of symptoms and/or given genetic or other susceptibility factors). Treatment can also be continued after the symptoms have resolved, for example, to prevent or delay their recurrence.

The term "delivery" or "delivery" encompasses ways of introducing the compound(s) into an individual to carry out its intended function. Examples of routes of administration that can be used include injection (subcutaneous, intravenous, parenteral, intraperitoneal, intrathecal), topical, oral, inhalation, rectal, and transdermal.

The term "effective amount" includes an amount that is effective in the doses and periods of time necessary to achieve the desired result. An effective amount of the compound can vary depending on factors such as the disease state, the age and weight of the subject, and the ability of the compound to elicit a desired response in the subject. Dosage regimens can be adjusted to provide the optimal therapeutic response.

The terms "systemic administration", "systemically administered", "peripheral administration" and "peripherally administered", as used herein, mean the administration of one or more compounds, drugs or other materials that enter the system of the patient and are therefore subject to metabolism. and other similar processes.

The term "therapeutically effective amount" means an amount of a compound of the present invention that (i) treats or prevents the specific disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of the disease, condition, or specific condition or disorder, or (iii) prevents or delays the onset of one or more symptoms of the specific disease, condition or disorder described herein. In the case of cancer, the therapeutically effective amount of the drug can reduce the number of cancer cells; reduce the size of the tumor; inhibit (ie, slow to some extent and preferably stop) the infiltration of cancer cells into peripheral organs; inhibit (ie, slow to some extent and preferably stop) tumor metastasis; to some extent, inhibit tumor growth; and/or alleviate, to some extent, one or more of the symptoms associated with the cancer. To the extent that the drug can prevent the growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic. For example, for cancer therapy, effectiveness can be measured by assessing time to disease progression (TTP) and/or determining response rate (RR).

The term "subject" refers to animals such as mammals, including, but not limited to, primates (eg, humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, and the like. In certain embodiments, the subject is a human.

combined therapies

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

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

Examples of agents with which the combinations of this invention may also be combined include, without limitation: treatments for Alzheimer's disease, such as Aricept® and Excelon®; HIV treatments such as ritonavir; treatments for Parkinson's disease such as L-DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexefendyl, and amantadine; agents for the treatment of multiple sclerosis (MS) such as interferon beta (eg, Avonex® and Rebif®), Copaxone® and mitoxantrone; asthma treatments such as Albuterol and Singulair®; agents for the treatment of schizophrenia such as Zyprexa, Risperdal, Seroquel and Haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporine, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophosphamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anticonvulsants, ion channel blockers, riluzole and antiparkinsonian drugs; agents for the treatment of cardiovascular diseases such as beta blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers and statins; agents for the treatment of liver diseases such as corticosteroids, cholestyramine, interferons and antivirals; agents used to treat blood disorders, such as corticosteroids, antileukemic agents, and growth factors; Agents that prolong or increase pharmacokinetics, such as cytochrome P450 inhibitors (ie, inhibitors of metabolic degradation) and CYP3A4 inhibitors (ie, ketoquenozole and ritonavir), and agents used to treat immunodeficiency disorders, such as gamma globulin.

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

These additional agents can be administered separately from any given combination therapy as part of a multiple dose regimen. Alternatively, these agents can form part of a single dosage form mixed with a compound of this invention in a single composition. When administered as part of a multiple dose regimen, the two active ingredients may be administered simultaneously, sequentially, or within a period of time, generally within five hours.

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

The amount of additional therapeutic agent present in the compositions of this invention is no greater than the amount that would normally be administered in a composition comprising that therapeutic agent as the sole active ingredient. Preferably, the amount of additional therapeutic agent in the compositions described herein is between about 50% and 100% of the amount normally present in a composition comprising that agent as the sole therapeutically active agent.

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

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

In another embodiment, the present invention provides a method of treating rheumatoid arthritis, comprising administering a compound of formula I and one or more additional therapeutic agents selected from non-steroidal anti-inflammatory drugs (NSAIDs), such as aspirin, to a patient whose need for . Ibuprofen, naproxen, etodolac (Lodine®), and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), methotrexate ( Rheumatrex ®), gold salts such as gold thioglucose (Solganal®), gold thiomalate (Myochrysine®) and auranofin (Ridaura®), D-penicillamine (Depen® or Cuprimine®), azathioprine (Imuran®), cyclophosphamide (Cytoxan ®), chlorambucil (Leukeran®), cyclosporine (Sandimmune®), leflunomide (Arava®), and "anti-TNF" agents such as etanercept (Enbrel®), infliximab (Remicade®), golimumab (Simponi®), certolizumab pegal (Cimzia ® ) and adalimumab (Humira®), "Anti-IL-1" agents such as anakinra (Kineret®) and Rilonacept (Arcalyst®), antibodies such as rituximab (Rituxan®), "anti-T cell" agents such as abatacept (Orencia® ) and "anti-IL-6" drugs, such as tocilizumab (Actemra®).

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

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

In some embodiments, the present invention provides a method of treating Crohn's disease, ulcerative colitis or inflammatory bowel disease comprising administering to a patient a compound of formula I and one or more additional therapeutic agents selected from mesalamine (Asacol®) sulfasalazine (Azulfidine®), antidiarrheals such as diphenoxylate (Lomotil®) and loperamide (Imodium®), bile acid chelators such as cholestyramine, alosetron (Lotronex®), lubiprostone (Amitiza®), laxatives such as milk of magnesia, polyethylene glycol ( MiraLax® ), Dulcolax®, Correctol® and Senokot® and anticholinergics or antispasmodics such as Dicyclomine (Bentyl®), anti-TNF therapies, steroids and antibiotics such as Flagyl or Ciprofloxacin.

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

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

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

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

In another embodiment, the present invention provides a method of treating a haematological malignancy comprising administering to a patient in need thereof a compound of formula I and a hedgehog (Hh) signaling inhibitor. In some embodiments, the hematologic malignancy is DLBCL (Ramirez et al. "Defining Causative Factors Contributing in the Activation of Hedgehog Signaling in Diffuse Large B-Cell Lymphoma" Leuk. Res. (2012), published online July 17 and incorporated here for reference included in its entirety).

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

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

In another embodiment, the present invention provides a method of treating or alleviating the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a BTK inhibitor, wherein the disease is selected from an inflammatory disease. intestinal, arthritis, cutaneous lupus erythematosus, systemic lupus erythematosus (SLE), vasculitis, idiopathic thrombocytopenic purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord, Graves' disease, autoimmune thyroiditis, Sjögren's syndrome, multiple sclerosis, systemic sclerosis, Lyme neuroborreliosis, Guillain-Barré syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia , autoimmune hepatitis, autoimmune gastritis, pernicious anemia, celiac disease, Goodpasture's disease, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, membranous glomerulonephropathy, endometriosis, interstitial cystitis, pemphigus vulgaris, bullous pemphigoid, neuromyotonia, scleroderma, vulvodynia, a hyperproliferative disorder, rejection of transplanted organs or tissues, immunodeficiency syndrome (AIDS) , also known as HIV), type 1 diabetes, graft-versus-host disease, transplantation, transfusion, anaphylaxis, allergies (eg. allergies to plant pollen, latex, medications, food, insect venom, animal dander, pet dander, house dust mites or cockroaches), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis and atopic dermatitis, asthma, appendicitis, dermatitis atopic, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic transplant rejection, colitis, conjunctivitis, Crohn's disease, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis , epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, Henoch-Schönlein purpura, hepatitis, hidradenitis suppurativa, immunoglobulin A nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, oophoritis, orchitis , osteitis, otitis, pancreatitis, mumps, pericarditis, peritonitis, pharyngitis, pleurisy, phlebitis, pneumonitis, pneumonia, polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendinitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis or vulvitis, B-cell proliferative disorder, eg B. Diffuse large B-cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, chronic lymphocytic leukemia, acute lymphoblastic leukemia, B-cell prolymphocytic leukemia, Waldenstrom lymphoplasma/macroglobulinemia lymphoma , splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non-Hodgkin lymphoma, Hodgkin lymphoma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mantle cell, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, mast cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia or lymphomatoid granulomatosis, breast cancer, prostate cancer, or mast cell cancer (p. g., mast cell tumor), mast cell leukemia, mast cell sarcoma, systemic mastocytosis) B. Bone cancer, colorectal cancer, pancreatic cancer, bone and joint diseases including, but not limited to, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic and Reiter's arthritis), Behçet's disease, Sjögren's syndrome, systemic sclerosis, osteoporosis, bone cancer, bone metastases, thromboembolic disease (eg B. myocardial infarction, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after CABG, restenosis after CABG, stroke, transient ischemia, peripheral arterial disease, pulmonary embolism, deep vein thrombosis), pelvic inflammatory disease, urethritis, sunburn of the skin, sinusitis, pneumonitis , encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, colocystitis, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irritable bowel syndrome, ulcerative colitis, Sjogren's disease , tissue transplant rejection, hyperacute organ transplant rejection, asthma, allergic rhinitis, chronic obstructive pulmonary disease (COPD), autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, sclerosis multiplex, scleroderma, vasculitis, autoimmune hemolytic and thrombocytopenic conditions, Goodpasture syndrome, atherosclerosis, Addison's disease, Parkinson's disease, Alzheimer's disease, diabetes, septic shock, cutaneous lupus erythematosus, systemic lupus erythematosus (SLE), rheumatoid arthritis, arthritis psoriatic, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Waldenstrom's macroglobulinemia, myasthenia gravis, Hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitarism, Guillain-Barré syndrome, Behçet's disease, scleroderma, mycosis fungoides, acute inflammatory reactions (such as acute respiratory distress syndrome and ischemia/reperfusion injury) and Graves' disease.

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

In another embodiment, the present invention provides a method of treating or reducing the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a PI3K inhibitor, wherein the disease is selected from Benign or malignant tumor, carcinoma, or solid tumor of the brain, kidney (eg, renal cell carcinoma [RCC]), liver, adrenal gland, bladder, breast, stomach, ovaries, colon, rectum, prostate, pancreas, lung, vagina , endometrium, cervix, testicles, urogenital tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastoma, neuroblastoma, multiple myeloma or gastrointestinal cancer, particularly colon or colorectal adenoma or head and neck tumor, epidermal hyperproliferation, psoriasis , prostatic hyperplasia, neoplasm, a neoplasm of an epithelial nature, an adenoma, an adenocarcinoma, a keratoacanthoma, a squamous cell carcinoma, a large cell carcinoma, a non-small cell lung carcinoma, lymphomas (including non-Hodgkin lymphoma ( NHL) and Hodgkin lymphoma (also known as Hodgkin lymphoma or Hodgkin disease)), breast carcinoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, or leukemia, diseases include Cowden syndrome, Lhermitte-Dudos and Bannayan-Zonana disease or diseases in which the PI3K/PKB pathway is abnormally activated, asthma of any type or origin, including intrinsic (non-allergic) and extrinsic (allergic) asthma, mild asthma, moderate asthma , severe asthma, bronchial asthma, exercise-induced asthma, occupational asthma, and asthma induced after bacterial infection, acute lung injury (ALI), adult respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease, respiratory or pulmonary ( COPD, COAD or COLD), including chronic bronchitis or associated dyspnea, emphysema, as well as exacerbation of airway hyperresponsiveness as a result of other drug therapy, particularly other inhaled drug therapy, bronchitis of any type or origin, including , but not limited to, acute, arabic, catarrhal, cropus, chronic or phthinoid bronchitis, pneumoconiosis (an inflammatory, general occupational lung disease often accompanied by chronic bronchitis or acute airway obstruction and caused by repeated inhalation of dusts ) of any type or origin, including, for example, aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tobacco and byssinosis, Löffler syndrome, eosinophilia, pneumonia, parasitic infestations (especially metazoans) (including tropical eosinophilia) , bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma and related eosinophilic diseases affecting the respiratory tract, such as drug reaction, psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforma, dermatitis herpetiformis , scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, pemphis, epidermolysis bullosa acquisita, conjunctivitis, keratoconjunctivitis primrose seca and blight Conjunctivitis, diseases of the nose, including allergic rhinitis, and inflammatory diseases in which autoimmune reactions are involved or have an autoimmune component or etiology, including hematologic autoimmune diseases (eg. hemolytic anemia, aplastic anemia, pure red cell anemia and idiopathic thrombocytopenia), cutaneous lupus erythematosus, systemic lupus erythematosus, rheumatoid arthritis, polychondritis, sclerodoma, Wegener's granulosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, Steven syndrome -Johnson, idiopathic sprue, autoimmune inflammatory bowel disease (eg fibrosis, psoriatic arthritis, and glomerulonephritis (with and without nephrotic syndrome, for example, including idiopathic nephrotic syndrome or nephropathy with mimic changes, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, ischemic stroke and heart failure, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease and cerebral ischemia and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity and hypoxia.

In some embodiments, the present invention provides a method of treating or alleviating the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a Bcl-2 inhibitor, where the disease is an inflammatory disease, a autoimmune disease, a proliferative disorder, an endocrine disorder, a neurological disorder, or a transplant-related disorder. In some embodiments, the disease is a proliferative disease, lupus, or lupus nephritis. In some embodiments, the proliferative disorder is chronic lymphocytic leukemia, diffuse large B-cell lymphoma, Hodgkin's disease, small cell lung cancer, non-small cell lung cancer, myelodysplastic syndrome, lymphoma, hematologic malignancy, or solid tumor.

In some embodiments, the present invention provides a method of treating or reducing the severity of a disease comprising administering to a patient in need of an HPK1-binding compound. In some embodiments, the disease is an autoimmune disease, an inflammatory disease, a proliferative disease, an endocrine disease, a neurological disease, or a transplant-related disease. In some embodiments, the HPK1-binding compound is a compound of formula I.

The compounds and compositions according to the method of the present invention can be administered using any amount and any route of administration useful for treating or reducing the severity of an autoimmune disease, an inflammatory disease, a proliferative disease, an endocrine disease, a neurological disorder or a transplant-related disorder. The exact amount needed will vary from person to person depending on the species, the age and general condition of the person, the severity of the infection, the particular agent, its mode of administration, and the like. The compounds of the present invention are preferably formulated in unit dosage form for ease of administration and uniformity of dosage. The term "unit dosage form", as used herein, refers to a physically discrete unit of composition suitable for the subject to be treated. However, it is to be understood that all daily use of the compounds and compositions of the present invention will be determined by the attending physician within the framework of sound medical judgment. The specific effective dose level for any particular patient or organism will depend on a variety of factors, including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition used; the age, body weight, general health, gender and diet of the patient; the time of administration, route of administration and rate of excretion of the specific compound used; the duration of treatment; Drugs used in combination or concurrently with the specific compound used and similar factors well known in the medical arts. The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.

The pharmaceutically acceptable compositions of this invention can be administered to humans and other animals by the oral, rectally, parenteral, intracisternal, intravaginal, intraperitoneal, topical (as powder, ointment, or drops), buccal, oral or nasal spray routes, or how, depending on the severity of the infection to be treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally in dosage amounts of from about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg of the subject's body weight. per day, once or several times a day to achieve the desired therapeutic effect.

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

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

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

In order to prolong the action of a compound of the present invention, it is often desirable to delay the absorption of the compound by subcutaneous or intramuscular injection. This can be achieved by using a liquid suspension of crystalline or amorphous material with low solubility in water. The rate of absorption of the compound then depends on its rate of dissolution, which in turn may depend on the size and shape of the crystal. Alternatively, delayed absorption of a parenterally administered form of compound is achieved by dissolving or suspending the compound in an oily vehicle. Injectable depot forms are made by forming microcapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of compound to polymer and the nature of the specific polymer used, the rate of release of the compound can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also manufactured by entrapping the compound in liposomes or microemulsions compatible with body tissues.

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

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

Solid compositions of a similar type can also be used as fillers in hard and soft gelatin capsules using excipients such as lactose or milk sugar, as well as high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and other coatings known in the art of pharmaceutical formulation. If necessary, they may contain opacifying agents and may also be composed in such a way that they release the active substance or active substances only or preferentially in a certain part of the intestinal tract, if necessary with a delay. Examples of coatings that can be used are polymeric substances and waxes. Solid compositions of a similar type can also be used as fillers in hard and soft gelatin capsules using excipients such as lactose or milk sugar, as well as high molecular weight polyethylene glycols and the like.

The active compounds may also be in microencapsulated form with one or more excipients as indicated above. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings, release control coatings, and other coatings known in the art of pharmaceutical formulation. In such solid dosage forms, the active ingredient may be mixed with at least one inert diluent such as sucrose, lactose, or starch. Such dosage forms may also comprise, as usual, additional substances in addition to inert diluents, e.g. B. Tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets, and pills, the dosage forms may also contain buffering agents. If necessary, they may contain opacifying agents and may also be composed in such a way that they release the active substance or active substances only or preferentially in a certain part of the intestinal tract, if necessary with a delay. Examples of coatings that can be used are polymeric substances and waxes.

Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, aerosols, inhalants, or plasters. The active component is mixed under sterile conditions with a pharmaceutically acceptable carrier and any necessary preservatives or buffers. Ophthalmic formulations, ear drops, and eye drops are also contemplated within the scope of this invention. Furthermore, the present invention contemplates the use of transdermal patches that have the additional benefit of providing controlled release of a compound in the body. Such dosage forms can be prepared by dissolving or dispensing the compound in the appropriate medium. Absorption enhancers can also be used to increase the flux of the compound through the skin. The rate can be controlled by providing a rate control membrane or by dispersing the compound in a gel or polymer matrix.

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

According to another embodiment, the invention relates to a method for inhibiting the activity of HPK1 or a mutant thereof in a biological sample, comprising the step of contacting the biological sample with a compound of this invention or a composition comprising this compound. In certain embodiments, the invention relates to a method for irreversibly inhibiting the activity of HPK1 or a mutant thereof in a biological sample, which comprises the step of contacting the biological sample with a compound of this invention or a composition comprising this compound.

In another embodiment, the invention provides a method for selectively inhibiting HPK1 at one or more kinases.

The term "biological sample" as used herein includes, without limitation, cell cultures or extracts thereof, biopsy material obtained from a mammal or extracts thereof, and blood, saliva, urine, feces, semen, tears, or other bodily fluids or extracts thereof.

Inhibition of HPK1 (or a mutant thereof) activity in a biological sample is useful for a variety of purposes known to those skilled in the art. Examples of such purposes include, but are not limited to, blood transfusions, organ transplants, storage of biological samples, and biological assays.

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

In another embodiment, the invention relates to a method of inhibiting the activity of HPK1 or a mutant thereof in a patient comprising the step of administering to the patient a compound of the present invention or a composition comprising said compound. According to certain embodiments, the invention relates to a method for reversibly or irreversibly inhibiting one or more activities of HPK1 or a mutant thereof in a patient, comprising the step of administering to the patient a compound of the present invention or a composition comprising said compound. In other embodiments, the present invention provides a method of treating a disorder mediated by HPK1 or a mutant thereof in a patient in need thereof, comprising the step of administering to the patient a compound according to the present invention or a pharmaceutically acceptable composition thereof. . Such disorders are described in detail here.

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

A compound of the present invention can also be used advantageously in combination with other therapeutic compounds. In some embodiments, the other therapeutic compounds are antiproliferative compounds. Such antiproliferative compounds include, but are not limited to, aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule agents; alkylating compounds; histone deacetylase inhibitors; compounds that induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platinum compounds; compounds that target/reduce protein or lipid kinase activity and other antiangiogenic compounds; compounds that target, reduce or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; antiandrogens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; Inhibitors of oncogenic Ras isoforms; telomerase inhibitors; proteasome inhibitors; compounds for use in the treatment of hematological malignancies; compounds that target, decrease, or inhibit the activity of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-desmethoxygeldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozolomide (Temodal®); kinesin spindle protein inhibitors such as SB715992 or SB743921 from GlaxoSmithKline or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array BioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer, and Leucovorin. As used herein, the term "aromatase inhibitor" refers to a compound that inhibits estrogen production, such as the conversion of androstenedione and testosterone substrates to estrone and estradiol, respectively. The term includes but is not limited to steroids, particularly atamestane, exemestane and formestane, and particularly non-steroids, particularly aminoglutethimide, rogletimide, pyridoglutamide, trilostane, testolactone, ketoconazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane is marketed under the trade name Aromasin™. Formestane is sold under the trade name Lentaron™. Fadrozol is marketed under the trade name Afema™. Anastrozole is marketed under the trade name Arimidex™. Letrozole is marketed under the brand names Femara™ or Femar™. Aminoglutethimide is marketed under the tradename Orimeten™. A combination of the invention comprising a chemotherapeutic agent that is an aromatase inhibitor is particularly useful for the treatment of hormone receptor-positive tumors, such as breast tumors.

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

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

The term "topoisomerase I inhibitor" as used herein includes, but is not limited to, topotecan, gimatecan, irinotecan, camptothecium and its analogs, 9-nitrocamptothecin, and the camptothecin macromolecular conjugate PNU-166148. For example, irinotecan can be sold as is, eg, under the trademark Camptosar™. Topotecan is marketed under the trade name Hycamptin™.

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

The term "microtubule drug" refers to microtubule-stabilizing compounds, microtubule-destabilizing compounds, and microtubule polymerization inhibitors, including, but not limited to, taxanes such as paclitaxel and docetaxel; vinca alkaloids such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolids; cochicine and epothilones and their derivatives. Paclitaxel is sold under the trade name Taxol™. Docetaxel is marketed under the trade name Taxotere™. Vinblastine sulfate is marketed under the tradename Vinblastine R.P™. Vincristine Sulfate is marketed under the trade name Farmistin™

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

The term "histone deacetylase inhibitors" or "HDAC inhibitors" refers to compounds that inhibit histone deacetylase and possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).

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

The term "platinum compound" as used herein includes, but is not limited to, carboplatin, cisplatin, cisplatin, and oxaliplatin. Carboplatin can e.g. be administered in the form in which it is marketed, e.g. under the trademark Carboplat™. Oxaliplatin can e.g. be administered in the marketed form, e.g. under the trademark Eloxatin™.

The term “compounds that target/reduce protein or lipid kinase activity; or a protein or lipid phosphatase activity; or other anti-angiogenic compounds" as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors such as platelet growth factor receptors (PDGFR), such as compounds that target, reduce or inhibit PDGFR activity, particularly compounds that inhibit the PDGF receptor, such as a derivative of N-phenyl-2-pyrimidinamine, such as Imatinib, SU101, SU6668 and GFB-111;b ) compounds that target, decrease, or inhibit fibroblast growth (FGFR); c) Compounds that target, reduce, or inhibit insulin-like growth factor I (IGF-IR) activity, such as compounds that target , reduce or inhibit the activity of IGF-IR, particularly compounds that target IGF kinase activity by inhibiting receptor -I or antibodies directed to the extracellular domain of the IGF-I receptor or its growth factors, d) compounds that target, reduce or inhibit the activity of the Trk receptor family of tyrosine kinases, or inhibitors of ephrin B4; e) compounds that target, reduce or inhibit the activity of the AxI receptor tyrosine kinase family; f) compounds that target, decrease or inhibit the activity of the Ret receptor tyrosine kinase; g) compounds that target, decrease or inhibit Kit/SCFR receptor tyrosine kinase activity, such as imatinib; h) Compounds that target, reduce or inhibit the activity of C-Kit receptor tyrosine kinases that are part of the PDGFR family, such as compounds that target, reduce or inhibit the activity of the c-receptor tyrosine kinase family -Kit, in particular compounds that inhibit the c-Kit receptor, such as imatinib; i) Compounds that target, reduce or inhibit the activity of c-Abl family members, their gene fusion products (eg BCR-Abl kinase) and mutants, eg B. Compounds intended to reduce or inhibit the activity of members of the c-Abl family. -Abl family and their gene fusion products, such as an N-phenyl-2-pyrimidinamine derivative such as imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; Parke Davis PD173955; or dasatinib (BMS-354825); j) Compounds that target, reduce or inhibit the activity of members of protein kinase C (PKC) and the Raf family of serine/threonine kinases, members of MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/ Akt, Ras/MAPK family, PI3K, SYK, BTK and TEC and/or members of the cyclin-dependent kinase (CDK) family, including staurosporine derivatives such as midostaurin; Examples of other compounds include UCN-01, safingol, BAY 43-9006, bryostatin 1, perifosine; illmofosine; RO 318220 and RO 320432; IR 6976; lsis 3521; LY333531/LY379196; isoquinoline compounds; FTI; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); k) compounds that target, reduce or inhibit the activity of protein tyrosine kinase inhibitors such as e.g. 50810; AD 99; tyrphostin AG 213; tyrphostin AG 1748; tyrphostin AG 490; tyrphostin B44; tyrphostin B44 (+) enantiomer; tyrphostin AG 555; AG 494; tyrphostin AG 556, AG957 and adafostin (4-{[(2,5-dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adafostin); 1) Compounds that target, reduce, or inhibit the activity of the epidermal growth factor receptor tyrosine kinase family (EGFR1, ErbB2, ErbB3, ErbB4 as homo or heterodimers) and their mutants, such as the EGFR1, ErbB2, ErbB3, ErbB4 family of receptor tyrosine kinases Epidermal Growth Agents are particular compounds, proteins, or antibodies that inhibit members of the EGF receptor tyrosine kinase family, such as the EGF receptor, ErbB2, ErbB3, and ErbB4, or bind EGF or EGF-related ligands, CP 358774 , ZD 1839, ZM 105180; Trastuzumab (Herceptin™), Cetuximab (Erbitux™), Iressa, Tarceva, OSI-774, Cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6. 4, E2.11, E6.3 or E7.6.3 and 7H-pyrrolo[2,3-d]pyrimidine derivatives; m) Compounds that target, decrease or inhibit c-Met receptor activity, such as compounds that target, decrease or inhibit c-Met activity, in particular compounds that target the kinase activity of c-Met receptors Met-inhibiting Met, or antibodies directed to the extracellular body. c-Met domain or HGF binding, n) compounds targeting the kinase activity of one or more members of the JAK family (JAK1/JAK2/JAK3/TYK2 and/or pan-JAK), including but not limited to PRT - 062070, SB-1578, baricitinib, pacritinib, mometinib, VX-509, AZD-1480, TG-101348, tofacitinib, and ruxolitinib; o) Compounds that target, decrease or inhibit PI3 kinase (PI3K) activity, including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib; AND; and q) compounds that target, decrease, or inhibit hedgehog protein (Hh) or smoothing receptor (SMO) signaling pathways, including, but not limited to, cyclopamine, vismodegib, itraconazole, erismodegib, and IPI-926 (saridegib).

The term "PI3K inhibitor," as used in the present document, includes, among others, compounds with inhibitory activity toward one or more enzymes of the phosphatidylinositol 3-kinases family, including, among others, PI3Kα, PI3Kγ, PI3Kδ, PI3Kβ, PI3K-C2α. , PI3K-C2β, PI3K-C2γ, Vps34, p110-α, p110-β, p110-γ, p110-δ, p85-α, p85-β, p55-γ, p150, p101 and p87. Examples of PI3K inhibitors useful in this invention include, inter alia, ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719 , Dactolisib, XL-147, XL-765 and Idelalisib.

The term "BTK inhibitor" as used herein includes, but is not limited to, compounds with inhibitory activity against Bruton's tyrosine kinase (BTK), including, but not limited to, AVL-292 and ibrutinib.

The term "SYK inhibitor" as used herein includes, but is not limited to, compounds with splenic tyrosine kinase (SYK) inhibitory activity, including, but not limited to, PRT-062070, R-343, R-333 , Excellair, PRT -062607 and fostamatinib.

The term "Bcl-2 inhibitor" as used herein includes, but is not limited to, compounds with inhibitory activity against B-cell lymphoma-2 (Bcl-2) protein, including, but not limited to, ABT-199, ABT -731, ABT-737, Apogosypol, Ascenta Pan-Bcl-2 Inhibitors, Curcumin (and its analogues), Dual Bcl-2/Bcl-xL Inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14 -1 (and its analogues, see WO2008118802), Navitoclax (and its analogues, see US Patent No. 7,390,799), NH-1 (Shenayng Pharmaceutical University), Obatoclax (and its analogues, see WO2004106328), p -001 (Gloria Pharmaceuticals), TW series compounds (Univ., Michigan) and venetoclax. In some embodiments, the Bcl-2 inhibitor is a therapeutic small molecule. In some embodiments, the Bcl-2 inhibitor is a peptidomimetic.

Other examples of BTK inhibitor compounds and conditions that can be treated with such compounds in combination with compounds of this invention can be found in WO2008039218 and WO2011090760, all of which are incorporated herein by reference.

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

Other examples of PI3K inhibitory compounds and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2004019973, WO2004089925, WO2007016176, US Patent 8,138,347, WO2002088112, WO2007084786, WO2007129161, WO2007129161, WO2005113554 and WO2007044729, all of which are incorporated herein by reference.

Additional examples of JAK inhibitor compounds and conditions that can be treated with such compounds in combination with compounds of this invention can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246 and WO2007070514, all of which are incorporated herein by reference. .

Other anti-angiogenic compounds include compounds with a different mechanism for their activity, e.g. independent of inhibition of protein or lipid kinases, e.g. Thalidomide (Thalomid™) and TNP-470.

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

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

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

The term cyclooxygenase inhibitor as used herein includes inhibitors of Cox-2, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives such as celecoxib (Celebrex™), rofecoxib (Vioxx™), etoricoxib, valdecoxib or a 5-alkyl-2-arylaminophenylacetic acid such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenylacetic acid, lumiracoxib.

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

As used herein, the term "heparanase inhibitor" refers to compounds that target, reduce, or inhibit the degradation of heparin sulfate. The term includes, but is not limited to, PI-88. The term "biological response modifier" as used herein refers to a lymphokine or interferons.

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

As used herein, the term "methionine aminopeptidase inhibitor" refers to compounds that target, reduce, or inhibit the activity of methionine aminopeptidase. Compounds that target, decrease, or inhibit methionine aminopeptidase activity include, but are not limited to, bengamide or a derivative thereof.

As used herein, the term "proteasome inhibitor" refers to compounds that target, reduce, or inhibit the activity of the proteasome. Compounds that target, reduce, or inhibit proteasome activity include, but are not limited to, bortezomib (Velcade™) and MLLN 341.

The term "matrix metalloproteinase inhibitor" or ("MMP" inhibitor) as used herein includes, but is not limited to, peptidomimetic and non-peptidomimetic inhibitors of collagen, tetracycline derivatives, e.g. Peptidomimetic hydroxamate inhibitor Batimastat and its oral bioavailable analogue Marimastat (BB-2516), Prinomastat (AG3340), Metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B, or AAJ996.

The term "compounds used in the treatment of hematologic malignancies" as used herein includes, but is not limited to, FMS-type tyrosine kinase inhibitors, which are compounds that affect receptor tyrosine kinase activity of type FMS (Flt-3R); interferon, 1-β-D-arabinofuransylcytosine (ara-c), and bisulfan; ALK inhibitors, which are compounds that target, reduce, or inhibit anaplastic lymphoma kinase, and Bcl-2 inhibitors.

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

The term "HSP90 inhibitors" as used herein includes, but is not limited to, compounds that target, reduce, or inhibit the intrinsic ATPase activity of HSP90; Degrade, target, reduce, or inhibit HSP90 client proteins via the ubiquitin-proteasome pathway. Specifically, compounds that target, decrease, or inhibit the intrinsic ATPase activity of HSP90 are compounds, proteins, or antibodies that inhibit the ATPase activity of HSP90, such as 17-allylamino, 17-desmethoxygeldanamycin (17AAG), geldanamycin derivative; other compounds related to geldanamycin; Radicicol and HDAC inhibitors.

As used herein, the term "antiproliferative antibodies" includes, but is not limited to, trastuzumab (Herceptin™), trastuzumab-DM1, Erbitux, bevacizumab (Avastin™), rituximab (Rituxan®), PR064553 (anti-CD40), and 2C4 antibodies Antibodies are understood to mean intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies and antibody fragments, provided they have the desired biological activity.

For the treatment of acute myelogenous leukemia (AML), the compounds of the present invention can be used in combination with standard anti-leukemia therapies, particularly in combination with therapies used for the treatment of AML. In particular, the compounds of the present invention may be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as daunorubicin, adriamycin, Ara-C, VP-16, teniposide, mitoxantrone. B idarubicin, carboplatin and PKC412. In some embodiments, the present invention provides a method of treating AML associated with an ITD and/or D835Y mutation, comprising administering a compound of the present invention together with one or more FLT3 inhibitors. In some embodiments, FLT3 inhibitors are selected from quizartinib (AC220), a staurosporine derivative (eg, midostaurin or lestaurtinib), sorafenib, tandutinib, LY-2401401, LS-104, EB-10, famitinib, NOV -110302, NMS-P948, AST-487, G-749, SB-1317, 5-209, SC-110219, AKN-028, fedratinib, tozasertib, and sunitinib. In some embodiments, the FLT3 inhibitors are selected from quizartinib, midostaurin, lestaurtinib, sorafenib, and sunitinib.

Other antileukemic compounds include, for example, Ara-C, a pyrimidine analog that is the 2'-alpha-hydroxyribose (arabinoside) derivative of deoxycytidine. Also included are the purine analogue of hypoxanthine, 6-mercaptopurine (6-MP), and fludarabine phosphate. Compounds that target, reduce, or inhibit the activity of histone deacetylase (HDAC) inhibitors, such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA), inhibit the activity of enzymes known as histone deacetylase. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), trichostatin A, and compounds listed in US Pat. 6,552,065, including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2-hydroxyethyl){2-(1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide or a pharmaceutically acceptable salt thereof, especially the lactate salt. Somatostatin receptor antagonists, as used herein, refer to compounds that target, treat, or inhibit the somatostatin receptor, such as octreotide and SOM230. Approaches that damage tumor cells are referred to as approaches such as ionizing radiation. The term "ionizing radiation" mentioned above and below means ionizing radiation that occurs as electromagnetic radiation (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiotherapy and is well known in the art. See Hellman, Principles of radiation therapy, cancer, in Principles and Practice of Oncology, Devita et al., eds., 4ºedition, vol. 1, pp. 248-275 (1993).

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

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

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

Angiostatic steroids as used herein refer to compounds that block or inhibit angiogenesis, such as. B. Anecortave, triamcinolone, hydrocortisone, 11-α-epihydrocotisol, cortexolone, 17α-hydroxyprogesterone, corticosterone, deoxycorticosterone, testosterone, estrone, and dexamethasone.

Implants that contain corticosteroids refer to compounds such as fluocinolone and dexamethasone.

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

The compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drugs, such as anti-inflammatories, bronchodilators or antihistamines, particularly in the treatment of obstructive or inflammatory diseases of the airways, such as those mentioned above, for example. , as enhancers of the therapeutic activity of said drugs or as a means to reduce the required dose or possible side effects of said drugs. A compound of the invention can be mixed with the other drug in a solid pharmaceutical composition, or it can be administered separately, before, simultaneously or after the other drug. Accordingly, the invention includes a combination of a compound of the invention as described above with an anti-inflammatory, bronchodilator, antihistamine or antitussive, the compound of the invention and the drug being contained in the same or different pharmaceutical compositions.

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

Suitable antihistamines include cetirizine hydrochloride, paracetamol, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, ativastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine.

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

The structure of the active substances identified by code numbers, generic or trade names can be found in the current edition of the standard compendium "The Merck Index" or in databases, e.g. Patents International (for example, IMS World Publications).

A compound of the present invention can also be used in combination with known therapeutic methods, for example, administration of hormones or radiation. In certain embodiments, a provided compound is used as a radiosensitizer, particularly for the treatment of tumors that have low susceptibility to radiotherapy.

A compound of the present invention may be administered alone or in combination with one or more therapeutic compounds, possible combination therapy taking the form of fixed combinations or administration of a compound of the invention and one or more therapeutic compound or staggered. administered independently, or the combined administration of fixed combinations and one or more therapeutic compounds. A compound of the present invention may be administered together with or in addition to, in particular, tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgery, or a combination thereof. Long-term therapy is also possible, as is adjuvant therapy as part of other treatment strategies as described above. Other possible treatments are therapy to maintain the patient's condition after tumor regression, or even chemopreventive therapy, for example, in patients at risk.

These additional agents can be administered separately from a composition containing a compound of the invention as part of a multiple dosing regimen. Alternatively, these agents can form part of a single dosage form mixed with a compound of this invention in a single composition. When administered as part of a multiple dose regimen, the two active ingredients may be administered simultaneously, sequentially, or within a period of time, generally within five hours.

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

The amount of a compound of the invention and an additional therapeutic agent (in compositions comprising an additional therapeutic agent as described above) that can be combined with the carrier materials to produce a single dosage form will vary depending on the host treated and the specific nature. vary by administration. Preferably, the compositions of this invention should be formulated so that a dose of between 0.01 and 100 mg/kg body weight/day of a compound of the invention can be administered.

In such compositions comprising an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions is less than that required in monotherapy using only that therapeutic agent. In such compositions, a dose of between 0.01 and 1000 µg/kg body weight/day of the additional therapeutic agent may be administered.

The amount of additional therapeutic agent present in the compositions of this invention is no greater than the amount that would normally be administered in a composition comprising that therapeutic agent as the sole active ingredient. Preferably, the amount of additional therapeutic agent in the compositions described herein is between about 50% and 100% of the amount normally present in a composition comprising that agent as the sole therapeutically active agent.

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

ILLUSTRATION

As illustrated in the examples below, in certain exemplary embodiments, the compounds are prepared according to the following general procedures. It is to be understood that while the general procedures set forth the synthesis of particular compounds of the present invention, the following general procedures and other procedures known to those skilled in the art can be applied to all compounds, subclasses, and species of any compound as described herein. . Additional compounds of the invention were prepared by procedures substantially similar to those described herein in examples and procedures known to those skilled in the art.

manufacture of intermediate products Method CA1 - Preparation of 2-(6-aminopyridin-3-yl)-N-ethyl-2-methylpropanamide (CA1)

Step 1: Ethyl-2-methyl-2-(6-nitropyridin-3-yl)propanoate (CA1.1)

A solution of ethyl 2-(6-nitropyridin-3-yl)acetate (CA1.0) (980 mg, 4.6 mmol) in DMF (20 mL) at 0 °C was treated with sodium hydride (dispersion of 60% mineral oil, 196 mg, 4.8 mmol). The mixture was stirred for 5 minutes and then iodomethane (0.316 mL, 5.0 mmol) was added dropwise. After a further 2 h, a further portion of sodium hydride (60% dispersion in mineral oil, 196 mg, 4.8 mmol) was added, followed 5 min later by iodomethane (0.316 mL, 5.0 mmol). The mixture was stirred at room temperature for 18h, quenched with water (30ml) and extracted into EtOAc (2 x 30ml). The combined extracts were washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated in vacuo. The residue was purified by column chromatography (0-50% gradient elution with EtOAc in cyclohexane) to give the title compound (CA1.1) (758 mg, 68%) as a yellow oil.1RMN-H (400 MHz, CDCl3): δ 8,63 (s, 1H), 8,22 (d, J=8,4 Hz, 1H), 7,99 (d, J=8,2 Hz, 1H ), 4,16 (q, J=7,2 Hz, 2H), 1,67 (s, 6H), 1,20 (t, J=7,2 Hz, 3H)

Step 2: 2-methyl-2-(6-nitropyridin-3-yl)propanoic acid (CA1.2)

A solution of ethyl 2-methyl-2-(6-nitropyridin-3-yl)propanoate (CA1.1) (750 mg, 3.1 mmol) in methanol-water (14 mL, 1:1) was treated with lithium hydroxide monohydrate (198 mg, 4.7 mmol) and stirred at room temperature overnight. The pH was adjusted to ~5 using 1 M HCl aqueous solution and the mixture was extracted with DCM-MeOH 9:1 (3 x 25 mL). The combined organic extracts were dried over Na2SO4filtered and concentrated in vacuo to give the title compound as an off-white crude solid (CA1.2) (423 mg, 63%).1H-RMN (400 MHz, DMSO): δ 12,87 (s, 1H), 8,67 (d, J = 1,8 Hz, 1H), 8,28 (d, J = 8,6 Hz, 1H ), 8,19 (dd, J = 2,4, 8,5). Hz, 1H), 1,59 (s, 6H).

Stage 3: N-ethyl-2-methyl-2-(6-nitropyridin-3-yl)propenamide (CA1.3)

To a suspension of 2-methyl-2-(6-nitropyridin-3-yl)propanoic acid (CA1.2) (430 mg, 2.0 mmol) in THF (5 mL) was added 1-hydroxybenzotriazole (359 mg , 2.6 mmol) and N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (510 mg, 2.6 mmol). The mixture was stirred at room temperature for 10 minutes. DIPEA (1.069 mL, 6.1 mmol) and ethylamine (2M soln in THF, 2.04 mL, 4.0 mmol) were added and the reaction stirred at room temperature overnight. The reaction was diluted with EtOAc (20ml) and the organic phase was washed with water (15ml) and brine (15ml). The organic phase was dried (MgSO4), filtered and concentrated in vacuo. The residue was purified by column chromatography (0-100% gradient elution with EtOAc in cyclohexane) to give the title compound (CA1.3) (375 mg, 77%) as a light yellow oil.1RMN-H (400 MHz, DMSO): δ 8,58 (d, J=2,0 Hz, 1H), 8,27 (d, J=7,8 Hz, 1H), 8,10 (dd, J =2,4, 8,5 Hz, 1H), 7,60 (dd, J=6,2, 6,2 Hz, 1H), 3,11-3,03 (m, 2H), 1,53 ( s, 6H), 0,97 (dd, J=7,2, 7,2 Hz, 3H).

Etapa 4: 2-(6-aminopyridin-3-yl)-N-etil-2-metilpropanamide (CA1)

N-ethyl-2-methyl-2-(6-nitropyridin-3-yl)propenamide (CA1.3) (375 mg, 1.5 mmol) dissolved in methanol (5 mL) 10% palladium on charcoal was added (35mg) . After hydrogenation under an atmosphere of hydrogen overnight, the mixture was filtered through Celite and the solvent was removed in vacuo to give the title compound (CA1) (320 mg, 96%) as a pale yellow oil which used without further purification.1RMN-H (400 MHz, DMSO): δ 7,83 (d, J=2,0 Hz, 1H), 7,26 (dd, J=2,5, 8,6 Hz, 2H), 6,38 (d, J=8,1 Hz, 1H), 5,74 (s, 2H), 3,05-3,01 (m, 2H), 1,36 (s, 6H), 0,93 (t, J=7,0 Hz, 3H).

Method CA2 - Preparation of 2-(6-aminopyridin-3-yl)-N-ethyl-N,2-dimethylpropanamide (CA2)

Intermediate CA2 was synthesized according to the CA1 method, but using N-methylethanamine for step 3. The crude product was used without further purification or characterization.

Method CA3 - Preparation of 5-(3-(2-Methoxyethoxy)azetidin-1-yl)pyridin-2-amine (CA3)

Intermediate CA3 was prepared according to the methods described in WO2015131080, but using 3-(2-methoxyethoxy)azetidine.1RMN H (400 MHz, CDCl3) δ 7,40 (d, J=2,3 Hz, 1H), 6,74 (dd, J=3,0, 8,6 Hz, 1H), 6,47–6,43 (m, 1H), 4,50–4,42 (m, 1H ), 4,11–4,01 (m, 2H), 3,65 (t, J=6,3 Hz, 2H), 3,60 -3,54 (m, 4H), 3,40-3,38 (m, 3H). 2H No se observan protones intercambiables.

Method PA1 - Preparation of 1-(6-aminopyridin-3-yl)piperidin-4-ol (PA1)

Schritt 1. 5-(3,4-Dihydro-2H-pyran-5-yl)-2-nitropyridine (PA1.1) .

To a stirred solution of 3,4-dihydro-2H-pyran-5-ylboronic acid, pinacol ester (0.217 g, 1.06 mmol, 1.0 eq.) in THF (4.5 mL) and water (0 0.5 mL) 5-Bromo-2-nitropyridine (0.336 g, 1.60 mmol, 1.5 eq.), S-Phos (0.043 g, 0.106 mmol, 0.1 eq.), and potassium phosphate (0.680 g , 3.206 mmol, 3.0 eq.). After degassing under argon for 5 minutes, palladium acetate (0.011 g, 0.053 mmol, 0.05 eq) was added and the reaction mixture degassed again under argon for 10 minutes. After stirring at 60°C for 1 hour, the reaction mixture was quenched with water (50ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was washed with brine (25 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 19% ethyl acetate in n-hexane to give pure PA1.1 (0.140 g, 63.51%). MS(ES): m/z 206.20 [M+H]+.

Stage 2. 1-(6-aminopyridin-3-yl)piperidin-4-ol (PA1)

To a suspension of palladium hydroxide (0.03 g) in methanol (3 mL) was added a solution of 5-(3,4-dihydro-2H-pyran-5-yl)-2-nitropyridine (PA1.1 ) (0.140 g) given, 0.678 mmol, 1.0 eq) in methanol (3 mL) under a nitrogen atmosphere. Hydrogen gas was bubbled through the reaction mixture for 1 hour. After the completion of the reaction, the reaction mixture was filtered through celite and washed with methanol. The filtrate was concentrated under reduced pressure to give pure PA1 (0.095 g, 78.45%). MS(ES): m/z 178.24 [M+H]+, RMN 1H (400 MHz, DMSO-d6) δ 7,77 (s, 1H), 7,28–7,25 (dd, J=2 Hz, 8,4 Hz, 1H), 6,37 (d, J=8,4, 1H), 5,72 (s, 2H), 3,94 (s, 2H), 3,84 (d, 12,4 Hz, 1H), 3,72 (d, J=8 ,4 Hz, 1H), 3,2 (t, J=11,4, 1H), 1,82 (s, 1H), 1,62 (s, 3H).

Method PA2 - Preparation of 5-(2-methyltetrahydro-2H-pyran-2-yl)pyridin-2-amine (PA2)

Step 1. 2-(6-Chloropyridin-3-yl)pent-4-en-2-ol (PA2.1)

To a stirred solution of 1-(6-chloropyridin-3-yl)ethane-1-one PA2.0 (1.2 g, 7.71 mmol, 1.0 eq) dissolved in THF (12 mL) and cooled to -78 °C, add allyl magnesium bromide (in ether) (11.56 mL, 11.56 mmol, 1.5 equiv.). After stirring for 30 minutes at -78°C, the reaction mixture was quenched with water (50 mL) and ammonium chloride solution (10 mL) and extracted with ethyl acetate (50 mL x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 15% ethyl acetate in n-hexane to give pure PA2.1 (1.31 g, 85.93%). MS(ES): m/z 197.66 [M+H]+.

Step 2. 5-(2-(Allyloxy)pent-4-en-2-yl)-2-chloropyridine (PA2.2)

To a stirred solution of 2-(6-chloropyridin-3-yl)pent-4-en-2-ol PA2.1 (1.31 g, 6.6 mmol, 1.0 eq.) dissolved in THF (5 mL), at 0°C. Sodium hydride (0.792 g, 19.8 mmol, 3.0 eq) was added. After stirring at 0 °C for 10 min, allyl bromide (2.0 g, 16.5 mmol, 2.5 eq.) was slowly added to the reaction mixture. After stirring at 0 °C for 15 min, the reaction mixture was refluxed for 6 h. After the completion of the reaction, the reaction mixture was quenched with cold 0.5N hydrochloric acid solution (100 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 8% ethyl acetate in n-hexane to give pure PA2.2 (1.05 g, 66.64%). MS(ES): m/z 237.73 [M+H]+.

Step 3. 2-Chloro-5-(2-methyl-3,6-dihydro-2H-pyran-2-yl)pyridine (PA2.3)

To a stirred solution of 5-(2-(allyloxy)pent-4-en-2-yl)-2-chloropyridine PA2.2 (0.5 g, 2.1 mmol, 1.0 eq.) in DCM ( 5 mL) of Grubbs' catalyst (0.021 g, 0.025 mmol, 0.012 eq.). After stirring at room temperature for 45 minutes, the solvent was concentrated in vacuo. The residue was purified by column chromatography eluting with 15% ethyl acetate in n-hexane to give pure PA2.3 (0.4 g, 90.70%). MS(ES): m/z 209.67 [M+H]+.

Step 4. 2-Chloro-5-(2-methyl-3,6-dihydro-2H-pyran-2-yl)pyridine (PA2.4)

To a stirred solution of 2-chloro-5-(2-methyl-3,6-dihydro-2H-pyran-2-yl)pyridine (0.4 g, 1.9 mmol, 1.0 eq) tris(dibenzylideneacetone )dipalladium(0) (0.174 g, 0.19 mmol, 0.1 eq) and (2-biphenyl)dicyclohexylphosphine (0.134 g, 0.38 mmol, 0.2 eq) in 1,4-dioxane (5 mL) , degassed with argon for 30 minutes, gave lithium hexamethyldisilazide (5.7 mL ), 5.7 mmol, 3.0 equiv.). After heating at 70°C for 3 hours, the reaction mixture was poured into water (50ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 2.5% methanol in DCM to give pure PA2.4 (0.255 g, 70.26%). MS(ES): m/z 190.25 [M+H]+.

Etapa 5.5-(2-Methyltetrahydro-2H-piran-2-il)piridin-2-amina (PA2)

To a suspension of 10% Pd/C (0.050 g) in methanol (3 mL) was added a solution of 2-chloro-5-(2-methyl-3,6-dihydro-2H-pyran-2-yl 0.4)pyridine PA2 (0.250 g, 1.3 mmol, 1.0 eq.) in methanol (3 mL). h2(Gas) was bubbled into the reaction mixture for 1 hour. After the reaction was complete, the reaction mixture was filtered through celite and washed with methanol (30 ml). The filtrate was concentrated under reduced pressure to give pure PA2 (0.220 g, 54.42%). MS(ES): m/z 192.26 [M+H]+, pureza LCMS: 97,40%, pureza HPLC: 96,52%.

Method PA4 - Preparation of tert-butyl 3-(6-aminopyridin-3-yl)-3,6-diazabicyclo[3.1.1]heptane-6-carboxylate (PA4)

Schritt 1. terc-butyl-3-(6-nitropyridin-3-yl)-3,6-diazabicyclo[3.1.1]heptano-6-carboxylate (PA4.1)

To a solution of 5-bromo-2-nitropyridine (1.1 g, 5.42 mmol, 1.0 eq.) in 1,4-dioxane (10 mL), tert-butyl-3,6-diazabicyclo[3.1 .1]heptane-6-carboxylate (1.18 g, 5.96 mmol, 1.1 eq) and potassium carbonate (2.24 g, 16.26 mmol, 3.0 eq), degassed with nitrogen gas for 10 min, 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (0.62 g, 1.08 mmol, 0.2 eq) and tris(dibenzylideneacetone)dipalladium (0.49 g, 0.542 mmol, 0, 1 eq). After stirring at 120 °C for 5 h, the reaction mixture was diluted with water (100 mL) and extracted into ethyl acetate (100 mL x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified using combi-flash silica eluting with 7% methanol/DCM to give material such as tert-butyl-3-(6-nitropyridin-3-yl)-3,6-diazabicyclo[3.1.1] to give heptane-6-carboxylate PA4.1 (1 g, yield - 57.03%). MS(ES): m/z 321.35 [M+H]+.

Schritt 2. terc-butyl-3-(6-aminopyridin-3-yl)-3,6-diazabicyclo[3.1.1]heptan-6-carboxylate (PA4)

To a suspension of 10% Pd/C (0.050 g) in methanol (3 mL) was added a solution of tert-butyl-3-(6-nitropyridin-3-yl)-3,6-diazabicyclo[3.1.1 ] heptane -6-carboxylate (0.850 g, 2.65 mmol, 1.0 eq.) in methanol (3 mL). After bubbling hydrogen gas through the solution for 1 hour, the reaction mixture was filtered through celite which was washed with MeOH. The filtrate was concentrated under reduced pressure to give crude PA4 (0.900 g, 55.16%). MS(ES): m/z 291.37[M+H]+.

Method PA9 - Preparation of tert-butyl 3-(6-aminopyridin-3-yl)-3,6-diazabicyclo[3.1.1]heptane-6-carboxylate (PA9)

Etapa 1,6-Fluoro-2-metil-3,4-dihydroisoquinolin-1(2H)-ona (PA9.1)

To a solution of 6-fluoro-3,4-dihydroisoquinolin-1(2H)-one PA9.0 (2.5 g, 15.13 mmol, 1.0 eq.) in dry THF (130 mL) was added to 0 °C sodium hydride (60% in mineral oil) (1.2 g, 30.27 mmol, 2.0 eq.). After stirring for 30 min, methyl iodide (4.3 g, 30.27 mmol, 2.0 eq.) was added. After stirring at room temperature for 1 h, the reaction mixture was diluted with saturated sodium bicarbonate solution (100 mL) and extracted with ethyl acetate (50 mL x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure to give PA9.1 (3.07 g, quantitative yield). MS (ES): m/z 180.2 [M+H]+

Etapa 2.6-Fluoro-2-metil-7-nitro-3,4-dihidroisoquinolina-1(2H)-ona (PA9.2)

To a solution of 6-Fluoro-2-methyl-3,4-dihydroisoquinolin-1(2H)-one PA9.1 (2.9 g, 16.201 mmol, 1.0 eq) dissolved in concentrated sulfuric acid (25 mL) , potassium nitrate (1.44 g, 17.821 mmol, 1.1 equiv.) at room temperature. After stirring at room temperature for 2 h, the reaction mixture was poured into ice-water and stirred for 10 min. The solid was filtered, washed with water and dried under high vacuum to give PA9.2 (3.1g, 85.44%). MS (ES): m/z 225 [M+H]+

Stage 3. 6-Fluoro-2-methyl-7-nitro-1,2,3,4-tetrahydroisoquinoline (PA9.3)

To a solution of 6-fluoro-2-methyl-7-nitro-3,4-dihydroisoquinolin-1(2H)-one PA9.2 (3.0 g, 13.392 mmol, 1.0 eq) dissolved in THF borane complex -THF (0.9 molar) (22 mL, 20.089 mmol, 1.5 eq) at 0 °C. After stirring for 2 hours at 80 °C, the reaction mixture was cooled to room temperature and diluted with water (100 mL). The aqueous layer was extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified using combi-flash silica eluting with 5% methanol/DCM to give PA9.3 (0.8 g, 28.44%). MS (ES): m/z 211.2 [M+H]+

Stage 4. 6-Fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-amine (PA9) .

To a suspension of 10% Pd/C (158 mg, 0.2% w/w) in methanol (3 mL) was added a solution of 6-fluoro-2-methyl-7-nitro-1,2, 3,4-Tetrahydroisoquinoline PA9.3 (0.79 g, 3.761 mmol, 1.0 eq.) in methanol (30 mL). After washing the solution with hydrogen gas for 1 hour, the reaction mixture was filtered through celite and washed with MeOH. The filtrate was concentrated under reduced pressure to give crude PA9 (0.56 g, 82.68%). MS (ES): m/z 181.3 [M+H]+

Method PA28 - Preparation of 3-(6-Aminopyridin-3-yl)-1-methylpyrrolidin-2-one (PA28)

Stage 1. 6-Fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-amine (PA28.1)

To a solution of 2-(6-chloropyridin-3-yl)acetonitrile PA28.0 (8 g, 52.4 mmol, 1.0 eq.) in ethanol (40 mL) at 0 °C was added sulfuric acid ( 15 ml). After stirring at 100°C for 3 hours, the reaction mixture was poured into ice-water (300ml) and extracted with ethyl acetate (50ml x 3). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to give the crude material PA28.1 which was used directly for the next step without further purification (8 g, 76.4%). MS(ES): m/z 200.30 [M+H]+.

Step 2. 3-(6-Chloropyridin-3-yl)-1-methylpyrrolidin-2-one (PA28.2)

To a solution of 6-fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-amine PA28.1 (1.0 g, 5.025 mmol, 1.0 eq) dissolved in DMSO (10 mL) to 10°C Sodium hydride (0.603 g, 25.12 mmol, 3.0 eq) was added. After stirring at the same temperature for 20 minutes, 2-bromo-N-methylethane-1-amine (1.3 g, 6.030 mmol, 1.2 eq.) was added. After stirring at room temperature for 1h, the reaction mixture was poured into ice-water (50ml) and extracted with ethyl acetate (50ml x 3). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to give the crude material PA28.2 (0.258 g, 24.48%), MS(ES): m/z 211.30 [M+H]+ .

Paso 3. N-(5-(1-Methyl-2-oxopyrrolidin-3-yl)pyridin-2-yl)cyclopropanecarboxamida (PA28.3)

The reaction of Step 3 was carried out according to the representative procedure described in FIG.

Method PA4 (Step 1) using 3-(6-chloropyridin-3-yl)-1-methylpyrrolidin-2-one PA28.2 and cyclopropanecarboxamide to give PA28.3 (0.80 g, yield - 64.99%). MS(ES): m/z 260.61 [M+H]+ Stage 4. 3-(6-aminopyridin-3-yl)-1-methylpyrrolidin-2-one (PA28) .

To a solution of N-(5-(1-methyl-2-oxopyrrolidin-3-yl)pyridin-2-yl)cyclopropanecarboxamide (0.500 g, 1.93 mmol, 1.0 eq.) in MeOH (7 mL) 5N sodium hydroxide solution (2.31 mL, 56.0 mmol, 6 eq.). After stirring at 60 °C for 16 h, the reaction mixture was poured into ice-water (100 mL) and extracted with 10% chloroform/isopropyl alcohol (100 mL x 3). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was further purified by diethyl ether solvent trituration (20 mL x 2) to give pure PA28 (0.250 g, 67.80%). MS(ES): m/z 192.51 [M+H]+

Method PA29 and PA30 - Preparation of (S)-5-(3-(dimethylamino)piperidin-1-yl)pyridin-2-amine (PA29) and (R)-5-(3-(dimethylamino)piperidin-1- yl)pyridin-2-amine (PA30)

Stage 1 and Stage 2. 5-(3-(dimethylamino)piperidin-1-yl)pyridin-2-amine (PA29.2)

The reaction was carried out according to the representative procedure described in Method PA4 to give PA29.2 (0.980 g, 45.15%). MS(ES): m/z 221.35 [M+H].

Schritt 3. (S)-5-(3-(Dimethylamino)piperidin-1-yl)pyridin-2-amine (PA29) y (R)-5-(3-(Dimethylamino)piperidin-1-yl)pyridin- 2-amine (PA30)

Compounds 29.2 (980 mg of racemate) were detected by Chiral SFC on a Shimadzu LC-20AP and UV detector with CHIRALPAK IC (250 x 21.0) mm, 5 micron column at a flow rate of 20 mL/min using mobile phase ( A) 0.1% IN DEA separated n-hexane and (B) 0.1% DEA in propan-2-ol:acetonitrile (70:30) to give pure compounds PA29 (350 mg) and PA30 (351 mg). Assign the stereochemistry arbitrarily.

Method PA31 - Preparation of 5-(oxetan-3-yl)pyridin-2-amine (PA31)

Step 1. 2-Bromo-5-(oxetan-3-yl)pyridine (PA32.1)

To a solution of (6-bromopyridin-3-yl)boronic acid (1.10 g, 5.44 mmol, 1 eq), nickel iodide (51 mg, 0.16 mmol, 0.2 eq), nickel iodide (51 mg, 0.16 mmol, 0.2 eq), sodium trans-2-aminocyclohexanol (24.73 mg, 0.16 mmol, 0.1 eq) in 2-propanol (5 mL) was added sodium bis(trimethylsilyl)amide (1.0 g, 5.44 mmol, 1 eq). After stirring under nitrogen for 10 minutes, a solution of 3-iodoxetane (0.5 g, 2.7 mmol) in 2-propanol (2 mL) was added. After shaking in a sealed flask at 80 for 16 h, the reaction was filtered through celite and washed with ethanol. The filtrate was concentrated and the residue was purified by flash chromatography on silica gel (10-100% ethyl acetate in hexanes) to give the title compound PA32.1 (0.28 g, 26%). MS(ES) m/z 215.87 [M+2+H]+

Stage 2 and Stage 3. 5-(oxetan-3-yl)pyridin-2-amine (PA31) .

The reaction of Steps 2 and 3 was carried out according to the representative procedure described in Method PA28 (Steps 3 and 4) using 2-bromo-5-(oxetan-3-yl)pyridine (PA32.1) and cyclopropanecarboxamide to give PA31 to give (0.15 g, 76.36% yield). MS(ES): m/z 151.2 [M+H]+

Method PA32 - Preparation of 3-(6-Aminopyridin-3-yl)-1-methylpyrrolidin-2-one (PA32)

Etapa 1. 6-Fluoro-1'-metil-[3,4'-bipyridina]-2'(1'H)-one (PA32.1)

To a solution of 2-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (2.0 g, 0.0089 mol, 1.0 eq) and 4-bromo-1-methylpyridin-2(1H)-one (1.3 g, 0.0074 mol, 0.8 eq) in 1,2-dimethoxyethane (10 mL) and water (2 mL) were treated in displaced shape.2CO3(2.8 g, 0.0267 mol, 3.0 eq) and [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) catalyst (0.36 g, 0.0004 mol, 0.05 eq ). After stirring at 120°C in a microwave for 30 minutes, the reaction mixture was diluted with water (60ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified using combi-flash silica eluting with 3% methanol in DCM to give PA32.1 (1.4 g, 76.46%), MS (ES): m/z 205.3 [M+H] +

Etapa 2. 4-(6-Fluoropyridin-3-yl)-1-methylpiperidin-2-one (PA32.2)

To a suspension of 10% Pd/C (558 mg, 0.5% w/w) in methanol (3 mL) was added a solution of 6-fluoro-1'-methyl-[3,4'-bipyridine] -2' given. (1'H)-one PA32.1 (1.0 g, 0.0049 mol, 1.0 eq) in methanol (30 mL). After hydrogen gas was bubbled through the solution for 1 hour, the reaction mixture was filtered through celite. The filtrate was concentrated under reduced pressure to give crude PA32.2 (1.0 g, 98.06%), MS (ES): m/z 209.2 [M+H]+

Etapa 3. 4-(6-aminopyridin-3-yl)-1-methylpiperidin-2-one (PA32)

A solution of 4-(6-fluoropyridin-3-yl)-1-methylpiperidin-2-one PA32.2 (0.850 g, 0.00048 mol, 1.0 eq) in ammonia (40 mL) in a steel bomb (40% headspace remaining) was heated at 150°C (100 psi) for 20 h. The reaction mixture was poured into water (50ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified using combi-flash silica eluting with 6.2% methanol in DCM to give PA32 (0.220 g, 26.26%), MS (ES): m/z 206.3 [M+H]+

Method PA35 - Preparation of 5-cyclopentyl-6-((dimethylamino)methyl)pyridin-2-amine (intermediate PA35)

Step 1. Methyl 6-Amino-3-bromopicolinate (PA35.1)

To a solution of PA35.0 (500 g, 3289.4 mmol, 1.0 eq) in acetonitrile (12.5 L) was added N-bromosuccinamide (644 g, 3618.4 mmol, 1.1 eq). Added in portions at room temperature for 30 min. After stirring at room temperature for 30 min, the reaction mixture was quenched with 10% Na.2S2o3The solution was washed with water (3.0 L) and concentrated to remove acetonitrile. The residue was diluted with 10% Na2S2o3The solution was washed with water (20L) and extracted with 50% ethyl acetate in hexanes (10L x 5). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The crude material was triturated with 25% ethyl acetate in hexane to give neat PA35.1 (methyl 6-amino-3-bromopicolinate). (57.07% yield). MS (ES): m/z 231-233 [M+2]+,1H NMR (400 MHz, CDCl3): δ 7.66 (d, 1H), 6.53 (d, 1H), 4.68 (S, 2H), 3.98 (s, 3H). Note: Isomers of other regions (methyl 6-amino-5-bromopicolinate) also form and can be separated by silica gel purification. Required regioisomer confirmed by 1H NMR and NOE analysis.

Step 2. Methyl 3-bromo-6-(bis(tert-butoxycarbonyl)amino)picolinate (PA35.2)

To a solution of PA35.1 (1100 g, 4782.6 mmol, 1.0 eq) in THF (20 L) was added dimethylaminopyridine (116.7 g, 956.5 mmol, 0.2 eq) and Boc anhydride. (2502 g, 11478.2 mmol, 2.4 eq) data). After stirring at 75 °C for 1.5 h, the solvent was evaporated and the residue was diluted with brine and extracted with ethyl acetate (2 x 10 L). The combined organic layer was dried over sodium sulfate and concentrated in vacuo. The residue was purified by column chromatography eluting with 5% ethyl acetate in hexane. The isolated material was triturated with hexanes (4 L) to give PA35.2 (1700 g, 82.79%) as a white solid. MS (ES): m/z 431-433 [M+2]+.1RMN-H (400 MHz, DMSO): δ 8,32 (d, 1H), 7,61 (d, 1H), 3,90 (s, 3H), 1,40 (s, 18H).

Stage 3. Methyl 6-(bis(tert-butoxycarbonyl)amino)-3-(furan-3-yl)picolinate (PA35.3) .

To a solution of PA35.2 (730 g, 1693.7 mmol, 1.0 eq) and furanboronic acid (379 g, 3387.4 mmol, 2 eq) and potassium phosphate tribasic (Sigma, 1078.3 g, 5086 0.2 mmol, 3.0 eq) was suspended in 1-4-dioxane (5.85 L) and water (1.46 L) and degassed for 20 min under a stream of nitrogen Dikis (59.5 g, 84 0.8 mmol, 0.05 eq). After stirring at 120 °C for 15 min, the reaction was cooled to room temperature. The organic layer was collected, filtered through a pad of celite, and concentrated under reduced pressure. The residue was purified by chromatography on silica gel eluting with 6.0% to 10% ethyl acetate/hexane. The isolated material was triturated with n-pentane to give PA35.3 (84% yield) as an off-white solid. MS(ES): m/z 418 [M+1]+1RMN-H (400 MHz, DMSO): δ 8,12 (d, 1H), 8,00 (d, 1H), 7,81 (s, 1H), 7,61 (s, 1H), 6,70 (S, 1H), 3,83 (S, 3H). ), 1,41 (s, 18H)

Stage 4. Methyl 6-(bis(tert-butoxycarbonyl)amino)-3-(furan-3-yl)picolinate (PA35.4) .

To a solution of PA35.4 (191 g, 456.9 mmol, 1.0 eq.) in methanol (1140 mL) and THF (955 mL) was added ammonium formate (115.1 g, 182.5 mmol, 4.0 eq.), acetic acid (133.7 mL, 0.7 V) and 20% NASS palladium hydroxide on carbon (133.7 g, 1:0.7 w/w). After stirring under an atmosphere of hydrogen gas for 24 h at room temperature, the reaction mixture was combined with 6 other batches of the same scale, prepared by the same procedure. The combined reaction mixture was then filtered through a celite bath and the filtrate was concentrated in vacuo. The residue was washed with sat. NaHCO3(10 L) and extracted with DCM (10 L x 3) to give PA35.4 (1251 g, 92.6%). MS(ES): m/z 423 [M+1]+1RMN-H (400 MHz, DMSO): δ 8,01 (d, 1H), 7,58 (d, 1H), 3,99 (t, 2H), 3,87 (S, 3H), 3,71 (m, 2H), 3,60 (m, 1H). ), 2,31 (m, 1H) 1,91 (d, 1H), 1,4 (S, 18H).

Step 5. tert-butyl (6-(hydroxymethyl)-5-(THF-3-yl)pyridin-2-yl)carbamate (PA35.5)

To a solution of PA35.4 (250 g, 592.4 mmol, 1.0 eq) in ethanol (2500 mL) was added sodium borohydride (135 g, 355.4 mmol, 6 eq) portionwise. After stirring at 60 °C for 2 h, the reaction was concentrated under reduced pressure, diluted with water (10 L) and extracted with DCM (4x10 L). The combined organic layer was washed with brine (10 L), dried over Na2SO4, filtered and concentrated under vacuum. The residue was combined with 4 other batches from the same scale, prepared by an identical procedure, to give PA35.5 (640 g, 73.49%) as a colorless gummy liquid that turned white after 2 days at room temperature and it became solid. MS(ES): m/z 295.0 [M+1]+1RMN-H (400 MHz, DMSO): δ 7,81 (d, 1H), 7,67 (d, 1H), 7,2 (d, 1H), 5,22 (d, 1H), 4,55 (t, 1H), 3,99 (s, 3H). ), 3,77 (m, 3H), 3,55 (m, 2H), 2,28 (d, 2H), 1,87 (d, 1H), 1,41 (s, 9H)

Schritt 6 & 7. tert-butyl-(6-((dimethylamino)methyl)-5-(THF-3-yl)pyridin-2-yl)carbamate (PA35.6).

To a solution of PA35.5 (440 g, 149.6 mmol, 1.0 eq.) in DCM (6.5 L) was added dropwise diisopropylethylamine (581.4 g, 448.9 mmol, 3.0 eq.) at 0ºC. Mesyl chloride (257.04 g, 2244 mmol, 1.5 eq) was added at 0 °C. After stirring at 0 °C at RT for 2 h, the reaction mixture was quenched with DI water (1 L) and with DCM (3 x 2 L). The combined organic layer was washed with brine (10 L), rinsed with Na2SO4The frit was washed and concentrated under reduced pressure to give a mesylated intermediate. The mesylated intermediate was combined with another batch (on a 200 g scale) made by an identical procedure. (700 g crude, 86.44%) as a pale yellow liquid. MS(ES): m/z 373.35 [M+1]+.

To a solution of mesylated intermediate (350 g, 940.0 mmol, 1.0 eq.) in MeCN (3.5 L) at room temperature was added dropwise diisopropylethylamine (529.23 g, 423.0 mmol, 4.5 eq.). ) followed by dimethylamine hydrochloride (152.41 g, 1880.0 mmol, 2.0 eq). After stirring at 90 °C for 3 h, the reaction mixture was concentrated to remove acetonitrile, quenched in DI water (1500 mL) and extracted with DCM (3 x 3 L). The combined organic layer was washed with brine (10L), dried over Na2SO4, and concentrated under reduced pressure to give PA35.6. The product was mixed with another batch of the same scale, produced by an identical process. (700 g, quantitative yield) as a brown semi-solid. MS(ES): m/z 322.39 [M+1]+,1RMN-H (400 MHz, DMSO): δ 9,61 (S, 1H), 7,67 (S, 2H), 3,96–3,94 (t, 2H, J=7,6), 3, 78–3,77 (d, 2H, J=4 Hz), 3,57 (S, 1H), 3,54 (s, 1H), 2,26-2,25 (t, 1H), 2,1 (S, 6H), 1,86 (S, 1H), 1,45 (s, 9H)

Step 8. 5-Cyclopentyl-6-((dimethylamino)methyl)pyridin-2-amine (PA35)

To a solution of PA35.6 (700 g, 2180.7 mmol, 1.0 eq.) in DCM (5.0 L) was added trifluoroacetic acid (2.1 L, 3 v) at 0 °C. After stirring at 70 °C for 2 h, the reaction mixture was concentrated, diluted with deionized water (2 L) and washed with heptane. The collected aqueous layer was neutralized with 10% NaOH solution and then extracted with 15% MeOH in DCM (4 x 3.0 L). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was triturated with 20% ethyl acetate in hexanes then diethyl ether to give neat PA35 as a tan solid. (330g, 68.47%). MS (ES): m/z 222.30 [M+1]+,1RMN-H (400 MHz, DMSO-d6) δ 7,33 (d, J=8,4 Hz, 1H), 6,36 (d, J=8,5 Hz, 1H), 5,71 (s, 1H), 3,96–3,85 (m, 1H), 3,72 (dq, J = 31,0, 7,7 Hz, 1H), 3,46–3,34 (m, 1H), 3 ,35 (s, 1H), 3,30 (d, J = 11,9 Hz, 0H), 2,19 (td, J = 7,8). , 4,2 Hz, 0H), 2,14 (s, 3H), 1,79 (dq, J=12,2, 8,0 Hz, 1H).

Method PA37 - Preparation of 6-((dimethylamino)methyl)-5-(tetrahydro-2H-pyran-4-yl)pyridin-2-amine (PA37)

Step 1. tert-butyl (5-bromo-6-(hydroxymethyl)pyridin-2-yl)carbamate (PA37.1).

To a solution of PA35.2 (50 g, 116.27 mmol, 1.0 eq) in ethanol (200 mL) was added sodium borohydride (26.3 g, 697.6 mmol, 6 eq) portionwise. After stirring at 70 °C for 2 h, the reaction was concentrated under reduced pressure, carefully diluted with water (200 mL) and extracted into DCM (3 x 150 mL). The combined organic layer was washed with brine (100 mL), passed through a hydrophobic filter, and concentrated under reduced pressure to give PA37.1 (27 g, 79%) as a white solid. MS(ES): m/z 395 [M+1]+1RMN-H (400 MHz, DMSO): δ 7,81 (d, 1H), 7,67 (d, 1H), 7,2 (d, 1H), 5,22 (d, 1H), 4,55 (t, 1H), 3,99 (s, 3H). ), 3,77 (m, 3H), 3,55 (m, 2H), 2,28 (d, 2H), 1,87 (d, 1H), 1,41 (s, 19H)

Step 2, 3. tert-butyl-(5-bromo-6-((dimethylamino)methyl)pyridin-2-yl)carbamate (PA37.2)

To a solution of PA37.1 (22.2 g, 73.2 mmol, 1.0 eq.) and NN-diisopropylethylamine (33.3 g, 256.3 mmol, 3.5 eq.) in DCM (200 mL ) at 0 °C methanesulfonyl chloride (12.5 g, 109.8 mmol, 1.5 eq.) was added. After stirring for 30 min, the reaction was quenched with water (100 ml) and extracted with ethyl acetate (3 x 40 ml). The combined organic layer was washed with brine, passed through a hydrophobic filter, and concentrated under reduced pressure.

To the mesylate intermediate dissolved in acetonitrile (200 mL) was added dimethylamine (15 g, 183.0 mmol, 2.5 eq) and NN-diisopropylethylamine (33.3 g, 256.3 mmol, 3.5 eq) was given. . After stirring at 70°C for 1 hour, the reaction was quenched with water (100 mL) and extracted into ethyl acetate (3 x 40 mL). The combined organic layer was washed with brine, passed through a hydrophobic filter, and concentrated under reduced pressure. The residue was purified by chromatography on silica gel eluting with 50% ethyl acetate/hexane to give PA37.2 (17.0 g, 94.3%). MS(ES): m/z 330 [M+H]+

Schritt 3. tert-butyl-(5-(3,6-dihydro-2H-pyran-4-yl)-6-((dimethylamino)methyl)pyridin-2-yl)carbamate (PA37.3) .

To a solution of PA37.2 (50 g, 151.5 mmol, 1.0 eq.) in 1,4-dioxane:water (400 mL:100 mL) with 2-(3,6-dihydro-2H -pyran -4-yl )-4,4,5,5-Tetramethyl-1,3,2-dioxaborolane (47.7 g, 227.2 mmol, 1.5 eq) and potassium tribasic phosphate (96.3 g 454.5 mmol, 3.0 eq)2X-phosPdG2 (11.9 g, 15.1 mmol, 0.1 eq) was added over 15 min. After stirring at 140 °C for 4 h, the reaction mixture was cooled to room temperature, diluted with water (1 L) and extracted with ethyl acetate (2 × 2 L). The combined organic extracts were washed with brine (1 L), dried over Na2SO4filtered and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 2% methanol in DCM to give PA37.3 (40 g, 79%), MS (ES): m/z 334.2 [M+H]+

Schritt 4. Tert-butyl-(6-((dimethylamino)methyl)-5-(tetrahydro-2H-pyran-4-yl)pyridin-2-yl)carbamate (PA37.4) .

To a suspension of palladium hydroxide (130 g) in methanol (600 ml) and THF (40 ml) was added the intermediate PA37.3 (130 g, 1.0 eq.). Hydrogen gas was bubbled through the reaction mixture for 4 h at room temperature. After the reaction was complete, the reaction mixture was filtered through a pad of celite which was washed with methanol. The filtrate was concentrated under reduced pressure to give PA37.4 (120 g, 91.75%). MS (ES): m/z 336.2 [M+H]+

Schritt 5. 6-((Dimetilamino)metil)-5-(tetrahidro-2H-piran-4-il)piridin-2-amina (PA37)

To a solution of intermediate PA37.4 (120 g, 356.9 mmol, 1.0 eq.) in DCM (1.2 L) was added trifluoroacetic acid (360 mL) dropwise. After stirring at 55 °C for 2 h, the reaction mixture was neutralized with saturated sodium hydroxide solution and extracted with 10% methanol in DCM (4 x 10 L). The combined organic layer was concentrated under reduced pressure to give PA37 (66 g, 78.40%). MS(ES): m/z 236.1 [M+H]+

Method PA39 - Preparation of 2-(1-(6-aminopyridin-3-yl)piperidin-3-yl)propan-2-ol (PA39)

Step 1 and Step 2. Ethyl 1-(6-Aminopyridin-3-yl)piperidinine-3-carboxylate (PA39.2).

Ethyl 1-(6-aminopyridin-3-yl)piperidine-3-carboxylate (PA39.2) was prepared in a similar manner as described in Method PA4 (1.2 g, 89.62%). MS(ES): m/z 250.15 [M+H]

Stage 3. 2-(1-(6-aminopyridin-3-yl)piperidin-3-yl)propan-2-ol (PA39) .

For a solution of ethyl 1-(6-aminopyridin-3-yl)piperidine-3-carboxylate (PA39.2) (2.0 g, 8.03 mmol, 1.0 eq) in THF (15 mL), a solution of methylmagnesium bromide (3N in THF, 20 mL) dropwise at 0°C. After stirring at room temperature for 1h, the reaction mixture was quenched with ice water (100ml) and filtered through a pad of celite. The filtrate was extracted with ethyl acetate (100 mL x 3) and the combined organic layer was washed with brine (100 mL) and concentrated under reduced pressure. The residue was purified by column chromatography (0-80% gradient elution with EtOAc in hexanes) to give PA39 (0.200 g, 10.60%). MS(ES): m/z 236.17 [M+H]+

Method PA40 - Preparation of 1-(6-amino-2-((dimethylamino)methyl)pyridin-3-yl)piperidin-4-ol (PA40)

Step 1. 6-Chloro-3-(4-hydroxypiperidin-1-yl)piconaldehyde (PA40.1)

For a solution of PA40.0 (1.0 g, 6.28 mmol, 1.0 eq) and piperidin-4-ol (1.0 g, 10.04 mmol, 1.6 eq) N,N-dimethylformamide (10 mL) was potassium carbonate (2.6 g, 18.84 mmol, 3.0 equiv.). After stirring at 100 °C for 1 hour, the reaction mixture was cooled to room temperature, diluted with ice water (100 ml) and extracted with ethyl acetate (3 x 40 ml). The combined organic extracts were washed with brine (80 mL), dried over Na2SO4filtered and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 30% ethyl acetate in hexane to give PA40.1 (1.2 g, 79.54%), MS (ES): m/z 241.07 [M+H ]+

Stage 2. 1-(6-Chloro-2-((dimethylamino)methyl)pyridin-3-yl)piperidin-4-ol (PA40.2) .

To a cooled solution of PA40.1 (1.2 g, 5.00 mmol, 1.0 eq.) in 1,2-dichloroethane (20 mL) was added acetic acid (2.4 mL) at 0 °C. . The reaction was bubbled with dimethylamine gas for 30 minutes before sodium triacetoxyborohydride (7.4 g, 35 mmol, 7.0 eq) was added portionwise. After stirring at room temperature for 16 h, the reaction mixture was diluted with ice water (100 mL) and extracted with DCM (4 x 40 mL). The combined organic extracts were washed with brine (90 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 3.0% methanol in DCM gradient to give PA40.2 (0.450 g, 33.46%), MS(ES): m/z 270.2 [M+H ]+

Stage 3 and Stage 4. 5-(oxetan-3-yl)pyridin-2-amine (PA40) .

The reaction in Steps 3 and 4 was carried out according to the representative procedure described in Method PA28 (Steps 3 and 4) using PA40.2 and cyclopropanecarboxamide to give PA40 (0.3 g, 64%). MS(ES): m/z 251.35 [M+H]+

The following aniline intermediates were prepared according to one of the PA1-PA40 intermediate processes as described above.

# STRUCTURE Method # STRUCTURE Method PA3 5-(Tetrahydro-2H-pyran-4-yl)pyridin-2-amine (as PA1) PA5 terc-butyl-3-(6-aminopyridin-3-yl)-3,8-diazabicyclo[3.2.1]octan-8-carboxylate (as PA4) PA6 (R)-1-(6-aminopyridin-3-yl)-3,3-dimethyl-piperidin-4-ol (as PA4) PA7 terc-butyl-4-(6-aminopyridin-3-yl)-1,4-diazepan-1-carboxylate (as PA4) PA8 (1-(6-aminopyridin-3-yl)piperidin-4-yl)(methyl)carbamate (as PA4) tert-butyl PA10 5-(4-oxa-7-azaspiro[2.5]octan-7-yl)pyridin-2-amine (as PA4) PA11 5-(2-Azaspiro[4.4]nonan-2-yl)pyridine-2-amine (as PA4) PA12 6-methoxy-2-methyl-1,2,3,4-tetra-hydro-isoquinoline-7-amine (as PA9) PA13 5-cyclohexyl-pyridine-2-amine (como PA1) PA14 tert-butyl-7-amino-6-methoxy-3,4-dihydro-isoquinoline-2(1H)-carboxylate (as PA9) PA15 5-Cyclopentyl-Pyridine-2-Amine (as PA1) PA16 (R)-(1-(6-aminopyridin-3-yl)pyrrolidin-3-yl)carbamate (tert-butyl) (as PA4) PA17 tert-butyl-3-(6-aminopyridin-3-yl)piperidin-1-carboxylate (as PA1) PA18 (S)-(1-(6-aminopyridin-3-yl)piperidin-3-yl)(methyl)carbamate (as PA4) PA19 (S)-(1-(6-aminopyridin-3-yl)pyrrolidin-3-yl)methanol (as PA4) PA20 (R)-(1-(6-aminopyridin-3-yl)piperidin-3-yl)(methyl)carbamate (as PA4) PA21 5-(3- Fluorpiperidin-1-yl)pyridine-2-amine (as PA4) PA22 tert-butyl-3-(6-aminopyridin-3-yl)piperidin-1-carboxylate (as PA1) PA23 (S)-(1-(6-aminopyridin-3-yl)piperidin-3-yl)carbamate (tert.-butyl) (as PA4) PA24 5-(3-Isopropyl-pyrrolidin-1-yl)pyridine-2-amine (as PA4) PA25 (R)-(1-(6-aminopyridin-3-yl)piperidin-3-yl)carbamate (tert.-butyl) (as PA4) PA26 (R)-(1-(6-aminopyridin-3-yl)pyrrolidin-3-yl)methanol (as PA4) PA27 (3S,5R)-4-(6-aminopyridin-3-yl)-3,5-dimethylpiperazine-1-carboxylic acid tert-butyl ester (as PA4) PA33 (S)-1-(6-aminopyridin-3-yl)-3,3-dimethylpiperidin-4-ol (as PA4) PA34 (3aR,6aS)-5-(6-aminopyridin-3-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-carboxylate of tert-butyl (as PA4) PA36 (1-(6-aminopyridin-3-yl)piperidin-4-yl)(methyl)carbamate (as PA4) tert-butyl PA38 2-(4-(6-aminopyridin-3-yl)morpholin-2-yl)propan-2-ol (using PA4) PA41 6-((dimethylamino)methyl)-5-morpholino-pyridine-2-amine (using PA40) PA42 1-(6-Amino-2-((dimethylamino)methyl)pyridin-3-yl)-4-(methoxymethyl)piperidin-4-ol (como PA40)

Method CB1 - Preparation of 5-Fluoro-2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)(piperidin-1-yl)methanone (CB1 ) )

Step 1. 5-Fluoro-2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborlan-2-yl)benzoic acid (CB1.1)

To a solution of methyl 5-fluoro-2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborlan-2-yl)benzoate (CB 1.0) (2.0 g, 6.45 mmol) in THF (60 mL) was added LiOH (2 M, 7.1 mL, 14.2 mmol). After stirring at room temperature for 16 h, the organic solvent was removed under reduced pressure and the residual material was neutralized with 2M aqueous HCl (20 mL), diluted with brine (10 mL), then extracted into ethyl acetate. (3×50ml). )ml). The combined organic layers were washed with brine (40 mL), passed through a hydrophobic frit, and concentrated under reduced pressure to give the product (CB 1.1) (1.66 g, 87%) as a white solid which was obtained without further purification. . m/z=297.1 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 10,83 (brs, 1H), 7,85 (d, J=8,7 Hz, 1H), 7,38 (d, J=4,0 Hz, 1H), 4,13 (s, 3H), 1,40 (s, 2H).

Schritt 2. 5-Fluor-2-methoxy-4-(4,4,5,5-tetrametil-1,3,2-dioxaborolan-2-yl)fenil)(piperidin-1-yl)methanone (CB1)

To a solution of (CB1.1) (1.66 g, 5.61 mmol) in THE (25 mL) were added hydroxybenzotriazole (0.98 g, 7.29 mmol) and EDC hydrochloride (1.40 g, 7th 0.29 mmol) provided. After stirring at room temperature for 1 h, N,N-diisopropylethylamine (2.9 mL, 16.8 mmol) and piperidine (0.66 mL, 6.73 mmol) were added sequentially. After stirring at room temperature for 22 h, the organic solvent was evaporated under reduced pressure, diluted with water (40 mL) and extracted with DCM (2 x 40 mL). The combined organic layer was passed through a hydrophobic frit and concentrated under reduced pressure. The resulting foam was dissolved in DCM (10 ml) and the product precipitated by addition of diethyl ether. The solvent was removed under reduced pressure to give crude (CB1) (2.07 g, quant.) as an off-white solid which was used without further purification. m/z=364.2 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,18 (d, J=4,2 Hz, 1H), 6,92 (d, J=8,1 Hz, 1H), 3,85 (s, 3H), 3,82–3, 64 (m, 4H), 1,56–1,41 (m, 4H). ), 1,36 (s, 12H), 1,33-1,25 (m, 2H).

Method CB2 - Preparation of N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-N-methylcyclopropanecarboxamide (CB2)

Step 1. N-(3-Fluoro-4-(4,4,5,5-tetrametil-1,3,2-dioxaborolan-2-il)fenil)ciclopropanocarboxamida (CB2.1)

For a solution of 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (CB2.0) (600 mg, 2.53 mmol) in chlorine of Se añadieron cyclopropanecarbonyl (0.24 ml, 2.66 mmol) and N,N-diisopropylethylamine (0.88 ml, 5.06 mmol) to DCM (12.0 ml). After stirring at room temperature for 2 h, the reaction was quenched with 2M HCl (20 ml) and extracted with DCM (3 x 10 ml). The combined organic layer was washed with brine (20 mL), passed through a hydrophobic frit and concentrated under reduced pressure to give the crude product (CB2.1) (650 mg, 85%) as a white solid which was obtained without further purification was used. m/z=306.1[M+H]+.1RMN-H (400 MHz, CDCl3): δ 7,61-7,55 (m, 1H), 7,47 (d, J=1,6 Hz, 1H), 7,25 (s, 1H), 7,18 (d, J=1 ,6 Hz, 1H), 1,54-1,50 (m, 1H), 1,38–1,30 (m, 12H), 1,88–1,83 (m, 2H), 0,75– 0,69 (m, 2H).

Paso 2. N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-N-methylcyclopropanecarboxamide (CB2)

For a solution of N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)cyclopropanecarboxamide (CB2.1) (650 mg, 2. Sodium hydride (60% in mineral oil, 145 mg, 3.62 mmol) was added portionwise to 13 mmol) in dry THF (6.0 mL) and methyl iodide (0.65 mL, 3.62 mmol). . After stirring at room temperature for 4 h, the reaction was quenched [carefully!] dropwise with water (1.0 mL) and extracted with EtOAc (2 x 5 mL). The combined organic layers were dried over anhydrous MgSO.4and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with 0-100% EtOAc in isohexane) to give the title compound (CB2) (390 mg, 57%) as an off-white solid. m/z=320.2 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 8,75 (s, 1H), 8,08 (d, J=1,6 Hz, 1H), 8,02 (d, J=1,6 Hz, 1H), 3,28 (s, 3H), 2,54 (s, 1H), 1,68-1,60 (m, 12H), 0,88–0,83 (m, 2H), 0,75–0,69 (m, 2H) .

Method CB3 - Preparation of 2-(2-Fluoro-4-methoxy-5-methylphenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (CB3)

To a mixture of 1-bromo-2-fluoro-4-methoxy-5-methylbenzene (CB3.0) (500 mg, 2.28 mmol), bis(pinacolato)diboron (696 mg, 2.74 mmol) and acetate Potassium (560 mg, 5.71 mmol) in 1,4-dioxane (10 mL) was converted to [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II), complexed with DCM (186 mg, 0. 23 mmol), admitted. After degassing with nitrogen, the mixture was heated at 100 °C for 6 h. The cooled mixture was diluted with EtOAc. The organic solution was collected, washed with water, brine, dried over Na2SO4and concentrated in emptiness. The residue was purified by silica gel chromatography (0-50% EtOAc in isohexane gradient) to give the title compound (CB3) (539 mg, 89%) as a tan solid.

Method CB4 - Preparation of 5-Fluoro-N,N-dimethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,3-di-hydro-1H -indeno-1-amine (CB4)

Stage 1: 6-bromo-5-fluoro-2,3-di-hydro-1H-inden-1-ol (CB4.1);

To a solution of 6-bromo-5-fluoro-2,3-dihydro-1H-inden-1-one (CB4.0) (300 mg, 1.3 mmol) in methanol (3.0 mL) was added sodium borohydride (141 mg) given, 3.9 mmol). After stirring at room temperature for 30 min, the mixture was diluted with DCM (20 mL) and washed with saturated aqueous NaHCO 3 .3(15 ml). The organic layer was passed through a hydrophobic frit and evaporated under reduced pressure to give the desired product (CB4.1) (307 mg, quant.) as an off-white solid, which was used in the next step without further purification. m/z=231.9 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,56 (d, J=6,3 Hz, 1H), 7,02–6,97 (m, 1H), 5,25–5,16 (m, 1H), 3,06–2 ,96 (m, 1H), 2,83–2,72 (m, 1H), 2,58–2,47 (m, 1H), 2,04–1,94 (m, 1H), 1,78 (d, J = 6,8 Hz, 1H).

Stage 2: 6-bromo-5-fluoro-N,N-dimethyl-2,3-di-hydro-1H-indeno-1-amine (CB4.2)

For a solution of 6-bromo-5-fluoro-2,3-dihydro-1H-inden-1-ol (CB4.1) (307 mg, 3.9 mmol) in THE (12 mL) and triethylamine (0. 94 mL, 5.1 mmol) at -15 °C, methanesulfonyl chloride (200 µL, 2.6 mmol) was added dropwise. After stirring for 3h at -15°C, a solution of dimethylamine (2M in THF, 7.8mL, 15.5mmol) was added. After stirring overnight, warming to room temperature, the reaction mixture was filtered and the solvent was evaporated under reduced pressure. The crude product was passed through an SCX cartridge (loaded with DCM, washed with 10% MeOH in DCM, eluting with 10% 7N methanolic ammonia in DCM). The solvent was evaporated under reduced pressure to give the desired product (CB4.2) (310 mg, 92%) as a reddish-brown oil, which was used in the next step without further purification. m/z=259.1 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,51 (d, J=6,7 Hz, 1H), 6,97–6,94 (m, 1H), 4,26 (t, J=6,8 Hz, 1H), 2, 93–2,72 (m, 2H), 2,23 (s, 6H). ), 2,12-2,03 (m, 2H).

Stage 3: 5-Fluoro-N,N-dimethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,3-di-hydro-1H-indene -1 -amine (CB4)

A large carousel tube was charged with 6-bromo-5-fluoro-N,N-dimethyl-2,3-dihydro-1H-indene-1-amine (CB4.2) (310 mg, 1.2 mmol), bis(pinacolato)diboron (475 mg, 1.8 mmol) and potassium acetate (354 mg, 3.6 mmol) in dioxane (10 mL) gave [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) , complexed with DCM (44 mg, 0.06 mmol). The tube was sealed and heated at 110°C for 3 hours with shaking. The reaction mixture was allowed to cool to room temperature and compound (CB4) was carried on to the next step without further purification. m/z=306.2 [M+H]+.

Método CB5 - 4-((3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)methyl)pyridine (CB5)

Step 1: 4-((4-Bromo-3-fluorophenoxy)methyl)pyridine (CB5.1)

To a solution of 4-bromo-3-fluorophenol (CB5.0) (1.00 g, 5.24 mmol) in DMF (8 mL) was added 4-(bromomethyl)pyridine (1.59 g, 6, 28 mmol) and potassium carbonate (1.81 g, 13.1 mmol). After stirring at room temperature overnight, the reaction mixture was diluted with ethyl acetate and washed with water (three times) and brine. The organic layer was dried over Na2SO4filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel eluting with 0-100% EtOAc in DCM to give the desired product (CB5.1) (162 mg, 11%) as a white solid. m/z=283.9 [M+H]+,1H-RMN (400 MHz, CDCl3): δ 8,63 (d, J=6,1 Hz, 2H), 7,45-7,38 (m, 1H), 7,32 (d, J=5, 8 Hz, 2H), 6,73 (dd, J=2,4, 8,4 Hz, 1H), 6,67 (dd, J=2,3, 11,0 Hz, 1H), 5,09 ( s, 2H).

Step 2: 4-((3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)methyl)pyridine (CB5)

A mixture of 4-((4-bromo-3-fluorophenoxy)methyl)pyridine (CB5.1) (155 mg, 0.55 mmol), bis(pinacolato)diboron (167 mg, 0.66 mmol), [1 ,1'-Bis( Diphenylphosphino)ferrocene]dichloropalladium(II) complex was degassed with DCM (45 mg, 0.05 mmol) and potassium acetate (135 mg, 1.37 mmol) in 1,4-dioxane ( 10 ml) and flushed with nitrogen. After heating at 100 °C for 6 h, the reaction mixture was cooled to room temperature, diluted with EtOAc, and washed with water and brine. The organic layer was dried over Na2SO4filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-100% EtOAc in DCM to give CB5 (179 mg, 99%) as an orange-brown residue. m/z=330.0 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 8,62 (d, J=6,1 Hz, 2H), 7,70-7,65 (m, 1H), 7,34 (d, J=5, 8 Hz, 2H), 6,74 (dd, J=2,4, 8,4 Hz, 1H), 6,63 (dd, J=2,3, 11,0 Hz, 1H), 5,10 ( s, 2H), 1,35–1,24 (m, 12H).

Method CB6 - 2-(5-Cyclobutyl-2-fluorophenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolan (CB6)

Step 1: 2-Bromo-4-cyclobutyl-1-fluorobenzene (CB6.1)

A solution of 2-bromo-1-fluoro-4-iodobenzene (CB6.0) (600 mg, 1.99 mmol), Xantphos Palladacycle Gen. 3 (95 mg, 0.10 mmol) and Xantphos (58 mg, 0 0.10 mmol) in THE (10 mL) was degassed and purged with nitrogen. After the addition of cyclobutylzinc bromide (0.5 M, 5.2 mL, 2.59 mmol), the reaction was heated at 50 °C for 7 h. After standing at room temperature for 3 days, the reaction mixture was quenched with saturated aqueous ammonium chloride and extracted into EtOAc (twice). The combined organic layer was washed with brine, dried over Na2SO4and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel eluting with 0-20% EtOAc in isohexane to give CB6.1 (385 mg, 84%) as a colorless oil.1H-RMN (400 MHz, CDCl3): δ 7,37 (dd, J=1,8, 6,6 Hz, 1H), 7,12–7,06 (m, 1H), 7,02 (dd, J=8,5, 8,5 Hz, 1H), 3,54–3,43 (m, 1H), 2,39–2,30 (m, 2H), 2,15–1,96 (m, 4H).

Etapa 2: 2-(5-ciclobutil-2-fluorofenil)-4,4,5,5-tetrametil-1,3,2-dioxaborolano (CB6)

A mixture of 2-bromo-4-cyclobutyl-1-fluorobenzene (CB6.1) (378 mg, 1.65 mmol), bis(pinacolato)diboron (503 mg, 1.98 mmol), [1,1'- bis(diphenylphosphine) Ferrocene]dichloropalladium(II) complex was degassed with DCM (135 mg, 0.17 mmol) and potassium acetate (405 mg, 4.12 mmol) in 1,4-dioxane (10 mL) and it was purged with nitrogen. After heating at 100 °C for 6 h, the reaction mixture was cooled to room temperature, diluted with EtOAc, and washed with water and brine. The organic layer was dried over Na2SO4filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-40% EtOAc in isohexane to give CB6 (286 mg, 63%) as a pale yellow oil.1RMN-H (400 MHz, CDCl3): δ 7,53 (dd, J=2,4, 5,6 Hz, 1H), 6,95 (dd, J=8,8, 8,8 Hz, 1H) , 3,57–3,46 (m, 1H), 2,37–2,28 (m, 2H), 2,18–1,95 (m, 3H), 1,89–1,83 (m, 1H), 1,36 (s, 12H).

Método CB7 - 2-(2-Fluoro-5-(1-methylcyclopropyl)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (CB7)

Step 1: 2-Bromo-1-fluoro-4-(prop-1-en-2-yl)benzene (CB7.1)

A suspension of methyltriphenylphosphonium bromide (1.73 g, 4.84 mmol) in THF (10 mL) was cooled to 0 °C and then treated portionwise with potassium tert-butoxide (0.57 g, 5.07 mmol). After stirring at 0 °C for 2 h, the reaction was quenched with the dropwise addition of a solution of 1-(3-bromo-4-fluorophenyl)ethane-1-one (CB7.0) (1.00 g, 4.61 mmol) treated) in DE (5 mL). After warming to room temperature and stirring at room temperature overnight, the reaction mixture was quenched with 1M aqueous HCl and extracted into EtOAc (twice). The combined organic layer was washed with brine, dried over Na2SO4filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-20% EtOAc in isohexane to give CB7.1 (352 mg, 36%) as a colorless oil.1RMN-H (400 MHz, CDCl3): δ 7,63 (dd, J=2,4, 6,7 Hz, 1H), 7,38–7,34 (m, 1H), 7,06 (dd, J=8,5, 8,5 Hz, 1H), 5,31 (s, 1H), 5,10 (s, 1H), 2,11 (s, 3H).

Step 2: 2-Bromo-1-fluoro-4-(1-methylcyclopropyl)benzene (CB7.2)

A solution of diethylzinc (1.0 M in hexane, 1.95 mL, 1.95 mmol) in DCM (5 mL) was placed under a nitrogen atmosphere and cooled to -15 °C. The solution was treated with the dropwise addition of a solution of dibutyl phosphate (0.39 mL, 1.95 mmol) in DCM (4 mL). After stirring at -15 °C for 15 min, the reaction was treated with diiodomethane (0.16 mL, 1.95 mmol). After stirring at -15 °C for 30 min, a solution of 2-bromo-1-fluoro-4-(prop-1-en-2-yl)benzene (CB7.1) (350 mg, 1.63 mmol ) in DCM (4 mL) was added. After warming to room temperature and stirring for 18 h, the reaction mixture was quenched with saturated aqueous ammonium chloride and extracted into EtOAc (twice). The combined organic layer was washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-30% EtOAc in isohexane to give CB7.2 (311 mg, 67%) as a colorless oil.1RMN-H (400 MHz, CDCl3): δ 7,36 (ddd, J=2,2, 4,7, 8,5 Hz, 1H), 7,17–7,10 (m, 2H), 2, 11 (s, 3H), 0,84–0,78 (m, 4H) .

Paso 3: 2-(2-Fluoro-5-(1-methylcyclopropyl)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (CB7)

A mixture of 2-bromo-1-fluoro-4-(1-methylcyclopropyl)benzene (CB7.2) (310 mg, 1.35 mmol), bis(pinacolato)diboron (412 mg, 1.62 mmol), [ 1,1'-Bis complex of (diphenylphosphino)ferrocene]dichloropalladium(II) with DCM (111 mg, 0.14 mmol) and potassium acetate (332 mg, 3.38 mmol) in 1,4-dioxane (10 mL ). degassed and flushed with nitrogen. After heating at 100 °C for 5 h, the reaction mixture was cooled to room temperature, diluted with EtOAc, and washed with water and brine. The organic layer was dried over Na2SO4filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-50% EtOAc in isohexane to give CB7 (357 mg) as an orange residue which was used without further purification in the next step.

Método CB8 - 1,1,1-Trifluoro-2-(4-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propan-2-ol (CB8)

Etapa 1: 2-(3-Bromo-4-fluorofenil)-1,1,1-trifluoropropan-2-ol (CB8.1)

A solution of 1-(3-bromo-4-fluorophenyl)ethan-1-one (CB8.0) (1.00 g, 4.61 mmol) and trimethyl(trifluoromethyl)silane (1.4 mL, 9.22 mmol) in THF (15 mL) was treated with the dropwise addition of tetrabutylammonium fluoride (1.0 M, 6.9 mL, 6.91 mmol). After stirring at room temperature overnight, the reaction mixture was quenched with saturated aqueous ammonium chloride and extracted into EtOAc (twice). The combined organic layer was washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-50% EtOAc in isohexane to give CB8.1 (1.39 g, quant) as a pale yellow oil.1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1H), 8,69 (s, 1H), 8,05 (dd, J=1,9, 7,5 Hz, 1H), 7 ,99 (d, J=2,8 Hz, 1H), 7,65-7,58 (m, 1H), 7,40 (dd, J=2,9, 9,0 Hz, 1H), 7, 35 (d, J=7,6 Hz, 2H), 2,43 (s, 6H).

Paso 2: 1,1,1-trifluoro-2-(4-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborlan-2-yl)phenyl)propan-2-ol (CB8)

A mixture of 2-(3-bromo-4-fluorophenyl)-1,1,1-trifluoropropan-2-ol (CB8.1) (521 mg, 1.82 mmol), bis(pinacolate)diboron (553 mg. 2.18 mmol ), complex of [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) with DCM (148 mg, 0.18 mmol) and potassium acetate (445 mg, 4.54 mmol) in 1 ,4-dioxane (10 ml) was degassed and purged with nitrogen. After heating at 100 °C for 5 h, the reaction mixture was cooled to room temperature, diluted with EtOAc and washed with water and brine. The organic layer was dried over Na2SO4filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-10% MeOH in DCM to give CB8 (348 mg) as an orange residue which was used in the next step without analysis.

CB9 method – (2-Etoxi-5-fluor-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)(piperidin-1-yl)methanone (CB9)

Step 1: (4-Bromo-5-fluoro-2-methoxyphenyl)(piperidin-1-yl)methanone (CB9.1)

A solution of 4-bromo-5-fluoro-2-methoxybenzoic acid (CB9.0) (215 mg, 0.82 mmol) in THF (5 mL) was treated with HOBt (144 mg, 1.06 mmol) and hydrochloride of EDC (204mg). mg) treated, 1.06 mmol). After stirring at room temperature for 1 hour, the reaction mixture was treated with DIPEA (0.43 mL, 2.45 mmol) and piperidine (0.10 mL, 0.98 mmol). After stirring at room temperature for 3 days, the reaction mixture was quenched with saturated aqueous sodium bicarbonate and diluted with DCM. The organic layer was separated, passed through a hydrophobic frit, and concentrated under reduced pressure to give CB9.1 (250 mg, 93%). m/z=318.0 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,07–7,00 (m, 2H), 3,81 (s, 3H), 3,78–3,61 (m, 4H), 3,21– 3,14 (m, 2H), 1,53–1,41 (m, 4H).

Step 2: (4-bromo-5-fluoro-2-hydroxyphenyl)(piperidin-1-yl)methanone (CB9.2)

A solution of (4-bromo-5-fluoro-2-methoxyphenyl)(piperidin-1-yl)methanone (CB9.1) (500 mg, 1.58 mmol) in DCM (30 mL) was brought to -78° C, refrigerated and treated with boron tribromide (1.0 M in DCM, 4.7 mL, 4.74 mmol). After stirring at -78 °C for 2 h, the reaction was quenched with water and neutralized with saturated aqueous sodium bicarbonate. The organic layer was separated, passed through a hydrophobic frit, and concentrated under reduced pressure to give CB9.2 (450 mg, 94%) as a tan solid. m/z=304.0 [M+H]+,1H-RMN (400 MHz, CDCl3): δ 7,37 (d, J=5,8 Hz, 1H), 6,97 (d, J=8,1 Hz, 1H), 3,58 (dd, J =5,2, 5,2 Hz, 4H), 3,21-3,14 (m, 2H), 1,53-1,41 (m, 4H).

Stage 3: (4-Bromo-2-ethoxi-5-fluorofenil)(piperidin-1-yl)methanone (CB9.3)

To a solution of (4-bromo-5-fluoro-2-hydroxyphenyl)(piperidin-1-yl)methanone (CB9.2) (220 mg, 0.73 mmol) in DMF (5 mL) was added carbonate of potassium (1.00 g), 7.28 mmol) and bromoethane (0.12 mL, 1.60 mmol). After stirring at 60 °C for 3 h, the reaction was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure to give crude CB9.3 (250mg) which was used in the next step without further purification. m/z=332.0 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,06–7,00 (m, 2H), 4,08–3,96 (m, 2H), 3,75–3,62 (m, 3H), 3,23–3,09 (m, 4H), 1,89 (s, 2H), 1,45-1,35 (m, 4H).

Step 4: (2-Ethoxy-5-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)(piperidine-1-yl)methanone (CB9)

A mixture of (4-bromo-2-ethoxy-5-fluorophenyl)(piperidin-1-yl)methanone (CB9.3) (250 mg, 0.76 mmol), bis(pinacolato)diboron (211 mg, 0. 83 mmol), complex of [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) with DCM (28 mg, 0.04 mmol) and potassium acetate (223 mg, 2.27 mmol) in 1.4 -dioxane (10 ml) was degassed and flushed with nitrogen. After stirring at 110°C overnight, the reaction mixture was cooled to room temperature, quenched with saturated aqueous ammonium chloride, and filtered. The filtrate was extracted into DCM and the organic layer was passed through a hydrophobic frit and concentrated under reduced pressure to give crude CB9, which was used in the next step without further purification. m/z=378.3 [M+H]+.

Method CB10 – (2,3-Difluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl)boros (CB10)

A solution of (2,3-difluoro-6-methoxyphenyl)(piperidin-1-yl)methanone (CB10.0) (2.30 g, 9.01 mmol) in THF (40 mL) was degassed and purged with nitrogen, heated to - cooled to 78 °C and then treated with the dropwise addition of n-butyllithium (2.50 M in hexanes, 3.6 mL, 9.01 mmol). After stirring at -78 °C for 30 min, trimethyl borate (1.2 mL, 10.8 mmol) was added dropwise. After stirring for 16h, warming to room temperature, the reaction was quenched with 1M aqueous HCl (40ml) and extracted into EtOAc (3 x 50ml). The combined organic layer was washed with brine (40 mL), dried over MgSO4, filtered, and concentrated under reduced pressure to give CB10 (2.76 g, quantitative) as an off-white solid. m/z=299.9 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,04 (s, 1H), 5,13–5,05 (m, 2H), 3,84 (s, 3H), 3,82–3,76 ( m, 1H), 3,71–3,67 (m, 1H), 3,21 (q, J = 5,1 Hz, 2H), 1,67–1,63 (m, 4H), 1,25 (s, 2H).

Method CB11 - 1-(2-(4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)pyrrolidine-1-yl)ethane-1- one (CB11)

Step 1: (4-(3-Bromo-4-fluorophenyl)-4-oxobutyl)carbamate (tert-butyl) (CB11.1)

A solution of 2-bromo-1-fluoro-4-iodobenzene (CB11.0) (3.00 g, 9.97 mmol) in THF (75 mL) was cooled to -40 °C and treated with dropwise addition. a drop of isopropylmagnesium chloride (2.0 M, 5.0 mL, 9.97 mmol). After stirring at -40°C for 1 hour and then cooling to -78°C, tert-butyl 2-oxopyrrolidine-1-carboxylate (1.85 g, 9.97 mmol) was added. After warming to room temperature for 90 minutes, the reaction was quenched with saturated aqueous ammonium chloride and extracted into EtOAc. The organic layer was washed with brine, passed through a hydrophobic frit, and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-60% EtOAc in cyclohexane to give CB11.1 (1.50 g, 42%) as a brown oil which solidified on standing. m/z=362.2 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 8,18 (dd, J=1,8, 6,6 Hz, 1H), 7,94–7,89 (m, 1H), 7,19 (dd, J=8,3, 8,3 Hz, 1H), 4,70–4,59 (m, 1H), 3,26–3,22 (m, 2H), 2,98 (dd, J=7, 1, 7,1 Hz, 2H), 1,99–1,88 (m, 2H), 1,27–1,14 (m, 9H).

Step 2: 2-(3-Bromo-4-fluorophenyl)pyrrolidine (CB11.2)

Trifluoroacetic acid (15 mL) was added given, 196 mmol). After stirring at room temperature for 30 min, the solvent was removed in vacuo. The crude residue was taken up in THF (30ml) and treated with trimethylamine (0.70ml, 5.00mmol) followed by sodium triacetoxyborohydride (2.65g, 12.5mmol). After 2 h, sodium cyanoborohydride (392 mg, 6.24 mmol) was added. After stirring for 30 min, the reaction was quenched with saturated aqueous sodium bicarbonate and extracted into EtOAc. The organic layer was separated, passed through a hydrophobic frit, and concentrated under reduced pressure to provide CB11.2 as an oil that was used in the next step without further purification. m/z=246.1[M+H]+.

Step 3: 1-(2-(3-Bromo-4-fluorophenyl)pyrrolidin-1-yl)ethan-1-one (CB11.3)

To a solution of 2-(3-bromo-4-fluorophenyl)pyrrolidine (CB11.2) (510 mg, 2.0 mmol) in THF (20 mL) and triethylamine (350 mL, 2.5 mmol) was added chloride of acetyl (220 mL) administered, 3.1 mmol). After stirring at room temperature for 4 h, the reaction was quenched with saturated NaHCO3.3solution and extracted with ethyl acetate. The organic layer was washed with brine, dried (MgSO4), filtered and concentrated under reduced pressure. The crude residue was purified by chromatography on silica gel using 0-60% ethyl acetate to give CB11.3 (310 mg, 52%) as a clear, colorless oil.1RMN-H (400 MHz, CDCl3) 7,38–7,28 (1H, m), 7,12–7,02 (2H, m), 5,14–4,86 (1H, m), 3, 77–3,57 (2H, m), 2,46–2,21 (1H, m), 2,13 (3H, s), 2,01-1,85 (3H, m).

Stage 4: 1-(2-(4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)pyrrolidine-1-yl)ethan-1- um (CB11)

The desired product was prepared according to the representative procedure described in Method CB3 using 1-(2-(3-bromo-4-fluorophenyl)pyrrolidin-1-yl)ethane-1-one (CB11.3) to give the product (CB11)) as a reddish-brown oil which was used in the next step without further purification.

Método CB12 - 6-(4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)selfish)-2-oxa-6-azaspiro[3.3]heptano ( CB12)

Etapa 1: 6-(3-Bromo-4-fluorobenzil)-2-oxa-6-azaspiro[3.3]heptano (CB12.1)

To a solution of 3-bromo-4-fluorobenzaldehyde (CB12.0) (250 mg, 1.23 mmol) in 2-propanol (10 mL) was added 2-oxa-6-azaspiro[3,3]heptane ( 244 mg, 2nd 46 mg) mmol) and titanium(IV) isopropoxide (0.53 mL, 1.79 mmol). After stirring at room temperature overnight, the reaction was treated with sodium borohydride (47 mg, 1.23 mmol). After stirring at room temperature for 1 h, the reaction was quenched with 6 M aqueous HCl, stirred at room temperature for 2 h, and neutralized to pH ~10 with 4 M aqueous NaOH. The reaction mixture was extracted into diethyl ether and The organic mixture layer was dried with MgSO4, filtered, and concentrated under reduced pressure. The starting material was loaded onto an SCX cartridge and diluted with 10% (7N NH3in MeOH) in DCM to give CB12.1 (234 mg, 66%) as a colorless oil. m/z=288.0 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,48–7,43 (m, 1H), 7,16–7,11 (m, 1H), 7,05 (dd, J=8,3, 8 ,3 Hz, 1H), 4,74 (s, 4H), 3,47 (s, 2H), 3,35 (s, 4H).

Schritt 2: 6-(4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)selfish)-2-oxa-6-azaspiro[3.3]heptano ( CB12)

To a degassed solution of 6-(3-bromo-4-fluorobenzyl)-2-oxa-6-azaspiro[3,3]heptane (CB12.1) (234 mg, 0.82 mmol) in 1,4-dioxane (5 mL) in a large carousel tube were bis(pinacolato)diboron (311 mg, 1.23 mmol), potassium acetate (241 mg, 2.45 mmol), and [1,1'-bis(diphenylphosphino)ferrocene ]dichloropalladium(II) (30 mg, 0.04 mmol). The reaction mixture was degassed and purged with nitrogen. After shaking the sealed tube at 115 °C for 3 h, the reaction mixture was cooled to room temperature and compound (CB12) was carried on to the next step without further purification.

Method CB13 - 1,1,1-Trifluoro-2-(4-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-N,N- Dimethylpropan-2-amine (CB13)

Stage 1: N-(2-(3-Bromo-4-fluorophenyl)-1,1,1-trifluoropropan-2-yl)acetamida (CB13.1)

A solution of 1-(3-bromo-4-fluorophenyl)ethane-1-one (CB13.0) (1.00 g, 4.61 mmol) in THF (15 mL) was placed under a nitrogen atmosphere and then washed with trimethyl (trifluoromethylsilane (0.82 mL, 5.53 mmol) and tetrabutylammonium fluoride (1 M in THF, 0.23 mL, 0.23 mmol). After stirring at room temperature for 24 h, the mixture of reaction was treated with additional trimethyl(trifluoromethyl)silane (0.82 mL, 5.53 mmol) After stirring at room temperature for 2 h, the solvent was evaporated in vacuo and the crude residue was taken up in acetonitrile (10 mL). The solution was treated with sulfuric acid (1.0 mL, 18.8 mmol) and heated at 80 °C for 24 h.The reaction mixture was cooled to room temperature, neutralized by the dropwise addition of sodium bicarbonate. saturated aqueous sodium and extracted into DCM (3 x 20 mL).The combined organic layer was washed with brine (30 mL), filtered through a hydrophobic frit, and concentrated under reduced pressure.The crude material was purified by Silica gel chromatography using 0-60% EtOAc in cyclohexane to give CB13.1 (275 mg, 18%) as a white solid. m/z=329.9 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,60 (d, J=4,8 Hz, 1H), 7,39-7,33 (m, 1H), 7,11 (dd, J=8, 5, 8,5 Hz, 1H), 6,05 (s, 1H), 2,05 (d, J=19,7 Hz, 6H).

Step 2: N-(2-(3-Bromo-4-fluorophenyl)-1,1,1-trifluoropropan-2-yl)-N-methylacetamide (CB13.2)

To a suspension of sodium hydride (60% w/w dispersion in mineral oil, 34 mg, 0.84 mmol) in DMF (3 mL) was added N-(2-(3-bromo-4-fluorophenyl) -1,1,1-trifluoropropan-2-yl)acetamide (CB13.1) (250 mg, 0.76 mmol). After stirring at room temperature for 2 h, iodomethane (0.05 mL, 0.84 mmol) was added. After heating at 85 °C for 16 h, the reaction was cooled to room temperature and quenched with 4% aqueous lithium chloride (10 mL). The mixture was extracted into DCM (3 x 15 mL) and the combined organic layer was washed with brine (20 mL), passed through a hydrophobic frit, and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-60% EtOAc in cyclohexane to give CB13.2 (120 mg, 46%) as a colorless oil. m/z=343.9 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,50 (d, J=5,1 Hz, 1H), 7,08 (dd, J=8,3, 8,3 Hz, 2H), 3,16 (s, 3H), 2,10 (s, 3H), 1,54 (s, 3H).

Step 3: 2-(3-Bromo-4-fluorophenyl)-1,1,1-trifluoro-N-methylpropan-2-amine (CB13.3)

A solution of N-(2-(3-Bromo-4-fluorophenyl)-1,1,1-trifluoropropan-2-yl)-N-methylacetamide (CB13.2) (115 mg, 0.34 mmol) in ethanol (2 ml) and water (0.2 ml) was treated with aqueous hydrochloric acid (11 M, 0.15 ml, 1.68 mmol). After stirring at 100 °C for 4 days, the reaction was cooled to room temperature and the solvent was removed under reduced pressure to give CB13.3 (89 mg, 88%) as a white solid, which was used in the next step. without further ado Cleanup. m/z = 302.0 [M+H]+.

Schritt 4: 1,1,1-trifluoro-2-(4-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-N-methylpropane- 2-amine (CB13)

A mixture of 2-(3-bromo-4-fluorophenyl)-1,1,1-trifluoro-N-methylpropan-2-amine (CB13.3) (89 mg, 0.30 mmol), bis(pinacolato)diboron (90 mg, 0.36 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) complex with DCM (12 mg, 0.01 mmol), and potassium acetate (87 mg, 0.89 mmol) in 1,4-dioxane (4 mL) was degassed and purged with nitrogen. After stirring at 100 °C for 2.5 h, the reaction was cooled to room temperature, filtered through a pad of Celite® (2.5 g) and eluted with DCM. The filtrate was concentrated under reduced pressure to give crude CB13, which was used in the next step without further purification.

Method CB14 - (2-Cyclopropyl-5-fluor-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)(piperidin-1-yl)methanone (CB14)

Stage 1: 4-bromo-5-fluoro-2-iodobenzoic acid (CB14.1)

Potassium permanganate (5.02 g, 31.8 mmol) was added to a solution of 4-bromo-5-fluoro-2-iodotoluene (CB14.0) (1.00 g, 3.18 mmol) in water ( 12 mL) and pyridine (8 mL). . After stirring at 100°C overnight, the reaction was allowed to come to room temperature, filtered through a pad of Celite and washed with water (20ml). The aqueous layer was washed with EtOAc (2 x 15 mL), then acidified with 2N HCl to pH ~1. The cloudy suspension was extracted with EtOAc (2 x 15 mL). The combined organic extracts were washed with water (20ml) and brine (20ml), dried over MgSO4, filtered and concentrated under reduced pressure. The desired product (CB14.1) was isolated as a white solid and used for the next step without further purification (520 mg, 47%), m/z = 344.9 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,6 (s, 1H), 8,37 (d, J=6,8 Hz, 1H), 7,76 (d, J=9,3 Hz, 1H ).

Step 2: Methyl 4-bromo-5-fluoro-2-iodobenzoate (CB14.2)

Sulfuric acid (0.04 mL, 0.754 mmol) was added to a solution of (CB14.1) (0.52 g, 1.51 mmol) in MeOH (0.8 mL). After stirring at 65 °C overnight, the reaction was quenched with saturated aqueous NaHCO3.3hydrochloric acid (10ml) and extracted with EtOAc (3 x 8ml). The organic solvent was evaporated under reduced pressure to give the desired compound (CB14.2) as a crystalline solid (420 mg, 78%). m/z=358.9 [M+H]+,1H-RMN (400 MHz, CDCl3): δ 8,20 (d, J=6,6 Hz, 1H), 7,62 (d, J=8,8 Hz, 1H), 3,90 (s, 3H ).

Stage 3: Methyl 4-bromo-5-fluoro-2-vinylbenzoate (CB14.3)

A degassed solution of (CB14.2) (500 mg, 1.39 mmol), potassium vinyl trifluoroborate (220 mg, 1.16 mmol), cesium carbonate (910 mg, 2.79 mmol) and [1, 1'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (100 mg, 0.14 mmol) in 1,4-dioxane (20 mL) and water (2 mL) was stirred at 100 °C overnight. After cooling to room temperature, the reaction mixture was filtered through a pad of celite and the solvent was evaporated. The crude product was purified by chromatography on silica gel eluting with 0-60% EtOAc in cyclohexane to give the title compound (CB14.3) (200 mg, 56%) as a clear solid. m/z=259.1 [M+H]+,1H-RMN (400 MHz, CDCl3): δ 7,76 (d, J=6,6 Hz, 1H), 7,65 (d, J=9,1 Hz, 1H), 7,39 (d, J =6,6 Hz, 1H), 5,60 (d, J=17,4 Hz, 1H), 5,38 (d, J=11,1 Hz, 1H), 3,90 (s, 3H).

Step 4: Methyl 4-bromo-2-cyclopropyl-5-fluorobenzoate (CB14.4)

A degassed solution of diethylzinc (1.5 mL, 1.54 mmol) in dry DCM (2 mL) was treated at 0 °C with a degassed solution of TFA (0.12 mL, 1.54 mmol) in dry DCM ( 1st ml). After stirring at 0 °C for 20 min, diiodomethane (0.12 mL, 1.54 mmol) in dry DCM (2 mL) was added. After stirring at 0 °C for 20 min, a solution of (CB14.3) (200 mg, 0.777 mmol) in dry DCM (1 mL) was added. After stirring at room temperature for 120 min, the reaction was quenched with NH4Cl (5mL). The organic phase was collected, concentrated and purified on silica gel (eluting with 0-70% EtOAc in cyclohexane) to give the title compound (CB14.4) (200 mg, 95%) as a pale solid. m/z=273.1 [M+H]+,1H-RMN (400 MHz, CDCl3): δ 7,56 (d, J=8,8 Hz, 1H), 7,20 (d, J=6,6 Hz, 1H), 3,91 (s, 3H ), 2,65–2,53 (m, 1H), 1,04-0,95 (m, 2H), 0,64 (q, J = 5,4 Hz, 2H).

Step 5: 4-bromo-2-cyclopropyl-5-fluorobenzoic acid (CB14.5)

To a solution of (CB14.4) (200 mg, 0.732 mmol) in dry THF (10 mL) was added LiOH (2 M in water, 0.81 mL, 1.61 mmol). After stirring at room temperature overnight, the resulting precipitate was collected and used for the next step without further purification, m/z = 259.1 [M+H]+.

Step 6: (4-bromo-2-cyclopropyl-5-fluorophenyl)(piperidin-1-yl)methanone (CB14.6)

To a stirred solution of (CB14.5) (120 mg, 0.463 mmol) in dry THF (5 mL) was added 1-hydroxybenzotriazole hydrochloride (81 mg, 0.60 mmol), N-(3-dimethylaminopropyl)- N'-ethylcarbodiimide, administered (115 mg, 0.60 mmol). After stirring at room temperature for 1 h, DIPEA (0.24 mL, 1.39 mmol) and piperidine (0.05 mL, 0.55 mmol) were added. After stirring at room temperature for 3h, the solvent was removed under reduced pressure. The crude residue was purified by chromatography on silica gel eluting with 0-60% EtOAc in cyclohexane to give the title compound (CB14.6) (74 mg, 49%) as a clear oil. m/z=326.2 [M+H]+,1RMN-H (400 MHz, CDCl3): δ 7,06 (d, J=6,3 Hz, 1H), 6,93 (d, J=8,1 Hz, 1H), 3,79-3,61 (m, 2H), 3,20 (dd, J=5,4 , 5,4 Hz, 2H), 1,92–1,79 (m, 1H), 1,67 (t, J=11,6 Hz, 4H), 1,50 (d, J=4,3 Hz, 4H), 1,01–0,75 (m, 2H).

Schritt 7: (2-cyclopropyl-5-fluor-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)(piperidin-1-yl)methanone (CB14)

To a mixture of (CB 14.6) (74 mg, 0.23 mmol), bis(pinacolato)diboron (69 mg, 0.27 mmol) and potassium acetate (67 mg, 0.68 mmol) in 1,4- dioxane (4 mL) under an atmosphere of N2[1,1'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) complex with DCM (9.3 mg, 0.01 mmol) was added. After stirring at 100 °C for 3.5 h, the reaction was cooled to room temperature, filtered through celite, and extracted with DCM (2 x 10 mL). The combined organic layer was concentrated in vacuo to give the product (CB 14) which was used without further purification for the next step (81 mg, 95%), m/z = 373.2 [M+H]+.

The following boronate intermediates were prepared according to any of Intermediate Procedures CB 1-CB 14 as described above.

intermediary CB Structure representative synthetic procedure CB15 Method CB1 using 4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoic acid and azetidine (Step 2) CB16 Method CB3 using 2-bromo-4-(tert-butyl)-1-fluorobenzene CB17 CB3 method using 2-(3-bromo-4-fluorophenyl)acetonitrile CB18 CB3 method using 2-(3-bromo-4-fluorophenyl)-2-methylpropanenitrile CB19 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and cyclohexanecarbonyl chloride (Step 1) CB20 Method CB14 with 4-bromo-3-fluoro-2-methylbenzoic acid and piperidine (steps 6 and 7) CB21 Method CB14 with 4-bromo-5-fluoro-2-methylbenzoic acid and piperidine (steps 6 and 7) CB22 CB3 method using 2-(3-bromo-4-fluorophenyl)propan-2-ol CB23 Method CB3 using tert-butyl 6-bromo-5-fluoro-1H-indole-1-carboxylate CB24 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and cyclobutanecarbonyl chloride (Step 1) CB25 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-methylcyclopropane-1-carbonyl chloride (Step 1) CB26 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and cyclopentanecarbonyl chloride (Step 1) CB27 Method CB14 using 5-bromo-4-fluoro-2-methoxybenzoic acid and piperidine (steps 6 and 7) CB28 Method CB14 using 4-bromo-3-fluoro-2-methoxybenzoic acid and piperidine (steps 6 and 7) CB29 Method CB14 with 5-bromo-4-fluoro-2-methylbenzoic acid and piperidine (steps 6 and 7) CB30 Method CB3 using tert-butyl 5-bromo-6-fluoro-3-methyl-1H-indazol-1-carboxylate CB31 CB3 method using 5-bromo-6-fluoro-2-methylbenzo[d]oxazole CB32 Method CB2 using 3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline chloride and bicyclo[2.2.1]heptane-2-carbonyl CB33 CB9 method using 2-iodopropane CB34 Método CB1 utilizando metil 2-methoxy-4-(4,4,5,5-tetrametil-1,3,2-dioxaborolan-2-yl)benzoato y piperidina (Etapa 2) CB35 Method CB3 with CB4.1 CB36 Method CB2 using 5-Fluoro-2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and cyclohexanecarbonyl chloride (Step 1) CB37 CB4 method with methylamine (Step 2) CB38 Method CB3 using 5-bromo-4-fluoro-2,3-di-hydro-1H-inden-l-ol CB39 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 3-methyloxetane-3-carbonyl chloride (Step 1) CB40 Method CB1 using methyl 5-fluoro-2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoate and pyrrolidine CB41 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 3-methyloxetane-3-carbonyl chloride (Step 1) CB42 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-methylcyclobutane-1-carbonyl chloride (Step 1) CB43 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-methylcyclopropane-1-carbonyl chloride (Step 1) CB44 Method CB3 using tert-butyl (4-bromo-3-fluorobenzyl)(methyl)carbamate CB45 Method CB3 using tert-Butyl 5-bromo-6-fluoro-3,4-dihydroisoquinoline-2(1H)-carboxylate CB46 Method CB3 using 5-bromo-6-fluoro-2-methyl-1,2,3,4-tetrahydroisoquinoline CB47 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-(difluoromethyl)cyclopropane-1-carbonyl chloride CB48 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-ethylcyclopropane-1-carbonyl chloride CB49 Method CB3 using tert-butyl (5-bromo-6-fluoro-1,2,3,4-tetrahydronaphthalen-1-yl)(methyl)carbamate CB50 Método CB1 utilizando metil 5-fluoro-2-methoxy-4-(4,4,5,5-tetrametil-1,3,2-dioxaborolan-2-yl)benzoato y dimethylamina CB51 Method CB14 using 4-bromo-2,5-difluorobenzoic acid and piperidine (steps 6 and 7) CB52 Method CB4 using 5-bromo-4-fluoro-2,3-di-hydro-1H-inden-1-ol CB53 Method CB4 using 5-bromo-6-fluoro-2,3-di-hydro-1H-inden-1-ol CB54 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-ethylcyclobutane-1-carbonyl chloride CB55 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and 1-ethylcyclobutane-1-carbonyl chloride CB56 Method CB4 with Piperidine (Step 2) CB57 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and endo-norbornanecarboxylic acid chloride CB58 CB4 Method with Morpholine (Step 2) CB59 Method CB12 with 2-azaspiro[3.3]heptane (step 1) CB60 CB12 method using 5-oxa-2-azaspiro[3.4]octane (Step 1) CB61 Method CB12 using 6-oxa-2-azaspiro[3.4]octane (stage 1) CB62 Method CB4 with 2-azaspiro[3.3]heptane (step 2) CB63 Method CB4 using 5-bromo-6-fluoro-2,3-di-hydro-1H-inden-1-ol and morpholine (step 2) . CB64 Method CB4 using 6-oxa-2-azaspiro[3.4]octane (Step 2) CB65 Method CB2 using 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline and exo-norbornanecarboxylic acid chloride CB66 Method CB2 with 5-fluoro-2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborlan-2-yl)anilinecyclohexanecarbonyl chloride (Step 1) CB67 Method CB1 using 6,6-difluoro-3-azabicyclo[3.1.0]hexane (step 2) CB68 CB12 method with methylamine (Step 1) CB69 Method CB3 with tert-butyl (1-(3-bromo-4-fluorophenyl)cyclopropyl)(methyl)carbamate CB70 Method CB3 using tert-butyl 2-(3-bromo-4-fluorophenyl)pyrrolidine-1-carboxylate

Method PB1 - Preparation of N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)cyclohexanecarboxamide (PB1)

Step 1. From N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)cyclohexanecarboxamide (PB1)

For a solution of 3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (CB2.0) (10 g, 0.0421 mol, 1, 0 eq.) in DCM (60 mL) and diisopropylethylamine (15 mL) was added to cyclohexanecarbonyl chloride (509 mL, 0.08435 mol, 2.0 eq.). After stirring at room temperature for 16 h, the reaction mixture was diluted with DCM (500 mL) and washed with saturated sodium bicarbonate (300 mL) and then water (300 mL). The combined organic layer was washed with brine (100ml), dried over sodium sulphate and concentrated under reduced pressure to give the crude material, which was purified by trituration using diethyl ether (2x30ml) to give the title compound PB1 (12.5g, 85.35%). ). ). MS (ES): m/z 348.2 [M+H]+.

PB2 Method - Preparation of 4-(3-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-3-one (PB2)

Etapa 1. 4-(3-Bromofenil)morpholina-3-one (PB2.1)

To a solution of morpholin-3-one (1 g, 9.90 mmol, 1.0 eq.) in 1,4-dioxane (100 mL) was added 1,3-dibromobenzene PB2.0 (2.6 g, 11.4 mmol, 1.1 eq.) at room temperature. The reaction mixture was degassed under argon for 10 min, followed by the addition of cesium carbonate (6.2 g, 19.8 mmol, 2.0 eq), (palladium acetate)Pd(OAc)2(0.21 g, 0.99 mmol, 0.1 eq.) and 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (Xanthphos) (0.8 g, 7.5 mmol, 0.1 eq. .). The reaction mixture was heated at 100 °C for 16 h in three parallel reactions. After the reaction was complete, the reaction mixture was cooled to room temperature, combined, filtered, and concentrated under reduced pressure. The crude product was subjected to flash chromatography eluting with 0-30% ethyl acetate in hexane. The solvent was evaporated under reduced pressure to give the desired product (PB2.1) (3.0 g, 39.48%). MS (ES): m/z 258.1 [M+2H]+

Paso 2. 6-4-(3-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-3-one (PB2)

To a solution of 4-(3-bromophenyl)morpholin-3-one (PB2.1) (3.0 g, 11.7 mmol, 1.0 eq.) in 1,4-dioxane (30 mL) was added bis(pinacolato)diborane (3.5 g, 14.0 mmol, 1.2 equiv.). The reaction mixture was degassed under argon for 10 min, followed by the addition of potassium acetate (3.4 g, 35.1 mmol, 3.0 eq.) and [1,1'-bis(diphenylphosphino) ferrocene]palladium(II) (Pd(dppf)Cl2) (0.85 g, 1.17 mmol, 0.1 eq.). After stirring at 100 °C for 4 h, the reaction mixture was cooled to room temperature, filtered, and concentrated in vacuo. The crude product was passed through flash chromatography eluting with 10-80% ethyl acetate in hexane. The solvent was evaporated under reduced pressure to give the desired product PB2 (3.2 g, yield: 90.11%). MS (ES): m/z 304.1 [M+H]+

Method PB3 - Preparation of 6-Amino-2-fluoro-N,N-dimethyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzamide (PB3)

Etapa 1. Metil 2-amino-6-fluorobenzoato (PB3.1)

To a solution of 2-amino-6-fluorobenzoic acid (2 g, 12.892 mmol, 1.0 eq.) in DCM (20 mL) and methanol (5 mL) at 0 °C was added (trimethylsilyl)diazomethane (0 0.6 M) (22 mL, 12.9 mmol, 1.0 eq) under a nitrogen atmosphere. After stirring at room temperature for 3 h, the reaction mixture was slowly quenched with acetic acid until bubbling ceased. The reaction mixture was diluted with DCM (50 mL) and washed with saturated sodium bicarbonate solution (50 mL). The combined organic layer was dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash column chromatography eluting with 2% ethyl acetate in hexane to give PB3.1 (2.1 g, 96.29%). MS (ES): m/z 170.1 [M+H]+

Etapa 2. Metil 6-amino-2-fluoro-3-bromobenzoato (PB3.2)

To a solution of methyl-2-amino-6-fluorobenzoic acid PB3.1 (2.1 g, 12.416 mmol, 1.0 eq) in ethanol (20 mL) was added silver sulfate (2.1 g, 14.899 mmol, 1.2 eq) and bromine in ethanol (2.98 g, 18.62 mmol, 1.5 eq) was added dropwise at 0 °C. After stirring at room temperature for 3h, the reaction mixture was quenched with sodium thiosulfate and ethyl acetate (100ml). The mixture was filtered through a pad of celite, and the combined organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash column chromatography eluting with 4% ethyl acetate in hexane to give PB3.2 (1.5 g, 48.71%). MS (ES): m/z 249.1 [M+H]+

Stage 3. 6-amino-2-fluoro-3-bromo-N,N-dimethylbenzamide (PB3.3)

To a solution of methyl 6-amino-3-bromo-2-fluorobenzoate PB3.2 (1.4 g, 5.645 mmol, 1.0 eq.) in toluene (10 mL) was added dimethylamine (2.0 M in THF) (4.8 mL, 9.596 mmol), 1.7 eq) trimethylaluminum (2.0 M in THF) (4.8 mL, 9.596 mmol, 1.7 eq) f. After 16 h it is stirred at room temperature. the reaction mixture was quenched with cold water (100 ml) and ethyl acetate (150 ml). The mixture was filtered through a pad of celite, and the combined organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash column chromatography eluting with 75% ethyl acetate in hexane to give PB3.3 (0.45 g, 30.54%), MS (ES): m/z 262.1 [M+ H ]+

Paso 4. 6-amino-2-fluoro-N,N-dimethyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzamide (PB3)

For a solution of methyl 6-amino-3-bromo-2-fluoro-n,n-dimethylbenzamide fluorobenzoate PB3.3 (0.4 g, 1.532 mmol, 1.0 eq) in 1,4-dioxane (8 mL) se g, 4.597 mmol, 3.0 eq.) and potassium acetate (0.45 g, 4.597 mmol, 3.0 eq.). After stirring and degassing with argon gas for 15 min, chloro(2-dicyclohexylphosphine-2',4',6'-triisopropyl-1,1'-biphenyl)[2-(2'-amino-1,1'- biphenyl) ) ] Palladium(II) (xphos PdG2) (0.12 g, 0.153 mmol, 0.1 eq) at room temperature. After stirring at 120 °C for 2.5 h, the reaction mixture was quenched with water (50 mL). The aqueous layer was washed with ethyl acetate (100 ml x 3). The combined organic layer is dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash column chromatography eluting with 2% methanol in DCM to give PB3 (0.4 g, 84.73%). MS (ES): m/z 309.1 [M+H]+

Method PB4 - Preparation of 1-ethyl-N-(3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)cyclobutane-1-carboxamide (PB4 )

Step 1. 1-Ethylcyclobutane-1-carboxylic acid (PB4.1)

To a stirred solution of cyclobutanoic acid (1 g, 5.0 mmol, 1 eq) in dry THF was added lithium diisopropylamide (2 M in THF) (10 mL, 20.00 mmol, 2.0 eq) at 0 °C in argon atmosphere. After stirring at 0 °C for 2 h, ethyl iodide (1.7 g, 11 mmol, 1.1 eq) was added. After stirring at room temperature for 16 h, the reaction mixture was poured into water (50 mL), extracted with ethyl acetate (50 mL x 3). The combined organic layer was washed with brine (30 mL), dried over sodium sulfate, and concentrated under reduced pressure to give PB4.1 which was used as such for the next step without purification. (1.2g, 100%).1RMN-H (DMSO-d6, 400 MHz): 12,08 (s, 1H), 1,82 (m, 4H), 1,60 (m, 2H), 1,48 (q, 2H), 0,94 (t, 3H).

Schritt 2. 1-Ethyl-N-(3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)cyclobutane-1-carboxamide (PB4)

To a stirred solution of PB4.1 (1.2 g, 9.375 mmol, 1.2 eq.) in THF were added phosphorous oxychloride and pyridine at 0 °C. After stirring at 0°C for 20 min, a solution of 3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (CB2.0) ( 2.554 g, 7.812 mmol, 1.0 eq.) in THF was added dropwise. After stirring at room temperature for 1 h, the reaction mixture was poured into water (100 mL), neutralized with sodium bicarbonate, and extracted with ethyl acetate (100 mL x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by combined flash chromatography eluting with 9% ethyl acetate in hexane to give pure PB4. (0.350g, 10.75%). MS(ES): m/z 348.24 [M+H]+.

Preparation of Compounds of the Invention Example 1: Method A - Preparation of 2-(2-Fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin- 5(6H)-one (I-42)

Step 1: 2,4-dichloro-1,6-naphthyridin-5(6H)-one(1.1)

To a solution of ethyl 4,6-dichloro-2-methylnicotinate (1.0) (20 g, 85.4 mmol) in tert-butanol (200 mL) was added 1,3,5-triazine (13, 8 g, 170.8 mmol) and potassium was administered. -butoxide (19.1 g, 170.8 mmol). After stirring at 90°C for 1 hour, the mixture was cooled to room temperature and evaporated under reduced pressure. The resulting residue was diluted with water (300 mL) and then acidified to ~pH 5 using 2M aqueous HCl solution. After stirring the resulting suspension at room temperature for 1 h, the solid was collected by filtration and washed with water. The crude solid was slurried in 4:1 DCM:acetonitrile (250 mL) and stirred for 20 minutes. The solid was collected by filtration and washed with 1:1 DCM:diethyl ether (100 mL). Residual water was removed from the solid by azeotroping with toluene (200 mL). Compound (1.1) was obtained as a brown solid (9.43 g, 51%) which was used without further purification. m/z=215.9 [M+H]+,1H-RMN (400 MHz, DMSO): δ 11,82 (s, 1H), 7,75 (s, 1H), 7,60 (t, J=5,9 Hz, 1H), 6,59 (d , J=7,1 Hz, 1H).

Schritt 2. 2-chloro-4-((5-(4-hidroxipiperidin-1-yl)pyridin-2-yl)amino)-1,6-naftiridin-5(6H)-ona (1,2)

In a 10-20 mL microwave tube filled with 2,4-dichloro-1,6-naphthyridin-5(6H)-one (1,1) (1.61 g, 7.5 mmol) in n -butanol (12.0 mL) and N ,N-diisopropylethylamine (2.6 mL, 15 mmol) was added to 1-(6-aminopyridin-3-yl)piperidin-4-ol (preparation described in WO2015131080, 1, 59 g, 8.25 mmol) provided. After heating in a Biotage Initiator® microwave at 160°C for 6 hours, the precipitate was isolated by filtration and washed sequentially with ethyl acetate (15 ml) and diethyl ether (40 ml). The reaction was carried out three times in parallel on the same scale and combined to give crude product (1.2) (5.02 g, 60%) as a brown solid which was used without further purification. m/z=372.5 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,57 (s, 1H), 11,90 (d, J=3,8 Hz, 1H), 8,43 (s, 1H), 8,20-8 ,15 (m, 1H), 7,53 (d, J =6,6 Hz, 2H), 7,07-7,02 (m, 1H), 6,53 (d, J=7,1 Hz, 1H), 4,76-4,72 (m, 1H), 3,70 (dd, J=3,4, 4,2 Hz, 1H), 3,61 (s, 2H), 2,98–2 ,88 (m, 2H), 1,89–1,87 (m, 2H), 1,57–1,50 (m, 2H).

Schritt 3 – 2-(2-Fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthiridin-5(6H)- ein (I-42)

A 2-5 mL microwaveable flask filled with 2-Chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H )- um ( 1.2) (100 mg, 0.269 mmol), 2-fluoro-5-methoxyphenylboronic acid (69 mg, 0.403 mmol) in dioxane (4.0 mL), water (0.5 mL), and K3AFTER4(228 mg, 1.08 mmol) XPhos Pd G2 catalyst (21 mg, 0.027 mmol) was added. The mixture was degassed, purged with N22and heated at 150°C for 45 minutes in a Biotage Initiator® microwave. The mixture was loaded onto a Biotage-ISOLUTE® HM-N cartridge and purified by chromatography on silica gel (eluting with 0-20% MeOH in DCM). The isolated yellow residue was treated with Et2O, filtered, washed with Et2Or, dried at 40°C in vacuo to give the title compound (I-42) (81mg, 65%) as a yellow solid. m/z=462 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1H), 11,68–11,62 (m, 1H), 8,72 (d, J=1,4 Hz, 1H), 8 ,07 (d, J=3,0 Hz, 1H), 7,50-7,46 (m, 2H), 7,42 (d, J=7,3 Hz, 1H), 7,29 (dd, J=9,0, 10,5 Hz, 1H), 7,11-7,01 (m, 2H), 6,60 (d, J= 7,3 Hz, 1H), 4,70 (d, J =4,3 Hz, 1H), 3,83 (s, 3H), 3,68–3,61 (m, 1H), 3,57–3,49 (m, 2H), 2,92–2, 83 (metro, 2H). ), 1,88–1,80 (m, 2H), 1,56–1,45 (m, 2H).

Example 2: Method B - Preparation of 2-(2,6-difluoro-3-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6- Naphthyridine-5(6H)-one (I-92)

Step 1: Ethyl-4-chloro-6-(2,6-difluoro-3-methoxyphenyl)-2-methylnicotinate (2.1)

To a solution of 2,6-difluoro-3-methoxybromobenzene (838 mg, 3.76 mmol) in dry THF (5 mL) under nitrogen at -78 °C was added dropwise a complex solution of sodium chloride lithium isopropylmagnesium chloride (1.3 M in THF, 4.2 mL, 5.40 mmol). After stirring at -78 °C for 40 min, zinc chloride (2M in THF, 5.6 mL, 11.28 mmol) was added dropwise. After warming to 0 °C for 20 min, the organozinc slurry was added to a degassed mixture and purged with ethyl 4,6-dichloro-2-methylnicotinate(1.0) nitrogen (550 mg, 2.35 mmol ) and Pd(PPh) given3)4(272 mg, 0.235 mmol) in THE (7 mL). After stirring at room temperature overnight, the reaction was washed with sat. N.H4Washed with Cl and extracted into EtOAc (2x). The combined organic phase was washed with brine, dried over Na2SO4, filtered, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (gradient elution with 0-60% EtOAc in cyclohexane) to give the title compound (2.1) (745 mg, 93%) as a colorless oil.1RMN-H (400 MHz, CDCl3): δ 7,39 (s, 1H), 7,02-6,90 (m, 2H), 4,49 (q, J=7,2 Hz, 2H), 3 ,90 (s, 3H), 2,63 (s, 3H). ), 1,44 (dd, J=7,2, 7,2 Hz, 3H).

Step 2: 4-chloro-2-(2,6-difluoro-3-methoxyphenyl)-1,6-naphthiridin-5(6H)-one (2,2)

The reaction was carried out according to the representative procedure described in Example 1, Method A, Step 1 to give the product (2.2).1RMN-H (400 MHz, DMSO): γ; 11,67–11,67 (m, 1H), 7,76 (s, 1H), 7,53 (d, J = 7,3 Hz, 1H), 7,41–7,33 (m, 1H) , 7,27–7,19 (m, 1H), 6,63 (d, J=7,3 Hz, 1H), 3,90 (s, 3H).

Schritt 3: 2-(2,6-difluoro-3-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthiridin-5(6H )-eins (I-92)

The reaction was carried out according to the representative procedure described in Example 1, Step 2 to give the product (I-92).1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1H), 11,64 (s, 1H), 8,35 (s, 1H), 7,97 (d, J = 2,8 Hz , 1H), 7,41–7,33 (m, 2H). ), 7,27-7,18 (m, 1H), 7,10 (dd, J=9,0, 9,0 Hz, 1H), 6,92 (d, J=9,1 Hz, 1H) , 6,47 (d, J=7,3 Hz, 1H), 4,60 (d, J=4,0 Hz, 1H), 3,82 (s, 3H), 3,60–3,50 ( m, 2H), 3,15–3,08 (m, 1H), 2,82–2,73 (m, 2H), 1,72 (d, J =9,6 Hz, 2H), 1,46 –1,34 (m, 2H).

Example 3: Method C - Preparation of 4-((1-ethyl-2-oxo-1,2-di-hydropyrimidin-4-yl)amino)-2-(2-fluoro-5-methoxy-4-(piperidine). ).-1 ).-Carbonyl)phenyl)-1,6-naphthyridin-5(6H)-one (1-228) .

Schritt 1. 4-Chloro-2-(2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl)-1,6-naphthyridine-5(6H)-on (3.1)

To a mixture of 2,4-dichloro-1,6-naphthyridin-5(6H)-one (1,1) (250 mg, 1.16 mmol), (5-fluoro-2-methoxy-4-(4). ,4 , 5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)(piperidin-1-yl)methanone (CB1) (422 mg, 1.16 mmol) and tetrakis(triphenylphosphine)palladium ( 134 mg, 10 mol %) under a nitrogen atmosphere in DME (5.0 mL) was added potassium carbonate solution (2 M in H).20, 1.3 mL, 2.56 mmol). The reaction mixture was purged with a stream of nitrogen for 5 minutes, then heated at 100°C for 1 hour in a Biotage Initiator microwave. The crude reaction mixture was cooled to RT and filtered through a pad of Celite® (2.5 g), rinsing with DCM. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (0-15% MeOH/DCM gradient elution) to give (3.1) (381 mg, 63% yield, 80% purity ), which was transferred to the next step without further cleanup. m/z=416.7 [M+H]+.

Schritt 2. 4-Chloro-2-(2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl)-6-((2-(trimethylsilyl)ethoxy)methyl)-1,6-naphthyridine- 5(6H)-eins (3.2)

To a mixture of 4-chloro-2-(2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl)-1,6-naphthyridin-5(6H)-one (3.1) (200 mg, 0.48 mmol) and 2-(trimethylsilyl)ethoxymethyl chloride (0.34 mL, 1.92 mmol) in THF (5.0 mL), degassed with a stream of nitrogen for 5 min, gave 1,8-diazabicycle [5.4.0]undec-7-ene (0.36 mL, 2.40 mmol). After stirring at room temperature for 16 h, the solvent was removed in vacuo, diluted with water (15 mL) and extracted with DCM (2 × 15 mL). The combined organic layers were washed with brine (15 mL), passed through a hydrophobic frit, and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel using 0-20% MeOH/DCM to give (3.2) (234 mg, 89% yield, 84% purity) as a yellow oil co-eluting with 16% ( 3.1) without reacting. The compound was taken to the next step without further purification. m/z=546.9 [M+H]+.

Step 3. 4-((1-ethyl-2-oxo-1,2-dihydropyrimidin-4-yl)amino)-2-(2-fluoro-5-methoxy-4-(piperidin-1-carbonyl)phenyl) -6-((2-(Trimethylsilyl)ethoxy)methyl)-1,6-naphthyridin-5(6H)-one (3.3)

To a mixture of 4-amino-1-ethylpyrimidin-2(1H)-one (42 mg, 0.30 mmol), Xantphos (23 mg, 0.04 mmol) and cesium carbonate (92 mg, 0.28 mmol ) in In 1,4-dioxane (2.5 mL) 4-chloro-2-(2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl)-6-((2-(trimethylsilyl) ethoxy)Methyl)-1 added ,6-naphthyridin-5(6H)-one(3.2) (110 mg, 0.20 mmol). The reaction mixture was stirred at room temperature with degassing under nitrogen flow for 10 min, treated with palladium(II) acetate (4.5 mg, 0.02 mmol) with degassing for a further 5 min, then warmed to 100 °C for 16 h. The crude material was filtered through a pad of Celite® (2.5 g frit), eluted with 5% MeOH/DCM, and then passed through a silica gel column with 0-20 MeOH. %/DCM to give the title compound (3.3) (88 mg, 67% yield, 62% purity) which was used without further purification. m/z=649.2 [M+H]+.

Schritt 4. 4-((1-Ethyl-2-oxo-1,2-di-hydropyrimidin-4-yl)amino)-2-(2-fluoro-5-methoxy-4-(piperidine-1-carbonyl) phenyl) -1,6-Naphthyridine-5(6H)-on (I-228)

A solution of crude 4-((1-ethyl-2-oxo-1,2-dihydropyrimidin-4-yl)amino)-2-(2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl) )-6-((2-(trimethylsilyl)ethoxy)methyl)-1,6-naphthyridin-5(6H)-one (3.3) (88 mg, 0.14 mmol) in DCM ( 1.0 ml) treated with trifluoroacetic acid (0.50 ml). After stirring at room temperature for 16h, the solvent was removed in vacuo. The crude material was neutralized with saturated aqueous sodium bicarbonate (5ml) and extracted into DCM (3 x 5ml). The combined organic layers were washed with brine (10 mL), passed through a hydrophobic frit, and concentrated under reduced pressure. The crude material was purified by preparative HPLC to give 4-((1-ethyl-2-oxo-1,2-dihydropyrimidin-4-yl)amino)-2-(2-fluoro-5-methoxy-4-(piperidine ) to give -1-carbonyl)phenyl)-1,6-naphthyridin-5(6H)-one (I-228) (23 mg, 33%) as a white solid. m/z=519.7 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,98 (s, 1H), 11,94 (s, 1H), 9,35 (s, 1H), 8,01 (d, J = 7,1 Hz , 1H), 7,58–7,49 (m, 2H). ), 7,27 (d, J=10,1 Hz, 1H), 6,73 (d, J=7,3 Hz, 1H), 6,13 (d, J=7,1 Hz, 1H), 3,86 (s, 3H), 3,81 (q, J =7,0 Hz, 2H), 3,66–3,54 (m, 2H), 3,19–3,13 (m, 2H) , 1,61–1,56 (m, 4H), 1,48–1,44 (m, 2H), 1,22 (t, J=7,1). hercios, 3H).

Example 4: Method D - Preparation of 2-(4-((dimethylamino)methyl)-2-fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl) Amino)-1,6-naphthyridine-5(6H)-one (I-215)

Step 1: 4-bromo-5-fluoro-2-methoxybenzaldeído (4.1)

To a solution of 2-bromo-1-fluoro-4-methoxybenzene (4.0) (2.65 g, 12.93 mmol) in DCM (45 mL) was added dropwise titanium (IV) at 0 °C in a nitrogen atmosphere. chloride (1 M) administered in DCM, 12.93 mL, 19.93 mmol). After 15 min, dichloromethyl methyl ether (1.4 mL, 15.51 mmol) and a further portion of titanium(IV) chloride (1M in DCM, 12.93 mL, 19.93 mmol) were added. After stirring at 0 °C for 3 h, the reaction was quenched by pouring into ice/water and extracted into DCM (3x). The combined extracts were washed with water and brine, dried over Na2SO4 and concentrated in vacuo to give the title compound (4.1) (3.69g) as a yellow/orange solid which was used as is in the next step. .

Step 2: 2-(4-Bromo-5-fluoro-2-methoxyphenyl)-1,3-dioxolano (4.2)

A mixture of 4-bromo-5-fluoro-2-methoxybenzaldehyde (4.1) (3.01 g, 12.93 mmol), ethylene glycol (1.4 mL, 25.86 mmol) and p-toluenesulfonic acid monohydrate (250). mg, first 0.29 mmol) in toluene (30 ml) was heated to reflux (Dean-Stark) for 18 h. The mixture was diluted with EtOAc, washed with sat solution. NaHCO3, water and brine, dried over Na2SO4and concentrated in emptiness. The residue was purified by chromatography on silica gel using 0-50% EtOAc in isohexane to give the title compound (4.2) (1.91 g, 53%) as a pale yellow oil.1RMN-H (400 MHz, CDCl3): δ 7,31 (d, J=8,6 Hz, 1H), 7,04 (d, J=5,1 Hz, 1H), 6,06 (s, 1H ), 4,14–3,99 (m, 4H), 3,85 (s, 3H).

Step 3: 2-(4-(1,3-dioxolan-2-yl)-2-fluoro-5-methoxyphenyl)-4,4,5,5-tetrametil-1,3,2-dioxaborolano (4.3)

The reaction was carried out according to the representative procedure described in Method CB3 using 2-(4-bromo-5-fluoro-2-methoxyphenyl)-1,3-dioxolane (4.2) to give the product (4.3).1RMN-H (400 MHz, CDCl3): δ 7,23–7,17 (m, 1H), 6,12 (s, 1H), 4,14–4,01 (m, 4H), 3,91– 3,86 (m, 2H), 1,38–1,34 (m, 9H), 1,29–1,25 (m, 6H).

Schritt 4: 2-(4-(1,3-dioxolan-2-yl)-2-fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl) amino)-1,6-naphthiridin-5(6H)-one (4.4)

The reaction was carried out according to the representative procedure described in Example 1, Method A, Step 3 using 2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino )-1 ,6-naphthyridin-5(6H)-one (1.2) and 2-(4-(1,3-dioxolan-2-yl)-2-fluoro-5-methoxyphenyl)-4,4,5, 5-tetramethyl-1,3,2-dioxaborolane (4.3) to give the product (4.4).1RMN H (400 MHz, DMSO): δ 12,47 (s, 1H), 11,73-11,68 (m, 1H), 8,77 (s, 1H), 8,11 (d, J=2 ,5 Hz, 1H), 7,60 (d, J =6,1 Hz, 1H), 7,52 (dd, J=2,9, 9,0 Hz, 1H), 7,47 (d, J =7,3 Hz, 1H), 7,38 (s, 1H), 7,07 (d, J=8,8 Hz, 1H). ), 6,66 (d, J=7,3 Hz, 1H), 6,08 (s, 1H), 4,74 (d, J=4,0 Hz, 1H), 4,14 (dd, J =6,8, 6,8 Hz, 2H), 4,03-3,91 (m, 4H), 3,72-3,66 (m, 1H), 3,59-3,52 (m, 2H ), 3,01-2,83 (m, 3H), 1,88 (d, J=9,9 Hz, 2H), 1,60-1,50 (m, 2H).

Schritt 5: 5-Fluoro-4-(4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridine- 2-yl)-2-methoxybenzaldehyde (4.5)

2-(4-(1,3-dioxolan-2-yl)-2-fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino mixture ) -1,6-naphthyridin-5(6H)-one (4,4) (240 mg, 0.450 mmol) and HCl (4N in 1,4-dioxane, 4 mL, 16 mmol) in THF (10 mL) were heated at 50°C for 2 h. The cooled mixture was washed with sat. NaHCO3and extracted into EtOAc (x2). The combined extracts were washed with brine, dried over Na2SO4filtered and concentrated in vacuo to give the title compound (4.5) (194 mg, 88%) as an orange solid which was used in the next step without purification.

Paso 6: 2-(4-((Dimetilamino)metil)-2-fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1, 6-naphthyridin-5(6H)-uno (I-215)

To a mixture of 5-fluoro-4-(4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6 at room temperature -Naphthyridin-2-yl)-2-methoxybenzaldehyde (4.5) (90 mg, 0.184 mmol), dimethylamine (370 µL, 0.735 mmol) in DCM (5 mL) and MeOH (1 mL) was treated with diacetoxyborohydride. sodium (58mg) . , 0.276 mmol) After stirring for 18 h, the mixture was filtered through a pad of Celite® and the filtrate was passed through an SCX cartridge (eluted with MeOH followed by 7N methanolic ammonia). The crude material was purified by preparative HPLC to give 2-(4-((dimethylamino)methyl)-2-fluoro-5-methoxyphenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2) give -yl)amino)-1,6-naphthyridin-5(6H)-one (I-215) (23 mg, 24%) as a yellow solid. m/z=519.2 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,41 (s, 1H), 11,66 (d, J=5,5 Hz, 1H), 8,73 (d, J=1,3 Hz, 1H ), 8,23 (s, 1H), 8,08 (d, J=3,0 Hz, 1H), 7,53 (d, J=6,4 Hz, 1H), 7,48 (dd, J =3,1, 9,0 Hz, 1H), 7,44–7,40 (m, 1H), 7,03 (d, J =8,8 Hz, 1H), 6,61 (d, J= 7,3 Hz, 1H), 3,86 (s, 4H), 3,68–3,60 (m, 2H), 3,57–3,49 (m, 3H), 2,92–2,83 (m, 2H), 2,23 (s, 6H), 1,88–1,80 (m, 2H), 1,56–1,45 (m, 2H).

Example 5: Method E - Preparation of 2-(3-Hydroxy-1-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine -5-one (I-50)

Schritt 1. 2-chloro-4-((5-(4-hidroxipiperidin-1-yl)pyridin-2-yl)amino)-1,6-naftiridin-5(6H)-ona (1,2)

The reaction was carried out according to the procedure described in Example 1, Method A, Step 2.

Schritt 2. 2-(3-Hydroxy-1-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one ( I-50)

For a reaction tube treated with 2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H)-one (1.2) ( 200 mg, 0.58 mmol), 3-hydroxypiperidine (65 mg, 0.64 mmol), potassium carbonate (149 mg, 1.0 mmol) and xanthophos (62 mg, 0.10 mmol) in dioxane (8 mL), rinsed with N2Bis(dibenzylideneacetone)palladium(0) (31 mg, 0.05 mmol) was added. After stirring at 125 °C for 8 h, the reaction mixture was allowed to cool to RT and filtered through Celite. The solvent was removed in vacuo to leave a crude residue which was purified by preparative HPLC to give 2-(3-hydroxy-1-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl to give [amino]-6H-1,6-naphthyridin-5-one (I-50) (16 mg, 7%) as a yellow solid. m/z=437 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,25 (s, 1H), 11,15 (s, 1H), 8,04 (s, 1H), 7,92 (s, 1H), 7,47– 7,41 (m, 1H), 7,21 (s, 1H), 6,92-6,87 (m, 1H), 6,22 (d, J=6,9 Hz, 1H), 4,30 (d, J=11,8 Hz, 1H), 4,14-4,06 (m, 1H), 3,62-3,50 (m , 4H), 3,04–2,98 (m, 1H ), 2,89–2,79 (m, 3H), 1,94–1,75 (m, 4H), 1,55–1,41 (m, 4H).

Example 6: Method F - Preparation of 2-[2-Fluoro-4-(pyrrolidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]- 6H-1,6-naphthyridin-5-one (I-115)

Step 1. 3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl]benzoic acid acid acid (I-134)

To a mixture of methyl 3-fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl ]benzoate (I-125) (1.43 g, 2.92 mmol) dissolved in THF (20 mL) and water (5 mL) was added lithium hydroxide monohydrate (200 mg, 4.67 mmol). After stirring at room temperature overnight. 4M HCl in dioxane (10 mL) was added and the mixture was evaporated to dryness. The residue was dried by azeotroping with dioxane (2 x 100 mL) to give crude product (I-134) (2.78 g) as a colorless gummy solid. The crude residue was used in the next step without further purification (HCl salt plus residual LiCl was assumed). 45 mg of the crude product was further purified by preparative HPLC to give pure 3-fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H- 1,6-naphthyridin-2-yl]benzoic acid (I-134) as a white solid. m/z=476 [M+H]+,1RMN H (400 MHz, DMSO): δ 13,47–13,46 (m, 1H), 12,50 (s, 1H), 11,72 (d, J = 5,3 Hz, 1H), 8, 80 (s, 1H), 8,17–8,09 (m , 2H), 7,95 (dd, J=1,3, 8,1 Hz, 1H), 7,86 (s, 1H), 7 ,55-7,44 (m, 2H), 7,08 (d, J=9,1 Hz, 1H), 6,65 (d, J=6,6 Hz, 1H), 4,73–4, 67 (m, 1H), 3,73–3,66 (m, 1H), 3,61–3,53 (m, 2H), 2,95–2,87 (m, 2H), 1,89 ( dd, J= 3,5, 8,8 Hz, 2H), 1,60–1,49 (m, 2H).

Step 2. Preparation of 2-[2-Fluoro-4-(pyrrolidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1 ,6-naphthyridine-5-one (I-115)

Crude 3-fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl]benzoic acid ( I-134) (94 mg, assuming 0.19 mmol) dissolved in DMF (900 µl), DIPEA (138 µl, 0.79 mmol), pyrrolidine (50 µl, 0.59 mmol) and HOPO solution (33 mg, 0.29 mmol) and EDCl.HCl (51 mg, 0.26 mmol) in DMF (300 µL). After stirring at room temperature overnight, the reaction mixture was diluted with DCM (6ml) and washed with water (3ml). The organic phase was collected and the aqueous phase was washed with more DCM. The combined organic phases were passed through a hydrophobic separator and the solvent was removed in vacuo to leave a crude residue which was purified by preparative HPLC to give 2-[2-fluoro-4-(pyrrolidin-1-carbonyl)phenyl] -4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one (I-115) (12 mg, 12%) as a solid yellow. m/z=529 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1H), 11,73 (s, 1H), 8,79 (s, 1H), 8,13–8,04 (m, 2H) , 7,57–7,50 (m, 3H), 7,47 (d, J=7,1 Hz, 1H), 7,08 (d, J=8,8 Hz, 1H), 6,64 ( d, J=7,3 Hz, 1H), 4,74 (d, J=1,3 Hz, 1H), 3,68 (d, J = 1,3 Hz, 1H), 3,61–3, 46 (m, 6H), 2,95–2,87 (m, 2H), 1,98–1,86 (m, 6H), 1,60–1,49 (m, 2H).

Example 7: Method G - Preparation of 2-(6-Fluoro-1,2,3,4-tetrahydroisoquinolin-5-yl)-4-[(5-tetrahydropyran-4-yl-1H-pyrazol-3-yl) Amino]-6H-1,6-naphthyridin-5-one (I-285)

For a solution of tert-butyl-6-fluoro-5-(5-oxo-4-((5-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-3-yl)amino)-5, 6-Dihydro-1,6-naphthyridin-2-yl)-3,4-dihydroisoquinolin-2(1H)-carboxylate (7,0) (77 mg, 0.13 mmol) in (2 mL) trifluoroacetic acid (0 .5 ml) was added. After stirring at room temperature for 3 h, the mixture was concentrated in vacuo and the resulting residue was purified by preparative HPLC to give 2-(6-Fluoro-1,2,3,4-tetrahydroisoquinolin-5-yl)-4 - [(5-Tetrahydropyran-4-yl-1H-pyrazol-3-yl)amino]-6H-1,6-naphthyridin-5-one (I-285) (47 mg, 74 %) as a yellow solid. m/z=461 [M+H]+,1RMN-H (400 MHz, DMSO): δ 12,02 (s, 1H), 8,29 (s, 1H), 7,82 (s, 1H), 7,45 (d, J=7,3 Hz , 1H), 7,39 (dd, J=5,7, 8,7 Hz, 1H), 7,29 (dd, J=8,8, 8,8 Hz, 1H), 6,59 (d, J=7,3 Hz, 1H), 5,99 (s, 1H), 4,32 (s, 2H), 3,45 (dd , J=9,9, 11,6 Hz, 2H), 2, 95-2,88 (m, 1H), 2,81 (dd, J=5,7, 5,7 Hz, 4H), 1,83 (dd, J=1,8, 12,9 Hz, 4H) , 1,70-1,58 (m, 4H).

Example 8: Method H - Preparation of 2-(2,6-difluorophenyl)-8-(1-hydroxyethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H -1,6-naphthyridin-5-one (I-15) and 2-(2,6-difluorophenyl)-8-(1-hydroxy-1-methyl-ethyl)-4-[[5-(4-hydroxy -1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one (I-16)

Stage 1. 8-Bromo-4-chloro-2-(2,6-difluorophenyl)-1,6-naphthyridin-5(6H)-one (8.1) .

For a solution of 4-chloro-2-(2,6-difluorophenyl)-1,6-naphthyridin-5(6H)-one (8.0, prepared according to Method B, steps 1 and 2, of 2 A difluorobromobenzene and ethyl (4,6-dichloro-2-methylnicotinate) (750 mg, 2.5 mmol) dissolved in DMF (10 mL) was added N-bromosuccinimide (460 mg, 2.5 mmol) with stirring at room temperature for 1 h, The solvent was removed in vacuo and DCM was added to the residue to promote precipitation of the product.The solid was collected by filtration and purified by trituration with a minimum of methanol to give the pure product (8.1) (450 mg, 47%) as a separate product -White solid.1RMN-H (400 MHz, DMSO) 12,02–12,02 (1H, m), 8,01–7,94 (2H, m), 7,75–7,66 (1H, m), 7, 36 (2H, t, J = 8,1 Hz).

Step 2. 8-Bromo-2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine-5-2. un (I-5)

The reaction was carried out according to the procedure described in Example 1, Method A, Step 2 using 8-bromo-4-chloro-2-(2,6-difluorophenyl)-1,6-naphthyridine-5(6H)- one (8.1) done. and 1-(6-aminopyridin-3-yl)piperidin-4-ol to give the desired product (I-5) as a yellow solid. m/z=528 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,65 (s, 1H), 12,13-12,09 (m, 1H), 8,60 (s, 1H), 8,11 (d, J=2 ,5 Hz, 1H), 7,91 (s, 1H), 7,69-7,60 (m, 1H), 7,52 (dd, J=2,9, 9,0 Hz, 1H), 7 ,32 (dd, J=8,0, 8,0 Hz, 2H), 7,07 (d, J=8,8 Hz, 1H), 4,73 (d, J=4,3 Hz, 1H) , 3,70–3,65 (m, 1H), 3,56 (d, J=12,6 Hz, 2H), 2,95–2,86 (m, 2H), 1,90–1,84 (m, 2H), 1,58-1,46 (m, 2H).

Etapa 3. 8-Acetyl-2-(2,6-difluorophenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridina-5(6H ). )-um (8.2)

For a solution of 8-bromo-2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine-5 -one (I-5) (75 mg, 0.14 mmol) dissolved in DMF (1 mL) and purged with nitrogen for 20 min, tributyl(1-ethoxyvinyl)tin (67 mg, 0.18 mmol) and tetrakis added (triphenylphosphine)palladium(0) (16 mg, 0.01 mmol). After stirring at 80 °C for 48 h, the mixture was diluted with water (6 mL) and extracted with DCM (3 x 3 mL). The combined organic phases were washed with water (4 ml) and then passed through a hydrophobic separator. The solvent was removed in vacuo and the crude residue was purified by column chromatography eluting with 0-20% methanol in DCM to give pure 8.2 (40 mg, 57%) as an off-white solid.1RMN-H (400 MHz, DMSO) 12,48 (1H, s), 12,26 (1H, s), 8,60 (1H, s), 8,13–8,08 (1H, m), 7 ,91 (1H, s), 7,68–7,59 (1H, m), 7,55–7,49 (1H, m), 7,36–7,29 (2H, m), 7,11 –7,03 (1H, m), 4,73 (1H, d, J=4,0 Hz), 3,71–3,65 (1H, m ), 3,63-3,53 (2H, m ), 2,95-2,88 (2H, m), 2,74-2,72 (3H, m), 1,90-1,80 (2H, m), 1,57-1,47 (2H , m).

Schritt 4. 2-(2,6-difluorophenyl)-8-(1-hydroxyethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6- Naphthyridin-5-one (I-15)

For a solution of 8-acetyl-2-(2,6-difluorophenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5( 6H)-one (8.2) (40 mg, 0.08 mmol) in ethanol (0.6 mL) was treated with sodium borohydride (31 mg, 0.81 mmol). After stirring at room temperature for 1h, the reaction was quenched with saturated aqueous ammonium chloride solution (1ml), stirred for 10min and extracted with ethyl acetate (3 x 3ml). The combined organic phases were dried (MgSO4), filtered and evaporated in vacuo to give a crude product which was purified by preparative HPLC to give (I-15) (16 mg, 42%) as a yellow solid. m/z=494 [M+H]+,1RMN H (400 MHz, MeOD) δ 8,44 (s, 1H), 8,09 (d, J=2,9 Hz, 1H), 7,56–7,48 (m, 2H), 7,38 (s, 1H), 7,14 (dd, J= 8,2, 8,2 Hz, 2H), 7,07 (d, J=8,8 Hz, 1H), 5,23 (q, J=6, 3 Hz, 1H), 3,82-3,74 (m, 1H), 3,62-3,54 (m, 2H ), 2,98-2,90 (m, 2H), 2,03-1,97 (m, 2H), 1,73-1,63 (m, 2H), 1,56 (d, J=6,4 Hz, 3H). 3 No se observan protones intercambiables.

Schritt 5. 2-(2,6-difluorophenyl)-8-(1-hydroxy-1-methylethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]- 6H- 1,6-naphthyridin-5-one (I-16)

For a solution of 8-acetyl-2-(2,6-difluorophenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5( 6H)-one (8.2) (73 mg, 0.14 mmol) dissolved in THF (3 mL) was added dropwise at 0 °C under nitrogen with methylmagnesium bromide (297 µL, 3 M solution in diethyl ether, 0.89 mmol) displaced. After stirring for 25 minutes, the reaction was quenched with saturated aqueous ammonium chloride solution (2 mL) and diluted with water (5 mL). The mixture was extracted with DCM (3 x 5 mL) and the combined organic extracts were passed through a phase separator and concentrated in vacuo to give a crude product which was purified by preparative HPLC to give pure (I-16). (17 mg) to give 23%) as a yellow solid. m/z=508 [M+H]+,1RMN-H (400MHz, MeOD) δ 12.51 (s, 1H), 8.48 (td, J=1.3, 11.9 Hz, 2H), 8.11 (d, J=2.9 Hz, 1H), 7.57-7.49 (m, 2H), 7.36 (s, 1H), 7.16 (dd, J=8.3, 8.3Hz, 2H), 7.08 (dd, J=1.6, 8.8Hz, 1H), 3.83-3.75 (m, 1H), 3.63-3.56 (m, 2H), 2.99-2.91 (m, 2H), 2.03-1.98 (m, 2H), 1.74-1.67 (m, 2H), 1.65 (s, 6H). 2 Exchangeable protons not observed.

Example 9: Method I - Preparation of 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 -naphthyridine-8-carbonitrile (I-33) and 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H -1,6-naphthyridine-8-carboxylic acid (I-39)

Schritt 1. 2-(2,6-Difluorfenil)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-8- Carbonitrile (I-33)

A microwave tube was charged with 8-bromo-2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6- naphthyridine -5-one (I-5) (200 mg, 0.37 mmol), cuprous cyanide (68 mg, 0.75 mmol) and DMF (2 mL). After stirring at 175 °C for 3 h in a microwave, the reaction mixture was cooled to room temperature and evaporated in vacuo to give a crude product which was purified by preparative HPLC to give pure I-33 (9 mg, 5 %) giving a yellow solid. m/z=475 [M+H]+,1RMN H (400 MHz, DMSO) δ 12,62-12,57 (m, 1H), 8,54 (s, 1H), 8,40 (s, 1H), 8,10 (d, J=2, 5 Hz, 1H), 7,69-7,60 (m , 1H), 7,51 (dd, J=2,7, 9,0 Hz, 1H), 7,32 (dd, J=8, 0 , 8,0 Hz, 2H), 7,07 (d, J=9,1 Hz, 1H), 4,73-4,72 ( m, 1H), 3,70-3,64 (m, 1H) , 3,56 (d, J=12,1 Hz, 2H), 2,90 (dd, J=10,0, 10,0 Hz, 2H), 1,84 (d, J=9,6 Hz, 2H ), 1,58-1,48 (m, 2H). Um proton trocável não foi observado

Step 2. 2-(2,6-Difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridine-8- carboxylic acid (I-39)

For a reaction tube filled with 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6- Naphthyridine - Concentrated sulfuric acid (168 µL, 3.09 mmol) was added to 8-carbonitrile (I-33) (70 mg, 0.14 mmol) in water (1.5 mL) and dioxane (2 mL). After stirring at 115 °C for 24 h, the pH of the reaction mixture was adjusted to pH 8 with NaHCO.3(formation of insolubles). The mixture was diluted with ethyl acetate and filtered through a pad of celite, washing with methanol and DMF. The filtrate was evaporated in vacuo to give a crude product which was purified by preparative HPLC to give I-39 (10 mg, 14%) as a yellow solid. m/z=494 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,83-12,77 (m, 1H), 8,65 (s, 1H), 8,38 (s, 1H), 8,15 (d, J=2 ,8 Hz, 1H), 7,75-7,67 (m, 1H), 7,54 (dd, J=2,8, 8,8 Hz, 1H), 7,40 (dd, J=8, 3, 8,3 Hz, 2H), 7,15 (d, J=8,8 Hz, 1H), 4,74 (s, 1H), 4,08 (s, 1H), 3,72-3, 67 (m, 1H), 3,60 (dd, J=4,4, 8,2 Hz, 2H), 2,98-2,91 (m, 2H), 1,87 (dd, J=3, 2, 12,8 Hz, 2H), 1,58–1,47 (m, 2H). 1 Austauschbares Proton nicht beobachtet

Example 10. Method J - Preparation of 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1,6 -Nafthyridine-5-one (I-25)

Etapa 1. 8-Bromo-4-chloro-2-(2,6-difluorofenil)-6-(4-methoxybenzyl)-1,6-naphthiridin-5(6H)-one (10.0)

To a solution of 8-bromo-4-chloro-2-(2,6-difluorophenyl)-1,6-naphthyridin-5(6H)-one (8.1) (460 mg, 1.24 mmol) dissolved in DMF (10ml). ) were added potassium iodide (51 mg, 0.31 mmol), potassium carbonate (210 mg, 1.55 mmol) and 4-methoxybenzyl chloride (200 µl, 1.49 mmol). After stirring at room temperature for 2h, the reaction mixture was diluted with water (20ml) and a precipitate formed. The solid was collected by filtration and dried to give the desired product (10.0) (550 mg, 90%) as an off-white solid.1RMN H (400 MHz, DMSO) 8,56 (1H, s), 7,99 (1H, s), 7,75–7,66 (1H, m), 7,45–7,33 (4H, m), 7,00–6,95 (2H, m), 5,16 (2H, s), 3,80–3,78 (3H, m).

Step 2. 4-Chloro-2-(2,6-difluorofenil)-6-(4-methoxybenzil)-8-metil-1,6-naphthiridin-5(6H)-one (10.1)

In a carousel tube with 8-bromo-4-chloro-2-(2,6-difluorophenyl)-6-(4-methoxybenzyl)-1,6-naphthyridin-5(6H)-one (10.0) ( 150 mg, 0.30 mmol), methylboronic acid (21 mg, 0.35 mmol), and cesium carbonate (298 mg, 0.91 mmol) in dioxane (1.5 mL) and water (0.15 mL) purged with nitrogen added [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) complex with DCM (30 mg, 0.03 mmol). After stirring at 100°C overnight, the reaction mixture was diluted with water (10 mL) and brine (2 mL) and washed with ethyl acetate (2x20 mL). The combined organic phases were dried (MgSO4), filtered and the solvent removed in vacuo to give a crude product which was purified by column chromatography eluting with 0-20% ethyl acetate in DCM to give pure (10.1) (44 mg, 34%) as a yellow solid.1RMN-H (400 MHz, CDCl3) 7,59 (1H, s), 7,44–7,35 (1H, m), 7,35–7,22 (3H, m), 7,06–6, 99 (2H, m), 6,91–6,85 (2H, m), 5,14–5,10 (2H, M), 3,81–3,79 (3H, M), 2,30– 2,28 (3H,M).

Paso 3. 2-(2,6-Difluorofenil)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-6-(4-methoxybenzyl)-8-metil-1 ,6-naphthyridin-5(6H)-one (10,2)

The reaction was carried out according to the procedure described in Example 1, Method A, Step 2 using 4-chloro-2-(2,6-difluorophenyl)-6-(4-methoxybenzyl)-8-methyl-1 ,6-naphthyridin-5(6H)-one (10.1) and 1-(6-aminopyridin-3-yl)piperidin-4-ol to give the desired crude product (10.2) as a yellow solid which is used directly in the next step. Yeah .

Etapa 4. 2-(2,6-Difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1,6-naphthyridina-5- un (I-25)

A microwave tube was charged with 2-(2,6-difluorophenyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-6-(4-methoxybenzyl)-8 - . Methyl-1,6-naphthyridin-5(6H)-one (10.2) (57 mg, 0.09 mmol) and TFA (1 mL). The tube was sealed and the reaction was heated at 165 °C for 4 h. The reaction solvent was removed in vacuo to give a crude product which was diluted in DCM (1 mL), methanol (2 mL) and 1 M methanolic ammonia (4 mL) [caution!] and stirred at room temperature for 1 h before the reevaporation dried. The residue was purified by preparative HPLC to give pure (I-25) (10 mg, 22%) as a yellow solid. m/z=464 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,78 (s, 1H), 11,63 (s, 1H), 8,55 (s, 1H), 8,09 (d, J=2,8 Hz, 1H), 7,67–7,58 (m, 1H), 7,51 (dd, J=2,9, 9,0 Hz, 1H), 7,37 (s, 1H), 7,31 (dd , J=8,0, 8,0 Hz, 2H), 7,04 (d, J=8,8 Hz, 1H), 4,73 (s, 1H), 3,70-3,65 (m, 1H), 3,54 (d, J=12,9 Hz, 2H), 2,94-2,85 (m, 2H), 2,21 (s, 3H), 1,90-1,84 (m , 2H), 1,58-1,47 (m, 2H).

Example 11. Method K - Preparation of 2-(2,6-difluorophenyl)-8-ethyl-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6- Naphthyridine-5(6H)-one (I-49)

Step 1. 8-Bromo-4-((5-(4-((tert-butildimethylsilyl)oxi)piperidin-1-yl)pyridin-2-yl)amino)-2-(2,6-difluorofenil)-6 -(4-methoxybenzyl)-1,6-naphthiridin-5(6H)-one (11.0)

The reaction was carried out according to the procedure described in Example 1, Method A, Step 2 using 8-bromo-4-chloro-2-(2,6-difluorophenyl)-6-(4-methoxybenzyl)-1 ,6-naphthyridin-5 carried out (6H)-one (10.0) and 5-(4-((tert-butyldimethylsilyl)oxy)piperidin-1-yl)pyridin-2-amine to give the desired crude product (11.0) as a yellow solid resulted, which was used in the next step without further purification.

Step 2. 4-((5-(4-((tert-butyldimethylsilyl)oxy)piperidin-1-yl)pyridin-2-yl)amino)-2-(2,6-difluorophenyl)-6-(4- Methoxybenzyl)-8-vinyl-1,6-naphthiridin-5(6H)-one (11.1)

The reaction was carried out according to the procedure described in Example 10. Method J, step 2 using 8-bromo-4-((5-(4-((tert-butyldimethylsilyl)oxy)piperidin-1-yl) pyridin-2-yl)amino)-2-(2,6-difluorophenyl)-6-(4-methoxybenzyl)-1,6-naphthyridin-5(6H)-one (11.0) and pinacol ester of acid vinylboronic acid to give the desired product (11.1) as a yellow solid of 75% purity. The product was used in the next step without further purification.

Step 3. 4-((5-(4-((tert-butyldimethylsilyl)oxy)piperidin-1-yl)pyridin-2-yl)amino)-2-(2,6-difluorophenyl)-8-ethyl-6 -(4-methoxybenzyl)-1,6-naphthiridin-5(6H)-one (11.2)

A solution of 4-((5-(4-((tert-butyldimethylsilyl)oxy)piperidin-1-yl)pyridin-2-yl)amino)-2-(2,6-difluorophenyl)-6-(4- Methoxybenzyl)-8-vinyl-1,6-naphthyridin-5(6H)-one (11.1) (117 mg, 0.16 mmol) in methanol (20 mL) was purified using 10% Pd/C by a H-Cube® with cartridge head (RT, atmospheric pressure). The solvent was removed in vacuo to give a crude product which was purified by column chromatography eluting with 0-100% diethyl ether in DCM to give the desired product (11.2) (53 mg, 45%) as a yellow solid.1RMN-H (400 MHz, CDCl3) 12,51–12,47 (1H, m), 8,49 (1H, s), 7,98–7,95 (1H, m), 7,30–7, 23 (1H, m), 7,22–7,18 (2H, m), 7,01-6,98 (1H, m), 6,91 (4H, q, J=7,7 Hz), 6 ,82 (2H, d, J=8,6 Hz), 5,06 (2H, s), 3,84-3,78 (1H, m), 3,75-3,71 (3H, m), 3,35-3,28 (2H, m), 2,96-2,88 (2H, m), 2,75-2,64 (2H, m), 1,84-1,76 (2H, m ), 1,65-1,59 (2H, m), 0,84-0,81 (12H, m), 0,00 (6H, s).

Etapa 4. 2-(2,6-Difluorophenyl)-8-ethyl-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridina-5(6H )-um (I-49)

The reaction was carried out according to the procedure described in Example 10, Method J, Step 4 using 4-((5-(4-((tert-butyldimethylsilyl)oxy)piperidin-1-yl)pyridin-2- yl)amino )-2-(2,6-difluorophenyl)-8-ethyl-6-(4-methoxybenzyl)-1,6-naphthyridin-5(6H)-one (11.2). The crude residue was purified by preparative HPLC to provide pure (I-49) (8 mg, 24%) as a yellow solid. m/z=478 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,78 (s, 1H), 11,71–11,65 (m, 1H), 8,60–8,53 (m, 1H), 8,09 (d , J=2,8 Hz, 1H), 7,67–7,57 (m, 1H), 7,51 (dd, J=2,8, 9,1 Hz, 1H), 7,35–7, 27 (m, 3H), 7,06–7,02 (m, 1H), 4,72 (s, 1H), 3,72–3,63 (m, 1H), 3,59-3,51 ( m, 2H), 2,93-2,87 (m, 2H), 2,71 (q, J=7,2 Hz, 2H), 1,86 (d, J=9,6 Hz, 2H), 1,58-1,47 (m, 2H), 1,22 (t, J=7,5 Hz, 3H).

Example 12. Method L - Preparation of 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-isopropyl-8-methyl-6H-1,6-naphthyridin-5-one (I-89) and 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1,6-naphthyridin-5-one (I-90)

Step 1. 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-isopropyl-8-methyl-6H-1,6-naphthiridin-5-one (I-89) e 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1,6-naphthiridin-5-one (I-90)

2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-8-methyl-1,6-naphthyridin-5(6H)-one (12.0) ( 100 mg, 0.25 mmol, prepared according to the procedure described in Example 1, Method A using ethyl 4,6-dichloro-2-ethylnicotinate) and ferric acetylacetonate (18 mg, 0.05 mmol), dissolved in THE ( 3 ml) and NMP (1 ml). The mixture was cooled to -45 °C and isopropylmagnesium chloride (1.9 mL, 3.89 mmol, 2M in THF) was added dropwise over 15 min. The reaction was stirred at -10 °C for 1 h, then at room temperature for 1.5 h. The reaction was cooled to -15 °C and then quenched using saturated aqueous ammonium chloride (5 mL). The mixture was allowed to warm to room temperature, diluted with water (5ml) and extracted with ethyl acetate (2 x 25ml). The combined organic extracts were passed through an Isolute phase separator and the solvent was removed in vacuo to give a crude product which was purified by preparative HPLC to give 4-[[5-(4-hydroxy-1-piperidyl)- 2- cigar to lend. Pyridyl]amino]-2-isopropyl-8-methyl-6H-1,6-naphthyridin-5-one (I-89) (10 mg, 10%) as a yellow solid. m/z=394 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,54 (s, 1H), 11,39 (s, 1H), 8,28 (s, 1H), 8,13 (d, J=3,0 Hz, 1H), 7,51 (dd, J=2,8, 8,8 Hz, 1H), 7,27 (s, 1H), 7,01 (d, J=8,8 Hz, 1H), 4, 74 (d, J=4,0 Hz, 1H), 3,72-3,66 (m, 1H), 3,58-3,52 (m, 2H), 3,05-2,97 (m, 1H), 2,96-2,88 (m, 2H), 2,25 (s, 3H), 1,93-1,87 (m, 2H), 1,61-1,50 (m, 2H) , 1,32 (d, J=7,1 Hz, 6H); e 4-[[5-(4-hidroxi-1-piperidil)-2-piridil]amino]-8-metil-6H-1,6-naftiridin-5-on (I-90) (17 mg, 19% ) como gelber Feststoff. m/z=352 [M+H]+,1H-RMN (400 MHz, DMSO) δ 12,62 (s, 1H), 11,51 (d, J=4,7 Hz, 1H), 8,55 (d, J=5,8 Hz, 1H) , 8,32 (d, J=6,0 Hz, 1H). ), 8,12 (d, J=2,8 Hz, 1H), 7,51 (dd, J=2,8, 8,8 Hz, 1H), 7,30 (d, J=4,8 Hz , 1H), 7,03 (d, J=8,8 Hz, 1H). ), 4,77 (s, 1H), 3,72–3,65 (m, 1H), 3,59–3,51 (m, 2H), 2,94–2,87 (m, 2H), 2,24 (s, 3H), 1,87 (dd, J=4,5). , 7,9 Hz, 2H), 1,61–1,50 (m, 2H).

Beispiel 13: Verfahren M – Herstellung von 2-Acetyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on (I-47 ), 2-(1-Hydroxyethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on (I-37), 2-(1-Hydroxy-1-methyl-ethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on (I -38) e 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(2,2,2-trifluoro-1-hydroxy-1-methyl-ethyl)-6H -1,6-Nafthyridine-5-on (I-77)

Step 1. 2-(1-Ethoxivinyl)-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthiridin-5(6H)-one (13, 0)

The reaction was carried out according to the procedure described in Example 8, Method H, Step 3 using 2-chloro-4-((5-(4-hydroxypiperidin-11-yl)pyridin-2-yl)amino) -1,6-naphthyridin-5(6H)-one (1.2) and tributyl(1-ethoxyvinyl)tin to give the desired product (13.0) as a yellow solid.1RMN-H (400 MHz, DMSO) 12,33 (1H, s), 11,56–11,52 (1H, m), 8,58 (1H, s), 8,08 (1H, d, J = 3,0 Hz), 7,50–7,46 (1H, m), 7,36 (1H, dd, J=5,8, 7,2 Hz), 7,02–6,99 (1H, m ), 6,52–6,49 (1H, m), 5,47 (1H, d, J=1,1 Hz), 4,72–4,70 (1H, m), 4,49 (1H, s), 3,95 (2H, q, J=7,0 Hz), 3,68–3,62 (1H, m), 3,57–3,49 (2H, m), 2,92–2 ,84 (2H, m) , 1,89–1,81 (2H, m), 1,57–1,46 (2H, m), 1,42–1,37 (3H, m).

Schritt 2. 2-Acetyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on (I-47)

For a solution of 2-(1-ethoxyvinyl)-4-((5-(4-hydroxypiperidin-11-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H)-one (13 .0) (191 mg, 0.46 mmol) dissolved in THF (5 mL) was treated with 4M HCl solution (5 mL). After stirring at room temperature overnight, the mixture was concentrated in vacuo and the residue was partitioned between aqueous NaHCO33and ethyl acetate. The organic phase was collected and washed with brine, dried (Na2SO4) and the solvent removed in vacuo to give (I-47) (158 mg, 88%) as an orange solid. m/z=380 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,46 (s, 1H), 11,78 (s, 1H), 8,71 (s, 1H), 8,15–8,10 (m, 1H), 7,53–7,42 (m, 2H), 7,02 (d, J=8,8 Hz, 1H), 6,63 (d, J=7,3 Hz, 1H), 4,71 (s , 1H), 3,71–3,49 (m, 3H), 2,96–2,84 (m, 2H), 2,65 (s , 3H), 1,85 (s, 2H), 1, 53 (t, J=9,3 Hz, 2H).

Schritt 3. 2-(1-Hydroxyethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on (I-37)

The reaction was carried out according to the procedure described in Example 8: Method H, Step 4 using 2-Acetyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H- 1,6-naphthyridin-5-one (I-47) to give (I-37) as a yellow solid. m/z=382 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,53 (d, J=5,0 Hz, 1H), 8,29 (s, 1H), 8,22 (s, 1H), 8,09 (d, J=3,0 Hz). , 1H), 7,48 (dd, J=3,1, 9,0 Hz, 1H), 7,35 (dd, J=5,5, 7,0 Hz, 1H), 7,02 (d, J=8,9 Hz, 1H), 6,48 (d, J= 7,3 Hz, 1H), 5,36 (s, 1H), 4,65 (q, J=6,5 Hz, 1H) , 3,69–3,51 (m, 3H), 2,93–2,84 (m, 2H), 1,89–1,81 (m, 2H). ), 1,57-1,46 (m, 2H), 1,37 (d, J=6,7 Hz, 3H).

Step 4. 2-(1-Hydroxy-1-methyl-ethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine-5- a (I-38)

The reaction was carried out according to the procedure described in Example 8: Method H, Step 5 using 2-Acetyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]- 6H-1,6-naphthyridin-5-one (I-47) to give (I-38) as a yellow solid. m/z=396 [M+H]+,1RMN H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,59–11,54 (m, 1H), 8,44 (s, 1H), 8,13 (d, J=2, 5 Hz, 1H), 7,51 (dd, J= 2,9, 9,0 Hz, 1H), 7,38 (d, J=7,3 Hz, 1H), 7,10-7,05 ( m, 1H), 6,53 (d, J=7,3 Hz, 1H), 5,26 (s, 1H), 4,75 (d, J=3,8 Hz, 1H), 3,74– 3,67 (m, 1H), 3,61–3,53 (m, 2H), 2,97–2,88 (m, 2H), 1,93–1,87 (m, 2H), 1, 62–1,52 (m, 2H), 1,50 (s, 6H).

Schritt 5. 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(2,2,2-trifluoro-1-hydroxy-1-methylethyl)-6H-1, 6-naphthyridin-5-one (I-77)

The reaction was carried out according to the procedure described in Method CB8, Step 1 using 2-Acetyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1 ,6-naphthyridin-5-one (I-47) to give (I-77) as a yellow solid. m/z=450 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,43 (s, 1H), 11,71 (s, 1H), 8,64 (s, 1H), 8,14 (d, J=2,8 Hz, 1H), 7,53 (dd, J=2,9, 9,0 Hz, 1H), 7,45 (d, J=7,3 Hz, 1H), 7,06 (d, J=8,8 Hz, 1H), 6,75 (s, 1H), 6,58 (d, J=7,1 Hz, 1H), 4,74 (d, J=4,3 Hz, 1H), 3,73– 3,67 (m, 1H), 3,63–3,55 (m, 2H), 2,98–2,90 (m, 2H), 1,88 (d, J=9,9 Hz, 2H) , 1,75 (s, 3H), 1,61–1,49 (m, 2H).

Example 14: Method N - Preparation of 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-tetrahydropyran-4-yl-6H-1,6-naphthyridin-5-one (I-36)

For a 2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H)-one (1,2) (70 mg , 0.18 mmol) in THF (2 mL) was added RuPhos Pd G2 (15 mg, 0.018 mmol). The mixture was flushed with nitrogen for 5 minutes and then (tetrahydro-2H-pyran-4-yl)zinc bromide (0.6M in THF, 3 mL, 1.8 mmol) was added. After stirring at 120 °C for 1 hour, the reaction mixture was applied dry on silica gel and purified by column chromatography, eluting with 0-20% 7N methanolic ammonia in DCM to give a crude residue which was purified. further by preparative HPLC to give pure. dae (I-36) (19 mg, 24%) as a yellow solid. m/z=422 [M+H]+,1RMN H (400 MHz, DMSO) δ 12,33 (s, 1H), 11,56–11,50 (m, 1H), 8,21 (s, 1H), 8,10 (d, J=3, 0 Hz, 1H), 7,47 (dd, J= 3,1, 9,0 Hz, 1H), 7,35 (dd, J=5,5, 7,0 Hz, 1H), 6,99 ( d, J=8,9 Hz, 1H), 6,49 (d, J=7,2 Hz, 1H), 4,72 (s, 1H), 4,00-3,94 (m, 2H), 3,69-3,61 (m, 1H), 3,57-3,42 (m, 4H), 2,92-2,82 (m, 3H), 1,88-1,75 (m, 6H ), 1,57-1,46 (M, 2H).

Example 15: Method O - Preparation of 2-(1-acetyl-4-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine -5-one (I-63) and 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(1-methyl-4-piperidyl)-6H-1,6 -Nafthyridine-5-one (I-75)

Schritt 1. 2-(1-Acetyl-4-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one ( I-63)

For a solution of 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(4-piperidyl)-6H-1,6-naphthyridin-5-one (I-34 ) (80 mg, 0.19 mmol) in DCM (5 mL) at 0 °C was added acetic anhydride (0.1 mL). After stirring at room temperature overnight, the reaction was quenched with 2 drops of 2N HCl solution and diluted with water (5 mL). The mixture was passed through an Isolute hydrophobic filter and the organic layer was evaporated in vacuo to give a crude residue which was purified by preparative HPLC to give pure (I-63) (7 mg, 8%) as a solid for delivery. yellow. m/z=463 [M+H]+,1H-NMR (400 MHz, DMSO) &dr; 12,34 (s, 1H), 11,55 (d, J = 2,3 Hz, 1H), 8,37–8,31 (m, 1H), 8,11 (d, J = 6,5 Hz , 1H), 8,06 (d, J=3,3 Hz, 1H), 7,46 (dd, J=3,0, 9,0 Hz, 1H), 7,34 (d, J=7, 3 Hz, 1H), 7,00 (d, J=8,8 Hz, 1H), 6,52 (d, J = 7,3 Hz, 1H), 4,52–4,47 (m, 1H) , 3,95–3,91 (m, 1H), 3,68–3,61 (m, 1H), 3,54–3,46 (m, 2H), 3,18–3,10 (m, 1H), 2,89–2,80 (m, 3H), 2,68–2,60 (m, 1H), 2,04 (s, 3H), 1,89–1,80 (m, 4H) , 1,71–1,44 (m, 4H). (1 Äq. Formiatsalz, 1 austauschbares Proton nicht beobachtet).

Schritt 2. 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(1-methyl-4-piperidyl)-6H-1,6-naphthyridin-5-one ( I-75)

For a solution of 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(4-piperidyl)-6H-1,6-naphthyridin-5-one (I-34 ) (30 mg, 0.07 mmol) in methanol (5 mL) with one drop of 37% formaldehyde solution (11 µL, 0.14 mmol) and sodium cyanoborohydride (5 mg, 0.07 mmol) with a drop of acetic acid). After stirring at room temperature overnight, the mixture was evaporated in vacuo to give a crude residue which was purified by preparative HPLC to give pure (I-75) (17 mg, 54%) as a yellow solid. m/z=435 [M+H]+,1RMN-H (400MHz, DMSO) δ 13.08 (s, 1H), 12.49 (s, 1H), 9.71-9.70 (m, 1H), 8.32-8.28 (m , 1H), 8.15-8.13 (m, 1H), 7.77 (s, 1H), 7.58-7.53 (m, 1H), 7.26-7.20 (m, 1H ), 6.70 (s, 1H), 3.73-3.56 (m, 5H), 3.17-3.08 (m, 3H), 3.02-2.94 (m, 2H), 2.87 (s, 3H), 2.24-2.20 (m, 2H), 2.01-1.82 (m, 4H), 1.55-1.45 (m, 2H). (1 eq. formate salt, 1 exchangeable proton not observed).

Example 16: Method P - Preparation of 2-(2,8-diazaspiro[4,5]decan-8-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino] . -6H -1,6-naphthyridin-5-one (I-235) and 2-[2-(cyclopropanocarbonyl)-2,8-diazaspiro[4,5]decan-8-yl]-4-[[5- (4-Hydroxy )-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one (I-233)

Schritt 1. tert.-butyl-8-(4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridine -2-yl)-2,8-diazaspiro[4.5]decan-2-carboxylate (16.0)

A reaction tube was treated with 2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H)-one (1, 2) (250 mg, 0.67 mmol), tert-butyl 2,8-diazaspiro[4,5]decane-2-carboxylate (323 mg, 1.34 mmol) in DMSO (6 mL). After stirring at 120°C overnight, the reaction mixture was diluted with ethyl acetate and washed with brine. The organic layer was dried (MgSO4), filtered and the solvent evaporated in vacuo to give a crude residue which was purified by column chromatography eluting with 0-100% ethyl acetate in hexane to give 16.0 (300 mg) with a purity of about 80% , which was used in the next step without further purification.

Schritt 2. 2-(2,8-Diazaspiro[4.5]decan-8-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6- Naphthyridine-5-on (I-235)

The reaction was carried out according to the procedure described in Example 7, Method G using 8-(4-((5-(4-Hydroxypiperidin-1-yl)pyridin-2-yl)amino)-5-oxo -5- 6-Dihydro-1,6-naphthyridin-2-yl)-2,8-diazaspiro[4,5]decane-2-carboxylate tert-butyl ester (16.0) to give I-235 as a yellow gum. m/z=476 [M+H]+,112.31 (s, 1H), 11.21 (s, 1H), 8.43 (s, 1H), 8.08 (d, J=2.5Hz, 1H), 7.99 (s, 1H ), 7.48 (dd, J=2.5, 8.8Hz), 1H), 7.26 (d, J=7.3Hz, 1H), 6.93 (d, J=8.8 Hz, 1H), 6.28 (d, J=7.3 Hz, 1H), 4.75 (s, 1H), 3.75-3.49 (m, 6H), 3.23 (s, 1H ), 3.19 (t, J=7.0Hz, 2H), 2.97 (s, 2H), 2.90 (t, J=11.2Hz, 2H), 1.90–1.77 (m, 4H), 1.66–1.51 (m, 6H). (1 Äq. Formiatsalz, 1 austauschbares Proton nicht beobachtet)

Schritt 3. 2-[2-(Cyclopropancarbonyl)-2,8-diazaspiro[4.5]decan-8-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]- 6H-1,6-Nafthyridin-5-on (I-233)

The reaction was carried out according to the procedure described in Method CB2, Step 1 using 2-(2,8-diazaspiro[4.5]decan-8-yl)-4-[[5-(4-hydroxy-1 -piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one (I-235) and cyclopropanecarbonyl chloride to give I-233 as a yellow solid. m/z=544 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,26 (d, J=2,6 Hz, 1H), 11,15 (s, 1H), 8,04 (dd, J=2,4, 2,4 Hz, 1H), 7,97 (d, J=4,9 Hz). , 1H), 7,44 (dd, J=3,0, 8,9 Hz, 1H), 7,25–7,19 (m, 1H), 6,90 (d, J=9,0 Hz, 1H), 6,24 (dd, J=1,6, 7,2 Hz, 1H), 4,71-4,70 (m, 1H), 3,78-3,41 (m, 10H), 3 ,26 (s, 1H), 2,89-2,80 (m, 2H), 1,92-1,75 (m, 5H), 1,63-1,46 (m, 6H), 0,76 -0,70 (m, 4H).

Example 17: Method Q - Preparation of 4-[[3-(2,6-difluorophenyl)-8-oxo-7H-2,7-naphthyridin-1-yl]amino]-N-ethylbenzamide (I-7)

The title compound was prepared from 6,8-dichloro-2,7-naphthyridin-1(2H)-one (17.0) according to the methods described in Tett Lett, 2013 54(15), p. 2014 and WO2012097683 using 4-amino-N-ethylbenzamide and 2-bromo-1,3-difluorobenzene to give I-7. m/z=421 [M+H]+,112,25 (s, 1H), 11,99 (s, 1H), 8,30 (dd, J=5,5, 5,5Hz, 1H), 7,89 (d, J=8,9Hz, 2H ), 7,82 (d, J=8,8 Hz, 2H ), 7,64–7,56 (m, 1H), 7,51 (d, J=6,9 Hz, 1H), 7,31 ( dd, J=8,2, 8,2 Hz, 2H), 7,19 (s, 1H), 6,63 (d, J=7,0Hz, 1H), 3,32–3, 23 (m, 2H), 1,12 (t, J=7,2Hz, 3H).

Example 18: Method R - Preparation of 7-(2,6-difluorophenyl)-5-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-3H-pyrido[2,3-d ] pyrimidin-4-one (I-11)

Step 1. 5,7-Dichloro-3-(4-methoxybenzyl)pyrido[2,3-d]pyrimidin-4(3H)-one (18.1)

The reaction was carried out according to the procedure described in Example 10, Method J, Step 1 using 5,7-dichloropyrido[2,3-d]pyrimidin-4(3H)-one (18.0) (described in WO2012097683) to give 18.1 as a yellow solid.1RMN-H (400 MHz, DMSO) 8,95–8,93 (1H, m), 7,91–7,89 (1H, m), 7,45–7,39 (2H, m), 6, 99–6,93 (2H, m), 5,15–5,12 (2H, m), 3,79-3,77 (3H, m).

Schritt 2. 7-chloro-5-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-3-(4-methoxybenzyl)pyrido[2,3-d]pyrimidin-4( 3H)-eins (18.2)

The reaction was carried out according to the procedure described in Example 1, Method A, Step 2 using 5,7-dichloro-3-(4-methoxybenzyl)pyrido[2,3-d]pyrimidin-4(3H) -one ( 18.1) and carried out 1-(6-aminopyridin-3-yl)piperidin-4-ol to give 18.2 as a brown solid.1RMN-H (400 MHz, DMSO) 11,78-11,75 (1H, m), 8,83 (1H, s), 8,48-8,46 (1H, m), 8,20-8, 16 (1H, m), 7,52 (1H, q, J= 3,8 Hz), 7,42 (2H, d, J=8,6 Hz), 7,14–7,09 (1H, m ), 7,01–6,95 (2H, m), 5,20–5,16 (2H, m), 4,77–4,73 (1H, m ), 3,78 (3H, s), 3,74-3,68 (1H, m), 3,62-3,54 (2H, m), 2,97-2,91 (2H, m), 1,88 (2H, d, J=14 ,0 Hz), 1,57- 1,48 (2H, m).

Schritt 3. 7-(2,6-difluorophenyl)-5-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-3-(4-methoxybenzyl)pyrido[2,3- d]pyrimidin-4(3H)-one (18,3)

For a carousel tube loaded with 7-chloro-5-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-3-(4-methoxybenzyl)pyrido[2,3-d] pyrimidine -4(3H)-one (18.2) (140 mg, 0.28 mmol), 2,6-difluorophenyltributyltin (229 mg, 0.56 mmol) and dioxane (2 mL) washed with N2was added. Copper I iodide (11 mg, 0.05 mmol) and bis(triphenylphosphine)palladium chloride (20 mg, 0.02 mmol). After stirring at 120°C overnight, the reaction was cooled and filtered through celite. The volatiles were removed under reduced pressure and the crude residue was purified by trituration with diethyl ether to give 18.3 (120 mg, 74%) as a brown solid.1RMN-H (400 MHz, DMSO) 11,80 (1H, s), 8,84–8,80 (1H, m), 8,54–8,52 (1H, m), 8,12–8, 08 (1H, m), 7,67–7,59 (1H, m) , 7,53-7,40 (3H, m), 7,31 (2H, t, J=8,0 Hz), 7 ,15-7,09 (1H, m), 7,02-6,95 (2H, m), 5,23-5,19 (2H, m), 4,72 (1H, d, J=4, 0 Hz), 3,80–3,77 (3H, m), 3,69–3,62 (1H, m), 3,56 (2H, d, J=12,6 Hz), 2,94– 2,86 (2H, m), 1,87-1,85 (2H, m), 1,55-1,47 (2H, m).

Etapa 4. 7-(2,6-Difluorophenyl)-5-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-3H-pyrido[2,3-d]pyrimidin-4- un (I-11)

The reaction was carried out according to the procedure described in Example 10. Method J, Step 4 using 7-(2,6-difluorophenyl)-5-((5-(4-hydroxypiperidin-1-yl)pyridin- 2-yl)amino)-3-(4-methoxybenzyl)pyrido[2,3-d]pyrimidin-4(3H)-one (18.3) to give I-11 as a yellow solid. m/z=451 [M+H]+,1RMN-H (400 MHz, DMSO) δ 11,87-11,82 (m, 1H), 8,38 (s, 1H), 8,22 (s, 1H), 7,98 (d, J=2 ,8 Hz, 1H), 7,55-7,46 (m, 1H), 7,39 (dd, J=2,9, 9,0 Hz, 1H), 7,19 (dd, J=8, 0, 8,0 Hz, 2H), 6,96 (d, J=8,8 Hz, 1H), 4,60-4,60 (m, 1H), 3,59–3,51 (m, 1H ), 3,48–3,39 (m, 3H), 2,83–2,74 (m, 2H), 1,75–1,71 (m, 2H), 1,45–1,33 (m , 2H).

Example 19: Method S - Preparation of 4-[[7-(2,6-difluorophenyl)-4-oxo-3H-pyrido[4,3-d]pyrimidin-5-yl]amino]-N-ethyl-benzamide (I-12)

Etapa 1. 6-((7-Chloro-4-oxo-3,4-di-hydropyrido[4,3-d]pyrimidin-5-yl)amino)-N-ethylnicotinamida (19.1)

The reaction was carried out according to the procedure described in Example 1, Method A, Step 2 using 5,7-dichloropyrido[4,3-d]pyrimidin-4(3H)-one (19.0) and 4-amino -N-ethylbenzamide was carried out to give (19.1) to 19.1 as a brown solid.1RMN-H (400 MHz, DMSO) 13,01–12,99 (1H, m), 11,56–11,50 (1H, m), 8,43 (1H, t, J = 5,4 Hz) , 8,36 (1H, s), 7,96–7,85 (4H, m), 7,04–7,02 (1H, m), 3,36–3,30 (2H, m), 1 ,21–1,14 (3H,m).

Step 2. 4-[[7-(2,6-difluorophenyl)-4-oxo-3H-pyrido[4,3-d]pyrimidin-5-yl]amino]-N-ethyl-benzamide (I-12)

The reaction was carried out according to the procedure described in Example 18: Method R, Step 3 using 6-((7-Chloro-4-oxo-3,4-dihydropyrido[4,3-d]pyrimidin-5 -yl)amino)-N-ethylnicotinamide (19.1) to give I-12 as a white solid. m/z=422 [M+H]+,1H RMN (400 MHz, DMSO) ? 11,67 (s, 1H), 8,25-8,20 (m, 2H), 7,81 (d, J=8,3Hz, 2H), 7,74 (d, J=8,3Hz, 2H ), 7,57 -7,48 (m, 1H), 7,22 (dd, J=8,1, 8,1 Hz, 2H), 7,02 (s, 1H), 3,19 (q, J=6,7 Hz, 2H), 1,04 (dd, J=6,8, 6,8 Hertz, 3H). Uma ressonância NH não foi observada

Example 20: Method T - Preparation of N-ethyl-4-[(2-isopropyl-5-oxo-6H-pyrido[4,3-d]pyrimidin-4-yl)amino]benzamide (I-10)

Title compounds were prepared from ethyl 4-chloro-2-isopropyl-6-methylpyrimidine-5-carboxylate (20.0) according to procedures described in KR2016035411 using 4-amino-N-ethylbenzamide to give I -10 as one to give yellow solid. m/z=352 [M+H]+,1H-NMR (400 MHz, DMSO) δ 12.13 (s, 1H), 12.00 (s, 1H), 8.41 (dd, J=5.4, 5.4 Hz, 1H), 7, 98 (d, J=8.9Hz, 2H), 7.91 (d, J=8.8Hz, 2H), 7.62 (d, J=7.2Hz, 1H), 6.48 ( d, J=7.2Hz, 1H), 3.32-3.26 (m, 2H), 3.09-2.99 (m, 1H), 1.33 (d, J=6.9Hz , 6H), 1.14 (dd, J=7.2, 7.2Hz, 3H).

Example 21: Method U - Preparation of 5-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-7-isopropyl-3H-pyrido[2,3-d]pyrimidin-4-one (I-59) and 5-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-7-propyl-3H-pyrido[2,3-d]pyrimidin-4-one (I -60)

Step 1. 5-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-7-isopropyl-3H-pyrido[2,3-d]pyrimidin-4-one (I-59) e 5-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-7-propyl-3H-pyrido[2,3-d]pyrimidin-4-one (I-60)

For a microwave tube loaded with 7-chloro-5-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-3-(4-methoxybenzyl)pyrido[2,3-d] pyrimidine-4(3H)-one (18.2) (100 mg, 0.2 mmol), tetrakis(triphenylphosphine)palladium(0) (23 mg, 0.02 mmol) in THE (2 mL), 2-bromide -propylzinc (0.5 M solution in THF, 4.1 mL, 2.03 mmol). After stirring at 120°C for 1 hour, the reaction was combined with a duplicate reaction and quenched by addition of methanol (1 mL) and diluted with DCM. The mixture was absorbed directly onto silica gel and purified by column chromatography, eluting with 0-10% methanol in DCM to give 5-((5-(4-Hydroxypiperidin-1-yl)pyridin-2-yl) amino) -7- isopropyl to give -3-(4-methoxybenzyl)pyrido[2,3-d]pyrimidin-4(3H)-one (200 mg) with a purity of ca. 80%, which was used in the next step without further purification.

Crude 5-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-7-isopropyl-3-(4-methoxybenzyl)pyrido[2,3-d]pyrimidine-4(3H) -um (200 mg) was dissolved in TFA (5 ml). After stirring at reflux for 4 hours, the reaction solvent was removed in vacuo to give a crude product which was diluted in DCM (1 mL), methanol (2 mL), and 1 M methanolic ammonia (4 mL) [caution! ] and stirred for 1 hour at room temperature. ambient before evaporating back to dryness. The residue was purified by preparative HPLC to give the product as a mixture of isomers (I-59 and I-60). The isomers were then separated by preparative SFC to give (I-59) (4 mg, 3%) and (I-60) (7 mg, 6%) both as a yellow solid. I-59: m/z=381 [M+H]+,1RMN H (400 MHz, DMSO) δ 12,06-11,99 (m, 1H), 8,26 (s, 1H), 8,21 (s, 1H), 8,14 (d, J=2, 8 Hz, 1H), 7,51 (dd, J=2,9, 9,0 Hz, 1H), 7,05 (d, J=9,1 Hz, 1H), 4,74 (s, 1H), 3,72–3,66 (m, 1H), 3,62–3,53 (m, 2H), 3,02–2,88 (m, 3H), 1,93–1,87 (m, 2H), 1,61-1,50 (m, 2H), 1,30 (d, J=7,1 Hz, 6H). 1 Protón intercambiable no observado. I-60 (7 mg, 6%) como un sólido amarillo. m/z=381 [M+H]+,1RMN-H (400 MHz, DMSO) δ 12,55 (s, 1H), 11,78 (s, 1H), 8,27 (s, 1H), 8,18–8,13 (m, 2H), 7,51 (dd, J=2,9, 9,0 Hz, 1H), 7,07 (d, J=8,8 Hz, 1H), 4,74 (d, J=3,5 Hz, 1H) ), 3,71–3,67 (m, 1H), 3,62–3,53 (m, 2H), 2,97–2,87 (m , 2H), 2,70 (dd, J=7 ,6, 7,6 Hz, 2H), 1,93–1,87 (m, 2H), 1,76 (dd, J=7,3, 14,9 Hz, 2H), 1,61–1, 49 (m, 2H), 0,99 (dd, J = 7,5, 7,5 Hz, 3H).

Example 22: AP Method - Preparation of 6-(2-Fluoro-5-isopropylphenyl)-8-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)isoquinoline-1(2H)- one (I-288)

Step 1. Preparation of 4-bromo-2-iodo-6-methylbenzoic acid (22.1)

To a stirred solution of 4-bromo-2-methylbenzoic acid (22.0) (15 g, 69.7 mmol, 1.0 eq) and iodobenzene diacetate (44.93 g, 139.5 mmol, 2.0 eq) in N,N-dimethylformamide (150 mL) at RT was treated with palladium(II) acetate (0.78 g, 3.48 mmol, 0.05 eq) followed by iodine (17.7 g, 69, 7 mmol, 1.0 eq) added. After stirring at 100 °C for 16 h, the reaction mixture was poured into ice-water (500 mL) and extracted with ethyl acetate (200 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 20% ethyl acetate in hexane to give 22.1. (11.25g, 47.31%). MS (ES): m/z 341.84 [M+H]+.

Step 2. 4-Bromo-N-(2,4-dimethoxybenzyl)-2-iodo-6-methylbenzamide (22.2)

N,N-Diisopropylethylamine (5.6 g, 44.10 mmol, 3.0 eq) and bromo-tris-pyrrolidino-phosphonium hexafluorophosphate (12.3 g, 26.46 mmol, 1.8). After stirring at room temperature for 30 min, 2,4-dimethoxybenzylamine (3.2 g, 19.11 mmol, 1.3 eq) was added. After stirring at room temperature for 16 h, the reaction mixture was poured into water (300 mL) and extracted with ethyl acetate (200 mL x 3). The combined organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 35% ethyl acetate in hexane to give 22.2 (6.5 g, 90.43%). MS (ES): m/z 492.72 [M+H]+.

Stage 3. 6-Bromo-2-(3,4-dimethylbenzyl)-8-iodioisoquinolin-1(2H)-one (22,3) .

To a solution of 4-bromo-N-(2,4-dimethoxybenzyl)-2-iodo-6-methylbenzamide (22.2) (2.5 g, 5.10 mmol, 1.0 eq.) in THF ( 50 mL) 2.5 M lithium diisopropylamide in THE (7.6 mL, 15.306 mmol, 3 eq) was added at -78 °C. After stirring at -78 °C for 30 min, N,N-dimethylformamide (1.49 g, 20.40 mmol, 4 eq) was added. After stirring at -78°C for 1 hour, the reaction mixture was acidified with hydrochloric acid and extracted with ethyl acetate (100ml x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 12% ethyl acetate in hexane to give 22.3 (1.5 g, 62.82%). MS (ES): m/z 500.85 [M+H]+.

Step 4. 6-Bromo-2-(3,4-dimethylbenzyl)-8-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)isoquinoline-1(2H)-one (22 ,4 )

To a solution of 6-bromo-2-(3,4-dimethylbenzyl)-8-iodisoquinolin-1(2H)-one (22.3) (0.7 g, 1.40 mmol, 1.0 eq.) 1-(6-aminopyridine-3-yl)piperidin-4-ol (0.325 g, 1.68 mmol, 1.2 eq) and cesium carbonate (1.36 g, 4 0.20 mmol, 3.0 eq ) added. After 10 minutes of degassing. Under argon atmosphere, tris(dibenzylideneacetone)dipalladium(0) (0.064 g, 0.07 mmol, 0.05 eq) and 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (0.08 g, 0.070 mmol, 0.1 eq) and degassed again for 5 min. After stirring at 140 °C for 1 h in the microwave, the reaction mixture was cooled to room temperature, poured into water (100 mL) and extracted with ethyl acetate (100 mL x 3). The combined organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by combi-flash using 75-80% ethyl acetate in hexane to give 22.4 (0.250 g, 31.34%). MS(ES): m/z 566.25 [M+H]+.

Step 5. 2-(3,4-Dimethylbenzyl)-6-(2-fluoro-5-isopropylphenyl)-8-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)isoquinoline- 1(2H)-ene (22.5)

For a solution of 6-bromo-2-(3,4-dimethylbenzyl)-8-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)isoquinolin-1(2H)-one ( 22.4) (0.1 g, 0.17 mmol, 1.0 eq) and (2-fluoro-5-isopropylphenyl)boronic acid (0.038 g, 0.21 mmol, 1.2 eq) in 1,4-dioxane HCl (5 mL) and water (1 mL), potassium phosphate (0.75 g, 0.35 mmol, 2 eq) was added. After degassing under argon for 10 min, [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (0.07 g, 0.008 mmol, 0.05 eq) was obtained. After stirring at 110 °C for 1 hour in the microwave, the reaction mixture was diluted with water (50 ml) and extracted with ethyl acetate (100 ml x 3). The combined organic layer was washed with brine, dried over sodium, sulfated, and concentrated under reduced pressure. The residue was purified by combined flash column chromatography, eluting with 65% ethyl acetate in hexane to give 22.5 (0.1 g, 90.30%), MS(ES): m/z 624.24 [ M+H]+

Schritt 6. 1-(6-((6-(2-Fluoro-5-isopropylphenyl)-1-oxo-1,2-dihydroisoquinolin-8-yl)amino)pyridin-3-yl)piperidin-4-yl 2 ,2,2-trifluoroacetate (22,6)

To a solution of 2-(3,4-dimethylbenzyl)-6-(2-fluoro-5-isopropylphenyl)-8-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)isoquinoline Trifluoroacetic acid (1.4 mL) was added to -1(2H)-one (22.5) (0.07 g, 0.11 mmol, 1.0 eq.) in DCM (2 mL) at room temperature. After stirring at 55 °C for 16 h, the reaction mixture was evaporated in vacuo to give 22.6 (0.07 g, quantitative), which was used in the next step without further purification. MS (ES): m/z 569.65 [M+H]+.

Schritt 7. 6-(2-Fluoro-5-isopropylphenyl)-8-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)isoquinolin-1(2H)-one (I-288 )

For a solution of 1-(6-((6-(2-Fluoro-5-isopropylphenyl)-1-oxo-1,2-dihydroisoquinolin-8-yl)amino)pyridin-3-yl)piperidin-4-yl 2,2,2-Trifluoroacetate (22.6 g) (0.07 g, 0.123 mmol, 1.0 eq.) in THF (3 mL) was treated at room temperature with sodium hydroxide (0.014 g, 0.36 mmol, 3.0 eq.) compensated. After stirring at room temperature for 2h, the reaction was concentrated in vacuo. The residue was purified by preparative HPLC using column: SUNFIRE C18 (250 × 19) mm 5u, mobile phase: 0.1% trifluoroacetic acid in water/acetonitrile as buffer, flow 15 mL/min, gradient 0-40% over 25 min. fractions were lyophilized to give the TFA salt. The salt was dissolved in methanol (3 mL) and neutralized with polymer-bound tetraalkylammonium carbonate (basic resin) to give the free base 22 (0.025 g, 42.97%). MS (ES): 473.87 m/z [M+H]+, LCMS purity: 100%, HPLC purity: 95.07% 1H-NMR (400 MHz, DMSO-d6) δ 1.26 (d, J =7.0 Hz, 6H) , 1.58 (s, 2H), 1.88 (s, 2H), 2.95-3.03 (m, 2H), 3.52 (s, 2H), 3 .69 (s, 1H), 6.63 (d, J=6.9 Hz, 1H ), 7.08 (s, 1H), 7.17-7.32 (m, 3H), 7.35 ( s, 1H), 7.43 (d, J=7.3 Hz, 1H), 7.67 (s, 1H), 8.10 (s, 1H), 8.61 (s, 1H), 11, 48 (s, 1H), 12.47 (s, 1H).

Example 23: BP Method - Preparation of N-(3-Fluoro-4-(4-((6-Fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-5-oxo -5,6-dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (I-312) .

Stage 1. Preparation of 2,4-dichloro-1,6-naphthyridin-5(6H)-one (23.1)

potassium tert-butoxide (23.79 g, 0.2136 mol, 2.0 eq.) and 1,3,5-triazine (16.25 g, 0.216 mol, 2.0 eq.). After stirring at 90 °C for 1 h, the reaction mixture was concentrated in vacuo. The residue was dissolved in ice water and acidified to pH 2-3 with 1N hydrochloric acid. After stirring at room temperature for 1h, the reaction mixture was filtered and washed with water (300ml). Additional product was extracted from the aqueous phase using DCM (500 mL x 3) to give crude material which was concentrated and then azeotroped with toluene (100 mL) to give 23.1 (18.5 g, yield: 80.55% ) as a result. MS (ES): m/z 216 [M+H]+.

Step 2. 2,4-Dichloro-6-(4-methoxybenzyl)-1,6-naphthyridin-5(6H)-one(23,2)

To a solution of 2,4-dichloro-1,6-naphthyridin-5(6H)-one (23.1) (10.0 g, 0.0465 mol, 1.0 eq.) in N,N-dimethylformamide HCl (40 mL) was added cesium carbonate (30.23 g, 0.093 mol, 2.0 eq.). After stirring at room temperature for 30 min, 1-(chloromethyl)-4-methoxybenzene (7.25 g, 0.0465 mol, 1.0 eq) was added. After stirring at room temperature for 4h, the reaction mixture was diluted with cold water (100ml), causing a solid to precipitate from solution. The solid was filtered and washed with water (50 mL) and hexane (50 mL). The solid was dried under reduced pressure to give 23.2 (9.98 g, yield: 64.03%). MS (ES): m/z 316.18 [M+H]+.

Schritt 3. N-(4-(4-Cloro-6-(4-metoxibenzil)-5-oxo-5,6-dihidro-1,6-naftiridin-2-il)-3-fluorfenil)ciclohexancarboxamida (23.3)

For a solution of 2,4-dichloro-6-(4-methoxybenzyl)-1,6-naphthyridin-5(6H)-one 23.1 (11.5 g, 0.0344 mol, 1.0 eq.) and N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborlan-2-yl)phenyl)cyclohexanecarboxamide (11.91 g, 0.0344 mol, 1.0 eq) in 1,2-dimethoxyethane (160 mL) and water (~40 mL) was added potassium phosphate (21.89 g, 0.1032 mol, 3.0 eq). After degassing with argon gas for 20 min, tetrakis(triphenylphosphine)palladium(0) (1.98 g, 0.0017 mol, 0.05 eq) was added. After stirring at 100 °C for 16 h, the reaction mixture was poured into water (500 ml) and extracted with ethyl acetate (500 ml x 2). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography eluting with 25% ethyl acetate in hexane to give 23.3 (5.2 g, yield: 28.03%). MS (ES): m/z 521.00 [M+H]+.

Schritt 4. N-(3-Fluoro-4-(4-((6-Fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-6-(4-methoxybenzyl)- . 5-oxo-5,6-dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (23.4) .

To a solution of N-(4-(4-chloro-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro-1,6-naphthyridin-2-yl)-3-fluorophenyl)cyclo-hexanecarboxamide 23.3 (0.30 g, 0.576 mmol, 1.0 eq) in 1,4-dioxane (5 mL) was 6-fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-amine ( PA9) (0.11 g, 0.634 mmol, 1.1 eq.) and potassium carbonate (0.22 g, 1.728 mmol, 3.0 eq.). After degassing with nitrogen gas for 10 min, 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (0.10 g, 0.115 mmol, 0.2 eq) and tris(dibenzylideneacetone)dipalladium (0.033 g, 0.057 mmol , 0.1 eq) obtained under a nitrogen gas atmosphere. After stirring at 120 °C for 16 h, the reaction mixture was diluted with water (100 mL) and extracted into ethyl acetate (100 mL x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified using combi-flash silica eluting with 7% methanol/DCM to give 23.4 (0.15 g, 39.17%). MS (ES): m/z 664.6 [M+H]+

Schritt 5. N-(3-Fluoro-4-(4-((6-Fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-5-oxo-5,6- Dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (I-312) .

For a solution of N-(3-Fluoro-4-(4-((6-Fluoro-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-6-(4-methoxybenzyl) -5-oxo-5,6-dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide 23.4 (0.14 g, 0.211 mmol, 1.0 eq) in DCM (4 mL) at room temperature it was triflic acid (0.54 g, 3.625 mmol, 17.1 eq.) and trifluoroacetic acid (0.55 g, 4.853 mmol, 23.0 eq.). After stirring at room temperature for 30 min, the reaction mixture was concentrated in vacuo. The residue was purified by preparative HPLC using: SUNFIRE C18 column (150 x 19) mm 5p Mobile phase: (A) 0.1% trifluoroacetic acid (TFA) in water and (B) acetonitrile, flow rate 13 mL/min, 0 -34% gradient over 23 min to give the TFA salt I-312 after lyophilization. The salt was dissolved in methanol (3 mL) and neutralized with polymer-bound tetraalkylammonium carbonate (basic resin) to isolate I-312 free base (0.042 g, 36.63%). MS (ES): 542.99 m/z [M -H]+, LCMS purity: 98.66%, HPLC purity: 99.43%, 1H-NMR (400 MHz, DMSO-d6) δ 1.15 –1.28 (m, 4H), 1.35–1.41 (m, 2H), 1.63–1.82 (m, 5H), 2.36 (d, J=12.9 Hz, 1H ), 2.43 (s, 2H), 2.74 (s, 2H), 2.88 (s, 2H), 3.60 (s, 2H), 6.56 (d, J=7.3 Hz , 1H), 7.04 (s, 1H), 7.19 (d, J=11.2 Hz, 1H), 7.32 (d, J=8.0 Hz, 1H), 7.42 (d , J=7.7 Hz, 2H), 7.72 (d, J=14.6 Hz, 1H), 7.93-8.06 (m, 1H), 10.23 (s, 1H), 11 .38 (s, 1H), 11.62 (s, 1H).

Example 24: CP Method - Preparation of N-(3-Fluoro-4-(4-((5-(4-hydroxy-4-methylpiperidin-1-yl)pyridin-2-yl)amino)-5-oxo- 5,6-dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (I-294)

Schritt 1. 2-Chloro-4-((5-(4-hydroxy-4-methylpiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H)-on (24.1)

For the stirred solution of 2,4-dichloro-1,6-naphthyridin-5(6H)-one (1,2) (0.50 g, 2.325 mmol, 1.5 eq.) and 1-(6-aminopyridine -3-yl )-4-methylpiperidin-4-ol (0.53 g, 2.558 mmol, 1.1 eq) in tert-butanol (10 mL) and diisopropylethylamine (0.60 g, 4.650 mmol, 2, 0 eq). After microwave heating at 150°C for 6 hours, the reaction mixture was diluted with water (50ml) and extracted with ethyl acetate (100ml x 2). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by preparative HPLC with X-SELECT PENYL HEXYL (150 x 19) mm, 5 µm. Column flow at 15.0 mL/min with mobile phase (A) 0.1% formic acid in water and (B) 100% acetonitrile with 0-43% solvent B gradient over 18 min to give 24 .1 (0.11 g, 12.26%) MS (ES): m/z 387.2 [M+H]+

Schritt 2. N-(3-Fluor-4-(4-((5-(4-hidroxy-4-methylpiperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-di -hidro -1,6-naftiridin-2-yl)phenyl)cyclohexancarboxamida (I-294)

For a solution of 2-chloro-4-((5-(4-hydroxy-4-methylpiperidin-1-yl)pyridin-2-yl)amino)-1,6-naphthyridin-5(6H)-one 24, 1 (0.10 g, 0.261 mmol, 1.0 eq) and N-(3-Fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl) Cyclohexanecarboxamide (0.13 g, 0.392 mmol, 1.5 eq) in 1,4-dioxane (6.0 mL) and water (1.0 mL) At room temperature tribasic potassium phosphate (0.39 g, 1.831 mmol, 7th 0.0 eq). After degassing for 20 min using argon gas, X-fos-aminobiphenylpalladium chloride precatalyst (0.020 g, 0.0261 mmol, 0.1 eq.) was added. After stirring at 150°C in a microwave for 20 minutes, the reaction mixture was poured into water (25ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was washed with brine (25 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by column chromatography using a Redisep gold column eluting with 15% methanol in DCM to give I-294 (0.10 g, 67.61%), MS(ES): 556.82 m/z. [M+H]+, LCMS purity: 95.09%, HPLC purity: 95.73% 1H NMR (400 MHz, DMSO-d6) δ 1.18 (s, 3H), 1.22-1, 34 (m, 3H), 1.37-1.51 (m, 2H), 1.59 (s, 4H), 1.59 (d, J=11.4 Hz, 1H), 1.68 (d, J=10.6Hz, 1H), 1.79 (d, J=11.7Hz, 2H), 1.85 (d, J=12.7Hz, 1H), 2.40-2.35 ( m, 2H), 3.14 (s, 2H), 3.08–3.19 (m, 1H), 3.32 (d, J=11.3 Hz, 2H), 4.35 (s, 1H ), 6.58 (d, J = 7.3 Hz). , 1H), 7.03 (d, J=8.9 Hz, 1H), 7.37-7.53 (m, 3H), 7.72-7.81 (m, 1H), 8.01 ( t, J=8.8 Hz, 1H), 8.09 (d, J=3.1 Hz, 1H), 8.74 (s, 1H), 10.25 (s, 1H), 11.65 ( s, 1H), 12.39 (s, 1H).

Example 25: DP Method - Preparation of N-(3-Fluoro-4-(4-((5-(1-methylpiperidin-3-yl)pyridin-2-yl)amino)-5-oxo-5,6- Dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (I-337)

Schritt 1. tert-butyl-3-(6-((2-(4-(cyclohexanecarboxamido)-2-fluorophenyl)-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro-1,6 - . naphthyridin-4-yl)amino)pyridin-3-yl)piperidin-1-carboxylate (25.1) .

To a solution of N-(4-(4-chloro-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro-1,6-naphthyridin-2-yl)-3-fluorophenyl)cyclo-hexanecarboxamide 23.3 (0.50 g, 0.96 mmol, 1.0 eq) in 1,4-dioxane (5.0 mL) was added sodium 3-(6-aminopyridin-3-yl)piperidine-1-carboxylate. tert-butyl (0.26 g, 0.96 mmol, 1.0 eq) and cesium carbonate (0.93 g, 2.8 mmol, 3.0 eq). After 10 minutes of degassing. Under argon atmosphere, tris(dibenzylideneacetone)dipalladium(0) (0.087 g, 0.09 mmol, 0.1 eq) and 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (0.111 g, 0. 19 mmol, 0.2 eq) and degassed again for 5 min. After stirring at 1H10C for 1 h, the reaction mixture was poured into water (100 mL) and extracted with ethyl acetate (100 mL x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by combi-flash elution with 35-40% ethyl acetate in hexane to give 25.1 (0.5 g, yield: 68.34%). MS(ES): m/z 761.94 [M+H]+.

Step 2. N-(3-Fluoro-4-(6-(4-methoxybenzyl)-5-oxo-4-((5-(piperidin-3-yl)pyridin-2-yl)amino)-5,6 -Dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (25.2)

For a solution of tert-butyl-3-(6-((2-(4-(cyclohexanecarboxamido)-2-fluorophenyl)-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro-1,6 25.1 -Naphthyridin-4-yl)amino)pyridin-3-yl)piperidine-1-carboxylate (0.24 g, 0.31 mmol, 1.0 eq.) in DCM (2 mL) at 0 °C treated with trifluoroacetic acid (1 ml). After stirring at room temperature for 15 min, the reaction mixture was poured into water (50 mL), neutralized with saturated sodium bicarbonate solution (15 mL) and extracted with DCM (50 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by trituration using diethyl ether (10 mL x 2) to give 25.2 (0.14 g, 67.17%). MS(ES): m/z 661.7 [M+H]+.

Paso 3. N-(3-Fluoro-4-(6-(4-methoxybenzyl)-4-((5-(1-metilpiperidin-3-yl)pyridin-2-yl)amino)-5-oxo-5 ,6-Dihidro-1,6-naphthiridin-2-yl)fenil)ciclohexancarboxamide (25.3)

For a solution of N-(3-Fluoro-4-(6-(4-methoxybenzyl)-5-oxo-4-((5-(piperidin-3-yl)pyridin-2-yl)amino)-5, 6-Dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (25.2) (0.14 g, 0.21 mmol, 1.0 equiv.) in methanol (2 mL) was treated with p- formaldehyde (0.019 g, 0.63 mmol, 3.0 equiv.), anhydrous zinc chloride (0.043 g, 0.31 mmol, 1.5 equiv.), and triethylamine (0.85 mL, 0.63 mmol, 3.0 eq.) . After stirring at 60 °C for 2 h, the reaction mixture was cooled to room temperature and sodium cyanoborohydride (0.033 g, 0.53 mmol, 2.5 eq) was added. After stirring overnight, the reaction mixture was poured into water (100 mL) and extracted with ethyl acetate (100 mL x 3). The combined organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by combi flash elution with 2-3% methanol in DCM to give 25.3 (0.1 g, 69.94%). MS(ES): m/z 675.8 [M+H]+.

Paso 4. N-(3-Fluoro-4-(4-((5-(1-methylpiperidin-3-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6 -naftiridin-2-yl)phenyl)cyclohexancarboxamida (I-337)

For a solution of N-(3-Fluoro-4-(6-(4-methoxybenzyl)-4-((5-(1-methylpiperidin-3-yl)pyridin-2-yl)amino)-5-oxo- 5,6-dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide (25.3) (0.1 g, 0.14 mmol, 1.0 eq) in DCM (1.0 mL). 0°C was converted to trifluoromethanesulfonic acid (∼0.6 mL). After stirring at room temperature for 0.5 h, the reaction mixture was poured into water (50 mL), neutralized with saturated bicarbonate solution (15 mL) and extracted with DCM (50 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC using X-SELECT PENYL HEXYL (150 x 19) mm, 5p mobile column with mobile phase (A): 0.1% formic acid in water/acetonitrile and (B) acetonitrile, flow rate 15 mL /min. , clean. Slope 0-22% for 16 min. The desired fractions were lyophilized to give the formate salt of I-337. The salt was dissolved in methanol (3 mL) and neutralized with polymer-bound tetraalkylammonium carbonate (basic resin) to give I-337 free base (0.025 g, 30.42%). MS (ES): 555.8 m/z [M+H]+, LCMS purity: 100%, HPLC purity: 100%,1RMN-H (400 MHz, DMSO-d6) δ 1,25-1,30 (m, 5H), 1,39-1,51 (m, 3H), 1,69-1,77 (m, 2H), 1,78-1,86 (m , 6H), 2,40 (s, 3H), 2,87-3,01 (s , 3H), 6,62 (d, J=7,2 Hz, 1H), 7,08 (d, J= 8,5 Hz, 1H), 7,40-7,50 (m, 2H), 7,71-7,81 (m, 2H), 8,01-8,05 (m, 1H), 8,32 (s, 1H), 9,06 (s, 1H), 10,26 (s, 1H), 11,71 (d, J=6,0 Hz, 1H), 12,69 (s, 1H).

Example 26: EP Method - Preparation of N-(4-(4-((5-(cyanomethyl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridin-2- yl)-3-fluorophenyl)cyclohexanecarboxamide (I-353)

Schritt 1. (2-(4-(cyclohexanecarboxamido)-2-fluorophenyl)-6-(4-methoxybenzyl)-5-oxo-5,6-di-hydro-1,6-naphthyridin-4-yl)carbamate of von tert-butyl (26.1)

The reaction was carried out according to the representative procedure described in Method BP (Step 4) using N-(4-(4-amino-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro- 1,6-Naphthyridin-2-yl)-3-fluorophenyl)cyclohexanecarboxamide (23.3) and tert-butyl carbamate. (1.0 g, 86.57%) MS (ES): m/z 601.7 [M+H]+.

Schritt 2. N-(4-(4-Amino-6-(4-metoxibenzil)-5-oxo-5,6-dihidro-1,6-naftiridin-2-il)-3-fluorfenil)ciclohexancarboxamida (26.2)

For a solution of tert-butyl-(2-(4-(cyclohexanecarboxamido)-2-fluorophenyl)-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro-1,6-naphthyridin-4-yl ) Carbamate (26.1) (1.0 g, 0.00161 mol, 1.0 eq) in DCM (5.0 mL) At 0 °C methylsulfonic acid (2.0 mL) was added. After stirring at room temperature for 30 min, the reaction mixture was poured into saturated NaHCO 33solution (50 ml) and extracted with ethyl acetate (100 ml x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 26.2 (800 mg, 96%). MS (ES): m/z 501.5 [M+H]+

Schritt 3. N-(4-(4-((5-(cyanomethyl)pyridin-2-yl)amino)-6-(4-methoxybencyl)-5-oxo-5,6-di-hydro-1,6 -naphthyridine -2-yl)-3-fluorophenyl)cyclohexancarboxamida (26.3)

The reaction was carried out according to the representative procedure described in Method BP (Step 4) using N-(4-(4-chloro-6-(4-methoxybenzyl)-5-oxo-5,6-dihydro- 1,6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexanecarboxamide (26.2) and 2-(6-bromopyridin-3-yl)acetonitrile to give (26.3) (0.060 g, 37.46%) MS (ES): m/z 617.7 [M+H]+.

Schritt 4. N-(4-(4-((5-(cyanomethyl)pyridin-2-yl)amino)-5-oxo-5,6-di-hydro-1,6-nafthyridin-2-yl)- 3- Fluorphenyl)cyclohexancarboxamide (I-353)

The reaction was performed according to the representative procedure described in Method DP (Step 4) to give I-353 (30 mg, 62.10%), MS (ES): m/z 597.39 [M+H]+.1RMN-H (DMSO-d6, 400 MHz): 13,26 (s, 1H), 12,41 (s, 1H), 10,40 (s, 1H), 8,96 (s, 1H), 8,45 (s, 1H), 7,89-7,85 (m, 3H), 7,47 (s, 1H), 7,55 (d, J=8,0 Hz, 1H), 7,34 (s, 1H), 6,77 (d, J=72 Hz, 1H), 4,21 (s, 2H), 2,42-2,36 (m, 1H), 1,87–1,77 (m, 4H), 1,69– 1,67 (m, 1H), 1,48–1,39 (m, 2H), 1,34–1,25 (m, 4H).

Document 27: Supplier FP – Derivation of (R)-8-((6-(Dimethylamino)methyl)-5-(THF-3-yl)pyridin-2-yl)amino)-5-(7-fluorimidazo[ 1). ,2-a]pyridin-3-yl)isoquinoline-1(2H)-one (I-356) and (S)-8-((6-((dimethylamino)methyl)-5-(THF-3-) yl)pyridin-2-yl)amino)-5-(7-fluorimidazo[1,2-a]pyridin-3-yl)isoquinolin-1(2H)-one (I-357) .

Step 1. 5-Bromoisoquinoline (27.1)

For a solution of isoquinoline (27.0) (20.0 g, 0.1550 mol, 1.0 eq) in H2SO4(120.0 mL) At 0 °C, N-bromosuccinamide (30.35 g, 0.465 mol, 3.0 eq) was added. After stirring at room temperature for 1.5 h, the reaction mixture was poured into water (500 mL), neutralized with concentrated ammonium hydroxide, and extracted with DCM (100 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 27.1 (20.5 g, 99%), MS (ES): m/z 208.97 [M+H]+

Step 2. 5-bromo-8-chloroisoquinoline (27.2)

For a solution of 5-bromoisoquinoline (27.1) (20.5 g, 0.0985 mol, 1.0 eq) in H2SO4(5.0 mL) at 0°C, N-chlorosuccinamide (19.75 g, 1.5 eq) was added. After stirring at 80 °C for 2 h, the reaction mixture was poured into water (50 mL), neutralized with concentrated ammonium hydroxide, and extracted with ethyl acetate (100 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 27.2 (18.5, 96%). MS (ES): m/z 243.93 [M+H]+

Step 3. 5-bromo-8-chloroisoquinolin-1(2H)-one(27,3)

To a solution of 5-bromo-8-chloroisoquinoline (27.2) (18.0 g, 0.0743 mol, 1.0 eq.) in DCM (15.0 mL) at 0 °C was added m- CPBA (19.23g, 0.1114, 1.5 eq.). After stirring at room temperature for 3 h, the reaction mixture was poured into saturated NaHCO3.3solution (200 ml) and extracted with ethyl acetate (100 ml x 3). The combined organic layer was dried over sodium sulphate and concentrated under reduced pressure to give crude material (15g) which was dissolved in acetic anhydride (200ml). After stirring at 100 °C for 1 h, 2N NaOH (150 mL) was added. After stirring at 110°C for 1 hour, the reaction mixture was poured into water (200ml) and extracted with ethyl acetate (100ml x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by combi flash elution with 20-30% ethyl acetate in hexane to give 27.3 (7 g, 60.94%). MS(ES): m/z 258.9 [M+H]+.

Step 4. 5-Bromo-8-chloro-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one (27,4) .

DBU (20.4 g, 0.13-0.5 eq.) and 2-(trimethylsilyl)ethoxymethyl chloride (17.8 g, 0.010, 4 eq.) were added. After stirring at room temperature for 1.5 h, the reaction mixture was poured into water (100 mL) and extracted with ethyl acetate (50 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by combi flash elution with 60-70% ethyl acetate in hexane to give 27.4 (4.0 g, 96%). MS(ES): m/z 389 [M+H]+.

Schritt 5. 8-Cloro-2-((2-(trimetilsilil)etoxi)metil)-5-(trimetilestanil)isoquinolina-1(2H)-on (27,5)

To a solution of 5-bromo-8-chloro-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one (27.4) (4.0 g, 0.0102 mol, 1 0.0 eq) in toluene (5 mL) was added hexamethyldithine (5.0 g, 0.0153, 1.5 eq). After 10 minutes of degassing. Bis(triphenylphosphine)palladium chloride (0.715 g, 0.00102 mol, 0.1 eq) was added under argon and degassed again for 5 min. After stirring at 110 °C for 4 h, the reaction mixture was poured into water (30 mL) and extracted with ethyl acetate (40 mL x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by Combiflash eluting with 2% ethyl acetate in hexane to give 27.5 (3.8 g, yield: 68.34%). MS(ES): m/z 473.06 [M+H]+.

Step 6. 8-Chloro-5-(7-fluoroimidazol[1,2-a]pyridin-3-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinoline-1(2H)-one ( 27 ). ,6 )

To a solution of 8-chloro-2-((2-(trimethylsilyl)ethoxy)methyl)-5-(trimethylstanyl)isoquinolin-1(2H)-one (27.5) (3.5 g, 0.0074 mol , 1.0 eq.) in 3-bromo-7-fluoroimidazo[1,2-a]pyridine dioxane (1.9 g, 0.0088 mol, 1.2 eq) was added. After 15 min degassing. Tetrakis (0.85 g, 0.00074 mol, 0.1 eq) and CuI (0.14 g, 0.00074 mol, 0.1 eq) were added under argon atmosphere. After stirring at 110 °C for 2 h, the reaction mixture was poured into water (50 mL) and extracted with ethyl acetate (50 mL x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by Combiflash eluting with 25% ethyl acetate in hexane to give 27.6 (1.9 g, yield: 64%), MS (ES): m/z 444.13 [M+H]+ .

Schritt 7. 8-((6-((Dimethylamino)methyl)-5-(THF-3-yl)pyridin-2-yl)amino)-5-(7-fluorimidazole[1,2-a]pyridin-3 -yl)isoquinolin-1(2H)-one(27.7)

To a solution of 8-chloro-5-(7-fluoroimidazo[1,2-a]pyridin-3-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one ( 27.6) (1.9 g, 0.00428 mol, 1.0 eq) in dioxane (1.5 mL) was added 6-((dimethylamino)methyl)-5-(THF-3-yl)pyridin- . 2-amine (0.94 g, 0.00428 mol). 1.0 equiv.) and K2CO3(1.7 g, 0.0128 mol, 3.0 equiv.). After 15 min degassing. under argon atmosphere 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (Xanthphos) (0.24 g, 0.428 mmol, 0.1 eq) and tris(dibenzylideneacetone)dipalladium (Pd2(db)3) (0.39 g, 0.428 mmol, 0.1 eq). After stirring at 110 °C for 2 h, the reaction mixture was poured into water (50 mL) and extracted with ethyl acetate (50 mL x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by Combiflash eluting with 4% DCM in methanol to give 27.7 (900 mg, yield: 45%), MS(ES): m/z 499.2 [M+H]+.

Schritt 8. (R)-8-((6-((Dimethylamino)methyl)-5-(THF-3-yl)pyridin-2-yl)amino)-5-(7-fluorimidazo[1,2-a). ).]pyridin-3-yl)isoquinolin-1(2H)-one (I-356) and (S)-8-((6-((dimethylamino)methyl)-5-(THF-3-yl)pyridine -2 -yl)amino)-5-(7-fluorimidazo[1,2-a]pyridin-3-yl)isoquinolin-1(2H)-one (I-357) .

27.7 (100 mg racemic) was resolved by Chiral SFC on Shimadzu LC-20AP and UV detector. The column used was CHIRALPAK IC (250 x 21.0) mm, 5 microns, the column flow was 20 mL/min. The mobile phase used was (A) DEA 0.1% IN n-hexane (B) DEA 0.1% IN propan-2-ol:acetonitrile (70:30) to add I-356 (25 mg) and I- 357 (28mg). The stereochemistry was arbitrarily assigned. I-356: MS(ES): m/z=500.2 [M+H]+, 1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 11.61 (d, J = 6.0 Hz, 1H), 8.89 (d, J=8.7 Hz, 1H), 7.93 (dd, J=7.5, 5.8 Hz, 1H), 7.77-7 .63 (m, 3H), 7.55 (dd, J=10.2, 2.6 Hz, 1H), 7.15 (dd, J=7.3, 5.8 Hz, 1H), 7, 01-6, 88 (m, 2H), 5.89 (dd, J=7.3, 1.3 Hz, 1H), 4.05-3.93 (m, 2H), 3.81 (q , J=7, 7 Hz, 2H), 3.54 (dd, J=8.2, 6.8 Hz, 1H), 2.36-2.18 (m, 6H), 1.90 (dq, J =12.1, 7.7Hz, 1H). I-357: MS(ES): m/z=500.2 [M+H]+, 1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 11.63 (d, J = 5.8 Hz, 1H), 8.80 (s, 1H), 7.93 (dd, J=7.6, 5.8 Hz, 1H), 7.77-7.63 (m, 3H) , 7.56 (dd, J=10.1, 2.6 Hz, 1H), 7.23-7.11 (m, 1H), 7.02 (d, J=8.3 Hz, 1H), 6.93 (td, J=7.6, 2.6 Hz, 1H), 5.97-5.81 (m, 1H), 4.15-3.93 (m, 3H), 3.82 ( q, J=7.7 Hz, 2H), 3.73 (s, 1H), 3.55 (t, J=7.6 Hz, 1H), 3.49 (t, J=5.3 Hz, 1H), 3.42 (t, J=5.2 Hz, 1H), 2.31 (s, 2H), 1.96-1.86 (m, 1H), 1.25 (d, J=8 , 0Hz, 1H).

Example 28: GP Method - Preparation of 8-((5-(4-Methylpiperazin-1-yl)pyridin-2-yl)amino)-5-(pyridin-4-yl)isoquinolin-1(2H)-one ( I-358)

Step 1. 8-Chloro-5-(pyridin-4-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one (28.1) .

For a solution of 5-bromo-8-chloro-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one (27.5) (1.0 g, 2.57 mmol, first 0.0 eq.) and pyridine Added -4-ylboronic acid (0.75 g, 3.08 mmol, 1.2) in 1,4-dioxane (8 mL) and water (2 mL) with potassium carbonate (1.06 g, 7.771 mmol, 3.0 eq). After 15 min degassing. under argon atmosphere, DCM complex of [1,1'-bis(diphenylphosphino)ferrocene]palladium(II) dichloride (Pd(dppf)Cl2).dcm (0.205 g, 0.257 mmol, 0.1 equiv) was added. After stirring at 110°C for 1 hour, the reaction mixture was poured into water (200ml) and extracted with ethyl acetate (100ml x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by Combiflash eluting with 4% DCM in methanol to give 28.1 (700 mg, yield: 60%), MS(ES): m/z 387.12 [M+H]+.

Paso 2. 8-((5-(4-Methylpiperazin-1-yl)pyridin-2-yl)amino)-5-(pyridin-4-yl)-2-((2-(trimethylsilyl)ethoxy)methyl) Isoquinoline-1(2H)-ona (28.2)

For a solution of 8-chloro-5-(pyridin-4-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one (28.1) (0.7 g. 1.813 mmol, 1.0 eq) and 5-(4-methylpiperazin-1-yl)pyridin-2-amine (0.383 g, 1.99 mmol, 1.1 eq) in toluene (7 mL) was added to K2CO3(0.750 g, 5.43 mmol, 3.0 eq.). After 15 min degassing. under argon atmosphere 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (Xanthphos) (0.104 g, 0.181 mmol, 0.1 eq) and tris(dibenzylideneacetone)dipalladium (Pd2(db)3) (0.165 g, 0.181 mmol, 0.1 eq). After stirring at 110°C for 2 hours, the reaction mixture was poured into water (200ml) and extracted with ethyl acetate (100ml x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by Combiflash eluting with 4% DCM in methanol to give 28.2 (300 mg, yield: 30.28%), MS(ES): m/z 543.28 [M+H]+.

Schritt 3. 8-((5-(4-Methylpiperazin-1-yl)pyridin-2-yl)amino)-5-(pyridin-4-yl)isoquinolin-1(2H)-ona (I-358)

For a solution of 8-((5-(4-methylpiperazin-1-yl)pyridin-2-yl)amino)-5-(pyridin-4-yl)-2-((2-(trimethylsilyl)ethoxy)methyl )Isoquinolin-1(2H)-one (28.2) (300 mg, 0.777 mmol, 1.0 eq.) in DCM (2 mL) at 0 °C 4 M HCl in 1.4 dioxane (8 mL) was added ) admitted. After stirring at room temperature for 1 h, the reaction mixture was poured into water (50 mL), neutralized with sodium bicarbonate, and extracted with DCM (40 mL x 3). The combined organic layer was washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by trituration with diethyl ether/pentane to give I-358 (200 mg, 87% yield). MS(ES): m/z 413.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 12.51 (s, 1H), 8.70-8.54 (m, 3H), 8.00 (d, J=3.0 Hz, 1H), 7 .54 (d, J=8.7 Hz, 1H), 7.50-7.39 (m, 3H), 7.17 (d, J=7.4 Hz, 1H), 6.96 (d, J=8.9Hz, 1H), 6.47 (d, J=7.5Hz, 1H), 3.13 ( t, J = 5.0 Hz, 4H), 2.5 (s, 4H, fusion in the DMSO peak), 2.26 (s, 3H).

Example 29: HP Method - Preparation of 5-(1-Methyl-1H-imidazol-5-yl)-8-((5-(piperazin-1-yl)pyridin-2-yl)amino)isoquinolin-1(2H). ). ). )-um (I-366)

Paso 1. 8-Chloro-5-(1-methyl-1H-imidazol-5-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinoline-1(2H)-one (29.1)

The reaction was carried out according to the representative procedure described in GP Method (Step 1) using 5-bromo-8-chloro-2-((2-(trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)- one 27,5 and 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-imidazole for 29.1 (1.4 g, 51, 87% yield) to give MS(ES): m/z 391.2 [M+H]+

Schritt 2 y 3. 5-(1-Methyl-1H-imidazol-5-yl)-8-((5-(piperazin-1-yl)pyridin-2-yl)amino)isoquinoline-1(2H)-on (I-366)

The reaction of Steps 2 and 3 was performed according to the representative procedure described in GP Method (Steps 2 and 3) using 8-chloro-5-(1-methyl-1H-imidazol-5-yl)-2- (( 2 -(Trimethylsilyl)ethoxy)methyl)isoquinolin-1(2H)-one 29.1 and tert-butyl 4-(6-aminopyridin-3-yl)piperazine-1-carboxylate followed by a 2-(trimethylsilyl) group ethoxymethyl (SEM) deprotection for I-366 (0.8 g, 53.87% yield) MS(ES): m/z 402.2 [M+H]+RMN 1H (400 MHz, DMSO-d6) δ 12,52 (s, 1H), 11,50 (s, 1H), 8,60 (d, J=8,7 Hz, 1H), 8,27 (s , 1H), 8,01 (d, J= 3,0 Hz, 1H), 7,76 (s, 1H), 7,51–7,43 (m, 2H), 7,15 (d, J= 5,7 Hz, 1H), 6,93 (d, J=8,9 Hz, 1H), 6,91 (td, J=7,5, 2,7 Hz, 1H), 6,14 (d, J=7,2 Hz, 1H), 3,36 (s, 3H), 3,26 (s, 4H), 3,17 (s, 4H).

Example 30: Method IP - Preparation of 8-((6-((dimethylamino)methyl)-5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-5-(1-methyl-1H- Pyrrolo[2,3-b]pyridin-4-yl)-2,6-naphthyridin-1(2H)-one (I-361)

Step 1. 2-Bromo-5-chloro-3-methylisonicotinic acid (30.1)

To a solution of 2-bromo-5-chloro-3-methylpyridine (30.0) (2.5 g, 12.13 mmol) in THE (25 mL) at -78 °C was added 2 M lithium diisopropylamide ( LDA) in THE (2 M, 9.1 mL, 18.15 mmol) dropwise under nitrogen. After the LDA addition was complete, the reaction mixture was purged of CO2Gas at -78°C for 1 h. After warming to 0°C, the reaction mixture was diluted with ethyl acetate (25 mL) and then with water (25 mL). The aqueous layer was extracted with ethyl acetate (2 x 100 mL). The aqueous layer was neutralized with citric acid and then extracted with ethyl acetate (4 x 150ml). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 2-bromo-5-chloro-3-methylisonicotinic acid (30.1) (1.7 g, 56%) which was used without further purification. MS(ES): m/z=251.4 [M+H]+.

Step 2. Methyl 2-bromo-5-chloro-3-methylisonicotinate (30.2)

To a solution of (30.1) (3 g, 12 mmol, 1.0 equiv.) in dimethylformamide (30 mL) under a nitrogen atmosphere was added potassium carbonate (3.31 g, 24 mmol, 2.0 equiv. .) and methyl iodide (2.04 g, 14.5 mmol, 1.1 equiv. ). After stirring at room temperature for 3h, the reaction mixture was diluted with water (100ml) and extracted with ethyl acetate (3 x 150ml). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified on Combiflash silica eluting with 5-10% ethyl acetate in hexane to give 30.2 (1.5 g, 47% yield). MS(ES): m/z = 265.5 [M+H]+

Stage 3. 5-Bromo-8-chloro-2,6-naphthyridin-1(2H)-one(30,3)

1,3,5-Triazine (0.92 g, 11.36 mmol, 2.0 eq) was added dropwise. After warming to room temperature, potassium tertiary butoxide (1.2 g, 11.36 mmol, 2.0 eq.) was added. After stirring at 110°C for 1 hour, the reaction mixture was poured into water (100ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was triturated with ether to give 30.3 (1.0 g, 68% yield). MS(ES): m/z 260.0 [M+H]+.

Step 4. 8-Chloro-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-2,6-naphthyridin-1(2H)-one (9.4);

To a solution of 5-bromo-8-chloro-2,6-naphthyridin-1(2H)-one (30.3) (1.0 g, 3.84 mmol, 1.0 eq) and 1-methyl- 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridine (1.0 g, 4.2 mmol, 1, 1 eq) in dioxane (10 ml) and water (2 ml) and potassium phosphate (2.4 g, 11.5 mmol, 3.0 eq) were added. After degassing under argon atmosphere for 15 minutes, [1,1'-bis(diphenylphosphino)ferrocene]palladium(II) dichloride is obtained. DCM complex (Pd(dppf)Cl2).dcm (0.31 g, 0.384 mmol, 0.1 equiv) was added. After stirring at 90°C for 1 hour, the reaction mixture was poured into water (100ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by Combiflash eluting with 100% ethyl acetate to give 30.4 (0.8 g, 66.8% yield). MS(ES): m/z 311.0 [M+H]+.

Schritt 5. 8-Chloro-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)-2,6- Naphthyridine -1(2H)-bound (30.5)

To a solution of 8-chloro-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-2,6-naphthyridin-1(2H)-one (30.4) ( 0.8 g, 2.58 mmol, 1.0 eq) in THE (8 mL) DBU (0.16 g, 12.9 mmol, 5.0 eq). After stirring at room temperature for 20 min, 2-(trimethylsilyl)ethoxymethyl chloride (1.71 g, 10.32 mmol, 4.0 eq) was added at 0 °C. After stirring at room temperature for 3h, the reaction mixture was poured into water (100ml) and extracted with ethyl acetate (50ml x 3). The combined organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified using Combiflash eluting with 50% ethyl acetate in acetic acid to give 30.5 (1.0 g, 88% yield). MS(ES): m/z 442.0 [M+H]+.

Paso 6. 8-((5-(4-Hidroxipiperidin-1-yl)pyridin-2-yl)amino)-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl) -2-((2-(Trimethylsilyl)ethoxy)methyl)-2,6-naphthyridin-1(2H)-one (30,6)

To a solution of 8-chloro-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-2-((2-(trimethylsilyl)ethoxy)methyl)-2,6- The naphthyridin-1(2H)-one (30.5) (1.0 g, 2.26 mmol, 1.0 eq) in dioxane (10 mL) dio1-(6-aminopyridin-3-yl)piperidin- 4-ol (0.48 g, 2.48 mmol , 1.1 eq) and cesium carbonate (2.2 g, 6.78 mmol, 3.0 eq). After 15 min of degassing. in argon atmosphere, 4,5-bis(diphenylphosphine)-9,9-dimethylxanthene (Xanthphos) (0.150 g, 0.26 mmol, 0.1 eq) and chlorine(2-dicyclohexylphosphine-2′,4′, 6). '-triisopropyl- 1,1'-biphenyl)[2-(2'-amino-1,1'-biphenyl)]palladium(II) (xphos PdG2) (0.204 g, 0.26 mmol, 0.1 eq). After stirring at 110 °C for 16 h, the reaction mixture was poured into water (100 mL) and extracted into DCM (50 mL x 3). The combined organic layer was washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified using Combiflash eluting with 15% methanol in DCM to give 30.6 (0.8 g, 59% yield). MS(ES): m/z 598.0 [M+H]+.

Schritt 7. 8-((5-(4-Hidroxipiperidin-1-yl)pyridin-2-yl)amino)-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl) -2,6-naphthyridine-1(2H)-on (I-361)

For a solution of 8-((5-(4-Hydroxypiperidin-1-yl)pyridin-2-yl)amino)-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl )-2-((2-(trimethylsilyl)ethoxy)methyl)-2,6-naphthyridin-1(2H)-one (30.6) (0.8 g, 1.0 eq) in DCM (10 mL) at 0 °C, 4 M HCl in dioxane (10 mL) was added portionwise. After stirring at room temperature for 2 h, the reaction mixture was neutralized with aqueous NaHCO.3and extracted with 10% methanol in DCM (50 mL x 3). The combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified using Combiflash eluting with 15% methanol in DCM to give I-361 (0.43 g, 69% yield). MS(ES): m/z 468.0 [M+H]+. RMN 1H (400 MHz, DMSO-d6) δ 11,41 (s, 1H), 9,33 (s, 1H), 8,32–8,07 (m, 2H), 8,04 (d, J= 3,1 Hz, 1H), 7,97–7,77 (m, 2H), 7,51-7,37 (m, 2H), 7,32 (t, J=6,4 Hz, 1H), 7,18 (d, J=7,4 Hz, 1H), 6,92 (d, J=4,9 Hz, 1H), 6,07 (d, J=3,5 Hz, 1H), 4, 70 (s, 1H), 3,84 (s, 3H), 3,71–3,58 (m, 1H), 3,49 (dd, J=11,2, 6,2 Hz, 2H), 2 ,93 -2,76 (m, 2H), 1,97-1,75 (m, 2H), 1,51 (q, J=11,1, 9,7 Hz, 2H).

Example 31: Method JP - Preparation of 8-((6-((dimethylamino)methyl)-5-morpholinopyridin-2-yl)amino)-5-(1-methyl-1H-pyrrolo[2,3-b]pyridine -4-yl)-2,6-naphthyridin-1(2H)-one (I-369)

The reaction was carried out according to the representative procedure described in Method IP (Step 6) using 8-chloro-5-(1-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)- 2,6-Naphthyridin-1(2H)-one 30.4 and 6-((dimethylamino)methyl)-5-morpholinopyridin-2-amine to give I-369 (0.8 g, 53.87% yield) MS(ES)): m/z 511.2. 0 [M+H]+. RMN 1H (400 MHz, DMSO-d6) δ 9,40 (s, 1H), 8,24 (d, J=4,9 Hz, 1H), 7,91–7,74 (m, 2H), 7 ,45 (dd, J=8,2, 4,9 Hz, 2H), 6,97 (dd, J=16,2, 6,1 Hz, 2H), 4,51 (s, 1H), 3, 86 (s, 2H), 3,81-3,74 (m, 2H), 2,95 (s, 2H), 2,85 (t, J = 4,5 Hz, 2H).

Example 32: The compounds of the invention

Compounds of the invention were prepared according to the methods described above, using known in the literature/commercially available boronates and anilines or with the listed intermediates. If the reagents are written, they are known in the literature/commercially available.

MESA 2 Method UE-# STRUCTURE NAME LCMS HRMN SM I-1 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[(5-methoxy-2-pyridyl)amino]-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 504 [M+H]+, Ret. time = 3.39 min. 1RMN-H (400 MHz, DMSO): δ 12,55 (s, 1H), 11,72 (s, 1 H), 8,78 (s, 1 H), 8,12 (d, J = 3, 0 Hz, 1 H), 7,57 (d, J = 6,1 Hz, 1 H), 7,49-7,42 (m, 2 H), 7,25 (s, 1 H), 7, 12 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 3,85 (s, 3 H), 3,82 (s, 3 H ), 3,65–3,55 (m, 2 H), 3,18–3,14 (m, 2 H), 1,62–1,54 (m, 4 H), 1,47 - 1, 41 (metro, 2H). Ein 5-methoxypyridin-2-amine CB1 I-2 N-Ethyl-2-[6-[[2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-5-oxo-6H-1,6-naphthiridin-4-yl] Amino]-3-pyridyl]-2-methyl-propanamide BicarbB EHC18 method, m/z = 587 [M + H]+, Rt. Time = 4.08 min. 1RMN-H (400 MHz, DMSO): δ 12,75 (s, 1 H), 11,78 (s, 1 H), 9,09 (s, 1 H), 8,32 (d, J = 1 ,8 Hz, 1 H), 7,67 (dd , J = 2,3, 8,6 Hz, 1 H), 7,59 (d, J = 6,1 Hz, 1 H), 7,45 ( d, J = 6,8 Hz, 2 H), 7,26 (d, J = 8,3 Hz, 1 H), 7,06 (d, J = 8,3 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 3,86 (s, 3 H), 3,62-3,55 (m, 2 H), 3,20-3,15 (m , 2 H ), 3,10-3,00 (m, 4 H), 1,60 (dd, J = 4,8, 20,7 Hz, 4 H), 1,47 (s, 6 H), 0,95 (dd, J = 7,2, 7,2 Hz, 3 H). A CA1 CB1 I-3 4-[(5-cyclopropyl-4-fluoro-2-pyridyl)amino]-2-[2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl]-6H-1,6-naphthyridine- 5-eins BicarbB EHC18 method, m/z = 532 [M + H]+, Ret. Time = 5.09 min. 1RMN-H (400 MHz, DMSO): δ 12,78 (s, 1 H), 11,79-11,77 (m, 1 H), 8,98 (d, J = 1,4 Hz, 1 H ), 8,16 (d, J = 6,0 Hz, 1 H), 7,57 (d, J = 6,0 Hz, 1 H), 7,48 (d, J = 7,3 Hz, 1 H), 7,27 (d, J = 7,3 Hz, 1 H), 7,05 (d, J = 7,3 Hz, 1 H), 6,66 (d, J = 7,3 Hz, 1 H), 3,87 (s, 3 H) 3,67–3,56 (m, 2 H), 3,17 (dd, J = 5,4, 5,4 Hz, 2 H), 1, 98-1,90 (m, 1 H), 1,65-1,45 (m, 6 H), 0,99-0,93 (m, 2 H), 0,86-0,81 (m, 2H). Ein 5-cyclopropyl-4-fluoro-pyridine-2-amine CB1 I-4 4-[[2-(2,6-Difluoro-phenyl)-5-oxo-6H-1,6-naphthyridin-4-yl]amino]-N-ethyl-benzamide 10cm_Bicarb_AQ method, m/z = 421 [M + H]+, Ret-Zeit = 2.87 min. 111,92 (s, 1H), 11,79 (s, 1H), 8,47 (dd, J = 5,4, 5,4 Hz, 1H), 7,94 (d, J = 8,6 Hz) , 2H), 7,63 - 7,54 (m, 1H), 7,52-7,46 (m, 3H), 7,26 (dd, J = 8,1, 8,1 Hz, 2H), 7,11 (s, 1H), 6,60 (d, J = 7,1 Hz, 1H), 3,32 (q, J = 7,2 Hz, 2H), 1,16 (t, J = 7 ,2Hz, 3H). B 4-Amino-N-etilbenzamida-2-bromo-1,3-difluorobenceno I-5 8-bromo-2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 528 [M + H]+, Ret. Time = 3.02 min. 1RMN-H (400 MHz, DMSO) δ 12,65 (s, 1H), 12,13-12,09 (m, 1H), 8,60 (s, 1H), 8,11 (d, J = 2 ,5 Hz, 1H), 7,19 (s, 1H), 7,69-7,06 (m, 1H), 7,52 (dd, J = 2,9, 9,0 Hz, 1H), 7 ,32 (dd, J = 8,0, 8,0 Hz, 2H), 7,07 (d, J = 8,8 Hz, 1H), 4,73 (d, J = 4,3 Hz, 1H) , 3,70–3,65 (m, 1H), 3,56 (d, J = 12,6 Hz, 2H), 2,95–2,86 (m, 2H), 1,90–1,84 (m, 2H), 1,58-1,46 (m, 2H). H 1-(6-Aminopyridin-3-yl)piperidin-4-ol 2-Bromo-1,3-difluorobenzene I-6 2-(2,6-Difluoro-phenyl)-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 435 [M + H]+, Ret. Time = 2.67 min. 1RMN-H (400 MHz, DMSO) δ 12,59 (s, 1H), 8,50 (s, 1H), 8,07 (d, J = 2,8 Hz, 1H), 7,67–7, 57 (m, 1H), 7,52–7,46 (m, 2H), 7,30 (dd, J = 8,0, 8,0 Hz, 2H), 7,06 (d, J = 8, 8 Hz, 1H), 6,60 (d, J = 7,3 Hz, 1H), 3,08 (dd, J = 4,7, 4,7 Hz, 4H), 2,87 (dd, J = 4,8, 4,8Hz, 4H). B und G tert-butyl-4-(6-amino-pyridin-3-yl)piperazine-1-carboxylate-2-bromo-1,3-difluorobenzene I-7 4-[[3-(2,6-Difluoro-phenyl)-8-oxo-7H-2,7-naphthyridin-1-yl]amino]-N-ethyl-benzamide Method 10cm_Bicarb_AQ, m/z = 421 [M + H]+, Ret. Time = 3.14 min. 112,25 (s, 1H), 11,99 (s, 1H), 8,30 (dd, J=5,5, 5,5Hz, 1H), 7,89 (d, J=8,9Hz, 2H ), 7,82 (d, J=8,8 Hz, 2H ), 7,64–7,56 (m, 1H), 7,51 (d, J=6,9 Hz, 1H), 7,31 ( dd, J=8,2, 8,2 Hz, 2H), 7,19 (s, 1H), 6,63 (d, J=7,0Hz, 1H), 3,32–3, 23 (m, 2H), 1,12 (t, J=7,2Hz, 3H). Q 4-Amino-N-ethyl-benzamida-2-bromo-1,3-difluoro-benzol I-8 6-(2,6-difluorophenyl)-8-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2H-2,7-naphthyridin-1-one Method 10cm_Formic_AQ, m/z = 450 [M + H]+, Ret. Time = 2.65 min. 1RMN-H (400 MHz, DMSO) δ 12,10 (s, 1H), 11,83–11,83 (m, 1H), 8,28 (d, J = 9,0 Hz, 1H) 7,94 (d, J = 2,9 Hz, 1H), 7,55– 7,46 (m, 1H), 7,40 (d, J = 7,0 Hz, 1H), 7,30 (dd, J = 3,0, 9,2 Hz, 1H), 7,21 (dd, J = 8,2, 8,2 Hz, 2H), 7,02 (s, 1H), 6,50 (d, J = 7 ,0 Hz, 1H), 4,60 (d, J = 3,6 Hz, 1H), 3,56-3,51 (m, 1H), 3,41 (dd, J = 3,9, 8, 3 Hz, 2H), 2,78–2,70 (m, 2H), 1,78–1,70 (m, 2H), 1,46–1,35 (m, 2H). Q 1-(6-Amino-piridin-3-il)piperidin-4-ol-2-brom-1,3-difluorbenzol I-9 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-isopropyl-6H-1,6-naphthiridin-5-one Method 10cm_Formic_AQ, m/z = 380 [M + H]+, Ret. Time = 2.4 min. 1RMN H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,50 (d, H = 3,8 Hz, 1H), 8,19 (s, 1H), 8,10 (d, J = 3,0 Hz, 1H), 7,47 (dd , J = 2,9, 9,0 Hz, 1H), 7,34 (dd, J = 6,3, 6,3 Hz, 1H), 6 ,99 (d, J = 8,8 Hz, 1H), 6,49 (d, J = 7,1 Hz, 1H), 4,71 (d, J = 4,0 Hz, 1H), 3,69 –3,62 (m, 1H), 3,57–3,48 (m, 2H), 2,97–2,83 (m, 3H), 1,85 (dd, J = 3,3, 12, 9Hz, 2H). ), 1,57–1,46 (m, 2H), 1,26 (d, J = 6,8 Hz, 6H). B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 2-propylzinc bromide I 10 N-ethyl-4-[(2-isopropyl-5-oxo-6H-pyrido[4,3-d]pyrimidin-4-yl)amino]benzamide Method 10cm_Bicarb_AQ, m/z = 352 [M + H]+, Ret. Time = 2.87 min. 1H-NMR (400 MHz, DMSO) δ 12.13 (s, 1H), 12.00 (s, 1H), 8.41 (dd, J = 5.4, 5.4 Hz, 1H), 7, 98 (d, J=8.9Hz, 2H) 7.91 (d, J=8.8Hz, 2H), 7.62 (d, J=7.2Hz, 1H), 6.48 (d , J = 7.2Hz, 1H), 3.32-3.26 (m, 2H), 3.09-2.99 (m, 1H), 1.33 (d, J = 6.9Hz, 6H), 1.14 (dd, J=7.2, 7.2Hz, 3H). T4-Amino-N-ethyl-benzamide I-11 7-(2,6-difluorophenyl)-5-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-3H-pyrido[2,3-d]pyrimidin-4-one Method 10cm_Formic_AQ, m/z = 451 [M + H]+, Ret. Time = 2.49 min. 1RMN-H (400 MHz, DMSO) δ 11,87–11,82 (m, 1H), 8,38 (s, 1H), 8,22 (s, 1H), 7,98 (d, J = 7 ,8 Hz, 1H), 7,55–7,46 (m, 1H), 7,39 (dd, J = 2,9, 9,0 Hz, 1H), 7,19 (dd, J = 8, 0, 8,0 Hz, 2H), 6,96 (d, J = 8,8 Hz, 1H), 4,60-4,60 (m, 1H), 3,59–3,51 (m, 1H ), 3,48–3,39 (m, 3H), 2,83–2,74 (m, 2H), 1,75–1,71 (m, 2H), 1,45–1,33 (m , 2H). R 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-2,6-difluorophenyl-tributylzine I-12 4-[[7-(2,6-Difluoro-phenyl)-4-oxo-3H-pyrido[4,3-d]pyrimidin-5-yl]amino]-N-ethyl-benzamide Method 10cm_Bicarb_AQ, m/z = 422 [M + H]+, Ret. Time = 2.9 min. 111,67 (s, 1H), 8,25-8,20 (m, 2H), 7,81 (d, J=8,3Hz, 2H), 7,74 (d, J=8,3Hz, 2H ), 7,57 - 7,48 (m, 1H), 7,22 (dd, J = 8,1, 8,1 Hz, 2H), 7,02 (s, 1H), 3,19 ( q, J = 6,7 Hz, 2H), 1,04 (dd, J = 6,8, 6,8 Hertz, 3H). Não foi observada ressonância NH. S 4- Amino-N-ethyl-benzamide 2,6- Difluorofenil-tributylzina I-13 7-(2,6-difluorophenyl)-5-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-3H-pyrido[4,3-d]pyrimidin-4-one Method 10cm_Formic_AQ, m/z = 451 [M + H]+, Ret. Time = 2.56 min. 1RMN H (400 MHz, DMSO) δ 12,80–12,79 (m, 1H), 11,41 (s, 1H), 8,29–8,22 (m, 2H), 7,97–7, 95 (m, 1H), 7,57–7,48 (m, 1H). ), 7,32 (dd, J = 2,4, 9,2 Hz, 1H), 7,22 (dd, J = 8,2, 8,2 Hz, 2H), 6,98 (s, 1H) , 4,60 (d, J = 4,0 Hz, 1H), 3,58 -3,50 (m, 1H), 3,45-3,37 (m, 2H), 2,74 (dd, J = 10,0, 10,0 Hz, 2H), 1,78-1,72 (m, 2H), 1,46-1,35 (m, 2H). S 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 2,6-difluorophenyl-tributylzina I-14 4-[[7-(2,6-Difluoro-phenyl)-4-oxo-3H-pyrido[2,3-d]pyrimidin-5-yl]amino]-N-ethyl-benzamide Method 10cm_Formic_AQ, m/z = 422 [M + H]+, Ret. Time = 2.55 min. 1RMN-H (400 MHz, DMSO) δ 12,65 (s, 1H), 11,11 (s, 1H), 8,39–8,33 (m, 1H), 8,23–8,20 (m , 1H), 7,83 (d, J = 8,6 Hz, 2H), 7,53–7,43 (m, 1H), 7,38 (dJ = 8,3 Hz, 2H), 7, 15 (dd, J = 8,1, 8,1 Hz, 2H), 7,03 (s, 1H), 3,20 (q, J = 6,7 Hz, 2H), 1,04 (dd, J = 7,2, 7,2 Hz, 3H). R 4-Amino-N-ethyl-benzamide-2,6-difluorophenyl-tributylzine I-15 2-(2,6-Difluorophenyl)-8-(1-hydroxyethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-Naphthyridine-5 -uno Method 10cm_Formic_AQ, m/z = 494 [M + H]+, Ret. Time = 2.66 min. 1RMN H (400 MHz, MeOD) δ 8,44 (s, 1H), 8,09 (d, J=2,9 Hz, 1H), 7,56-7,48 (m, 2H), 7, 38 (s, 1H), 7,14 (dd, J =8,2, 8,2Hz, 2H), 7,07 (d, J=8,8Hz, 1H), 5,23 (q, J = 6, 3 Hz, 1H), 3,82-3,74 (m, 1H), 3,62-3,54 (m, 2H), 2,98-2,90 (m, 2H), 2, 03-1 ,97 (m, 2H), 1,73-1,63 (m, 2H), 1,56 (d, J=6,4 Hz, 3H). 3 protones trocáveis ​​não observados H 1-(6-Aminopyridin-3-yl)piperidin-4-ol 2-Bromo-1,3-difluorobenzene I-16 2-(2,6-difluorophenyl)-8-(1-hydroxy-1-methylethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H - 1,6 -naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 508 [M + H]+, Ret. Time = 2.71 min. 1RMN-H (400MHz, MeOD) δ 12.51 (s, 1H), 8.48 (td, J=1.3, 11.9Hz, 2H), 8.11 (d, J=2.9 Hz, 1H), 7.57-7.49 (m, 2H), 7.36 (s, 1H), 7.16 (dd, J = 8.3, 8.3 Hz, 2H), 7.08 (dd, J=1.6, 8.8Hz, 1H), 3.83-3.75 (m, 1H), 3.63-3.56 (m, 2H), 2.99-2.91 (m, 2H), 2.03-1.98 (m, 2H), 1.74-1.67 (m, 2H), 1.65 (s, 6H). 2 Exchangeable protons not observed H 1-(6-Aminopyridin-3-yl)piperidin-4-ol 2-Bromo-1,3-difluorobenzene I-17 2-Cyclopropyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on Method 10cm_Bicarb_AQ, m/z = 378 [M + H]+, Ret. Time = 2.8 min. 1RMN-H (400 MHz, DMSO) δ 12,78 (s, 1H), 11,63 (s, 1H), 8,55 (s, 1H), 8,09 (d, J = 2,8 Hz, 1H), 7,67–7,58 (m, 1H), 7,51 (dd, J = 2,9, 9,0 Hz, 1H), 7,37 (s, 1H), 7,31 (dd , J = 8,0, 8,0 Hz, 2H), 7,04 (d, J = 8,8 Hz, 1H), 4,73 ( s, 1H), 3,70–3,65 (m, 1H), 3,54 (d, J = 12,9 Hz, 2H), 2,94–2,85 (m, 2H), 2,21 (s, 3H), 1,90–1,84 (m , 2H), 1,58-1,47 (m, 2H). B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol Cyclopropylcinbromet I-18 2-(2,6-Difluoro-phenyl)-4-[(5-methyl-sulfonyl-2-pyridyl)amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 429 [M + H]+, Ret. Time = 2.99 min. 1RMN-H (400 MHz, DMSO) δ 13,29 (s, 1H), 11,95–11,89 (m, 1H), 8,78 (s, 1H), 8,75 (d, J = 2 ,5 Hz, 1H), 8,12 (dd, J = 2,6, 8,7 Hz, 1H), 7,58–7,49 (m, 1H), 7,45 (d, J = 7, 3 Hz, 1H), 7,24–7,19 (m, 3H), 6,60 (d, J = 7,3 Hz, 1H), 3,20 (s, 3H). B 5-(Methylsulfonyl)pyridin-2-amine 2-Bromo-1,3-difluorobenzol I-19 2-cyclohexyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 420 [M + H]+, Ret. Time = 3.02 min. 1RMN H (400 MHz, DMSO) δ 12,31 (s, 1H), 11,52–11,45 (m, 1H), 8,16 (s, 1H), 8,10 (d, J = 3, 0 Hz, 1H), 7,46 (dd, J = 3,1, 9,0 Hz, 1H), 7,33 (d, J = 7,3 Hz, 1H), 6,98 (d, J = 8,9 Hz, 1H), 6,48 (d, J = 7,3 Hz, 1H), 4,71 (d, J = 4,1 Hz, 1H), 3,68–3,61 (m, 1H), 3,56–3,49 (m, 2H), 2,91–2,83 (m, 2H), 2,64–2,55 (m, 1H), 1,87–1,68 ( m, 6H), 1,58-1,24 (m, 6H). B 1-(6-amino-pyridin-3-yl)piperidin-4-ol Cyclohexylzincobromate I-20 2-Cyclopentyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on Method 10cm_Bicarb_AQ, m/z = 406 [M + H]+, Ret. Time = 2.92 min. 1RMN-H (400 MHz, DMSO) δ 12,30 (s, 1H), 11,49 (s, 1H), 8,20 (s, 1H), 8,08 (d, J = 3,0 Hz, 1H), 7,46 (dd, J = 3,1, 9,0 Hz, 1H), 7,33 (d, J = 7,3 Hz, 1H), 6,98 (d, J = 8,9 Hz, 1H), 6,47 (d, J = 7,3 Hz, 1H), 4,70 (d, J = 4,1 Hz). , 1H), 3,68–3,61 (m, 1H), 3,56–3,48 (m, 2H), 3,14–3,04 (m, 1H), 2,91–2,83 (m, 2H), 2,02–1,94 (m, 2H), 1,89 -1,75 (m, 6H), 1,75-1,64 (m, 2H), 1,57-1 ,46 (metro, 2H). B 1-(6-amino-pyridin-3-yl)piperidin-4-ol Cyclopentylzincobromet I-21 2-isopropyl-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-5-one Method 10cm_Formic_AQ, m/z = 365 [M + H]+, Ret. Time = 2.1 min. 1RMN-H (400 MHz, DMSO) δ 12,33 (s, 1H), 11,51–11,50 (m, 1H), 8,20 (s, 1H), 8,07 (d, J = 2 ,9 Hz, 1H), 7,45 (dd, J = 3,1, 8,9 Hz, 1H), 7,34 (d, J = 7,3 Hz, 1H), 7,00 (d, J = 8,9 Hz, 1H), 6,49 (d, J = 7,3 Hz, 1H), 3,08–3,04 (m, 4H), 3,00–2,98 (m, 1H) , 2,88–2,84 (m, 4H), 1,26 (d, J = 6,9 Hz, 6H). B und G tert-butyl-4-(6-amino-pyridin-3-yl)-piperazin-1-carboxy-lat-2-propylzincobromate I-22 2-isopropyl-4-[(5-morpholino-2-pyridyl)amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 366 [M + H]+, Ret. Time = 2.96 min. 1RMN-H (400 MHz, DMSO) δ 12,36 (s, 1H), 11,51 (s, 1H), 8,23 (s, 1H), 8,11 (d, J = 3,0 Hz, 1H), 7,48 (dd, J = 3,1, 9,0 Hz, 1H), 7,34 (d, J = 7,3 Hz, 1H), 7,02 (d, J = 8,8 Hz, 1H), 6,50 (d, J = 7,3 Hz, 1H), 3,79-3,75 (m, 4H), 3,16–3,12 (m, 4H), 3,00 –2,89 (m, 1H), 1,26 (d, J = 6,9 Hz, 6H). B 5- morpholino- pyridine- 2- amine 2- bromuro de propylzinc I-23 2-isopropyl-4-[(5-methyl-sulfonyl-2-pyridyl)amino]-6H-1,6-naphthiridin-5-one Method 10cm_Bicarb_AQ, m/z = 359 [M + H]+, Ret. Time = 2.9 min. 1RMN-H (400 MHz, DMSO) δ 13,21 (s, 1H), 11,81–11,74 (m, 1H), 8,88 (d, J = 2,5 Hz, 1H), 8, 65 (s, 1H), 8,18 (dd, J = 2,5, 8,8 Hz, 1H), 7,43 (d, J = 7,3 Hz, 1H), 7,23 (d, J = 8,5 Hz, 1H), 6,61 (d, J = 7,3 Hz, 1H), 3,30 (s, 3H), 3,11-3,00 (m, 1H), 1,30 (d, J = 6,9 Hz, 6H). B 5-(Methylsulfonyl)pyridine-2-amino-2-propylzincobromo I-24 2-(2,6-Difluoro-phenyl)-4-[[5-(1-piperidyl-sulfonyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 498 [M + H]+, Ret. Time = 3.36 min. 1RMN-H (400 MHz, DMSO) δ 13,29 (s, 1H), 11,93 (s, 1H), 8,77 (s, 1H), 8,58 (d, J = 2,4 Hz, 1H), 7,94 (dd, J = 2,5, 8,8 Hz, 1H), 7,58-7,49 (m, 1H), 7,45 (d, J = 7,2 Hz, 1H) ), 7,24-7,17 (m, 3H), 6,59 (d, J = 7,3 Hz, 1H), 2,85 (dd, J = 5,3, 5,3 Hz, 4H) , 1,50–1,43 (m, 4H), 1,30 (d, J = 4,0 Hz, 2H). B 5-(Piperidin-1-ylsulfonyl)pyridin-2-amin-2-bromo-1,3-difluorobenzol I-25 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 464 [M + H]+, Ret. Time = 2.63 min. 1RMN-H (400 MHz, DMSO) δ 12,78 (s, 1H), 11,63 (s, 1H), 8,55 (s, 1H), 8,09 (d, J = 2,8 Hz, 1H), 7,67–7,58 (m, 1H), 7,51 (dd, J = 2,9, 9,0 Hz, 1H), 7,37 (s, 1H), 7,31 (dd , J = 8,0, 8,0 Hz, 2H), 7,04 (d, J = 8,8 Hz, 1H), 4,73 ( s, 1H), 3,70–3,65 (m, 1H), 3,54 (d, J = 12,9 Hz, 2H), 2,94–2,85 (m, 2H), 2,21 (s, 3H), 1,90–1,84 (m , 2H), 1,58-1,47 (m, 2H). J 1-(6-Aminopyridin-3-yl)piperidin-4-ol 2-Bromo-1,3-difluorobenzene I-26 2-Cyclobutyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on Method 10cm_Formic_AQ, m/z = 392 [M + H]+, Ret. Time = 2.43 min. 1RMN H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,51 (s, 1H), 8,14 (s, 1H), 8,08 (d, J = 3,0 Hz, 1H), 7,46 (dd, J = 3,1, 9,0 Hz, 1H), 7,34 (d, J = 7,3 Hz, 1H), 6,98 (d, J = 8,9 Hz, 1H) 6,51 (d, J = 7,3 Hz, 1H), 4,71 (d, J = 4,0 Hz, 1H), 3,68-3,48 (m, 4H), 2 ,91-2,83 (m, 2H), 2,36-2,25 (m, 4H), 2,10-1,98 (m, 1H), 1,89-1,81 (m, 3H) , 1,57-1,46 (M, 2H). B 1-(6-amino-pyridin-3-yl)piperidin-4-ol Cyclo-butylzincobromate I-27 N-ethyl-6-[(2-isopropyl-5-oxo-6H-1,6-naphthiridin-4-yl)amino]pyridine-3-carboxamide Method 10cm_Bicarb_AQ, m/z = 352.246 [M + H]+, Ret. Time = 2.83 min. 1RMN-H (400 MHz, DMSO) δ 12,93 (s, 1H), 11,70 (d, J = 5,3 Hz, 1H), 8,89 (d, J = 2,1 Hz, 1H) , 8,63 (s, 1H), 8,51 (dd , J = 5,5, 5,5 Hz, 1H), 8,19–8,5 (m, 1H), 7,42–7,38 (m, 1H), 7,11 (d, J = 8,3 Hz, 1H), 6,57 (d, J = 7,0 Hz, 1H), 3,09–2,97 (m, 1H) , 1,30 (d, J = 6,8 Hz, 6H), 1,15 (t, J = 7,3 Hz, 3H). B 6- Amino-N-ethyl-nicotinamide 2- Propylzincbromuro I-28 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-isobutyl-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 394 [M + H]+, Ret. Time = 2.48 min. 1RMN-H (400 MHz, DMSO) δ 12,30 (s, 1H), 11,53 (d, J = 4,6 Hz, 1H), 8,11–8,07 (m, 2H), 7, 46 (dd, J = 3,1, 9,0 Hz, 1H), 7,36-7,31 (m, 1H), 6,98 (d, J = 8,9 Hz, 1H), 6,48 (d, J = 7,3 Hz, 1H), 4,70 (s, 1H), 3,69-3,62 (m, 1H), 3,57–3,48 (m, 2H), 2, 91–2,83 (m, 2H), 2,55 (d, J = 7,2 Hz, 2H), 2,16–2,07 (m, 1H), 1,88–1,81 (m, 2H), 1,56– 1,46 (m, B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-t-butylzincobromate 2H), 0.92 (d, J=6.6Hz, 6H). I-29 N-ethyl-4-[(2-isopropyl-5-oxo-6H-1,6-naphthiridin-4-yl)amino]benzamide Method 10cm_Bicarb_AQ, m/z = 351 [M + H]+, Ret. Time = 2.91 min. 1RMN-H (400 MHz, DMSO) δ 11,65 (s, 1H), 11,45–11,43 (m, 1H), 8,37 (dd, J = 5,5, 5,5 Hz, 1H ), 7,84 (d, J = 8,7 Hz, 2H) , 7,35 (d, J = 8,7 Hz, 2H), 7,27 (dd, J = 4,5, 7,0 Hz , 1H), 6,85 (s, 1H), 6,41 (d, J = 7,3 Hz, 1H), 3,24–3,18 (m, 2H), 2,88–2,80 ( m, 1H), 1,13 (d, J = 6,9 Hz, 6H), 1,06 (t, J = 7,2 Hz, 3H). B 4- Amino-N-ethyl-benzamide 2- Propylzincobrometo I-30 2-(2,6-difluoro-phenyl)-4-[[5-(methyl-sulfonyl-methyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ Method, m/z = 443 [M + H]+, Ret. Time = 2.5 min. 1RMN-H (400 MHz, DMSO) δ 12,98 (s, 1H), 11,92-11,85 (m, 1H), 8,77 (s, 1H), 8,35 (d, J = 2 ,1 Hz, 1H), 7,81 (dd, J = 2,4, 8,4 Hz, 1H), 7,64 7,56 (m, 1H), 7,49 (d, J = 7,3 Hz, 1H), 7,28 (dd, J = 8,0, 8,0 Hz, 2H), 7,16 (d, J = 8,4 Hz). , 1H), 6,63 (d, J = 7,3 Hz, 1H), 4,50 (s, 2H), 2,95 (s, 3H). B 5-((Methylsulfonyl)methyl)pyridin-2-amino-2-bromo-1,3-difluorobenzol I-31 2-Isopropyl-4-[[5-(1-piperidyl-sulfonyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 428 [M + H]+, Ret. Time = 2.79 min. 1RMN-H (400 MHz, DMSO) δ 13,19 (s, 1H), 11,80–11,73 (m, 1H), 8,71 (d, J = 2,4 Hz, 1H), 8, 64 (s, 1H), 8,00 (dd, J = 2,5, 8,8 Hz, 1H), 7,43 (d, J = 7,3 Hz, 1H), 7,21 (d, J = 8,8 Hz, 1H), 6,61 (d, J = 7,3 Hz, 1H), 3,11–3,00 (m, 1H), 2,96 (dd, J = 5,3, 5,3 Hz, 4H), 1,62–1,53 (m, 4H), 1,45–1,36 (m, 2H), 1,30 (d, J = 6,9 Hz, 6H). B 5-(Piperidin-1-ylsulfonyl)pyridin-2-amino-2-propylzincobromet I-32 2-(1-ethylpropyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one Method 10cm_Formic_AQ, m/z = 408 [M + H]+, Ret. Time = 2.52 min. 1RMN H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,50 (d, J = 4,8 Hz, 1H), 8,13 (s, 1H), 8,09 (d, J = 3,1 Hz, 1H), 7,47 (dd , J = 3,1, 9,0 Hz, 1H), 7,35–7,30 (m, 1H), 6,97 (d, J = 8,9 Hz, 1H), 6,50 (d, J = 7,2 Hz, 1H), 4,71 (s, 1H), 3,69-3,61 (m, 1H), 3,57- 3,48 (m, 2H), 2,91-2,83 (m, 2H), 2,49-2,40 (m, 1H), 1,88-1,80 (m, 2H), 1, 73-1,48 (m, 6H), 0,77 (dd, J = 7,4, 7,4 Hz, 6H). B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-pentan-3-ylzincobromate I-33 2-(2,6-Difluor-fenil)-4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-8-carbonitrile Method 10cm_Formic_AQ, m/z = 475 [M + H]+, Ret. Time = 2.93 min. 1RMN H (400 MHz, DMSO) δ 12,62-12,57 (m, 1H), 8,54 (s, 1H), 8,40 (s, 1H), 8,10 (d, J=2, 5 Hz, 1H), 7,69-7,60 (m , 1H), 7,51 (dd, J=2,7, 9,0 Hz, 1H), 7,32 (dd, J=8, 0 , 8,0 Hz, 2H), 7,07 (d, J = 9,1 Hz, 1H), 4,73-4,72 ( m, 1H), 3,70-3,64 (m, 1H) , 3,56 (d, J = 12,1 Hz, 2H), 2,90 (dd, J = 10,0, 10,0 Hz, 2H), 1,84 (d, J = 9,6 Hz, 2H ), 1,58-1,48 (m, 2H). Um proton trocável não foi observado I 1-(6-Amino-piridin-3-il)piperidin-4-ol 2-Brom-1,3-difluorbenzol I-34 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(4-piperidyl)-6H-1,6-naphthiridin-5-one Method 10cm_Formic AQ, m/z = 421 [M + H]+, Ret. Time = 2.07 min. 1RMN-H (400 MHz, DMSO) δ 12,34 (s, 1H), 8,17 (s, 1H), 8,10 (d, J = 3,0 Hz, 1H), 7,47 (dd, J = 3,0, 9,0 Hz, 1H), 7,34 (d, J = 7,3 Hz, 1H), 6,98 (d, J = 8,9 Hz, 1H), 6,47 ( d, J = 7,3 Hz, 1H), 4,71 (s, 1H), 3,68-3,62 (m, 1H), 3,56–3,50 (m, 2H), 3,03 (dd, J = 3,1, 8,8 Hz, 2H), 2,92–2,83 (m, 2H), 2,71–2,58 (m, 5H), 1,88–1,84 (m, 2H), 1,76-1,46 (m, 6H). B and G 1-(6-Amino-pyridin-3-yl)piperidin-4-ol (1-(tert-Butoxy-carbonyl)piperidin-4-yl)zincobromite I-35 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-norbornan-2-yl-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ Method, m/z = 432 [M + H]+, Ret. Time = 2.61 min. 1RMN H (400 MHz, DMSO) δ 12,27 (s, 1H), 11,49–11,47 (m, 1H), 8,19 (s, 1H), 8,08 (d, J = 3, 0 Hz, 1H), 7,46 (dd, J = 3,1, 9,0 Hz, 1H), 7,34–7,31 (m, 1H), 6,99–6,96 (m, 1H ), 6,48 (d, J = 7,3 Hz, 1H), 4,71 (d, J = 4,0 Hz, 1H), 3,69–3,61 (m, 1H), 3,56 –3,48 (m, 2H), 2,92–2,83 (m, 2H), 2,77 (dd, J = 5,4, 8,8 Hz, 1H), 2,41–2,32 (m, 2H), 2,08-2,00 (m, 1H), 1,89-1,81 (m, 2H), 1,74 (d, J = 9,4 Hz, 1H), 1, 64-1,46 (m, B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol Exo-2-norborn-ylzinc bromide 5H), 1,42-1,35 (m, 1H), 1,34-1,23 (m, 1H), 1,12 (d, J = 9,9 Hz, 1H). I-36 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-tetra-hydropyran-4-yl-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 422 [M + H]+, Zeit = 2.32 min. 1RMN-H (400 MHz, DMSO) δ 12,33 (s, 1H), 11,56–11,50 (m, 1H), 8,21 (s, 1H), 8,10 (d, J = 3 ,0 Hz, 1H), 7,47 (dd, J = 3,1, 9,0 Hz, 1H), 7,35 (dd, J = 5,5, 7,0 Hz, 1H), 6,99 (d, J = 8,9 Hz, 1H), 6,49 (d, J = 7,2 Hz, 1H), 4,72 (s, 1H), 4,00-3,94 (m, 2H) , 3,69-3,61 (m, 1H), 3,57-3,42 (m, 4H), 2,92-2,82 (m, 3H), 1,88-1,75 (m, 6H), 1,57-1,46 (M, 2H). N 1-(6-amino-pyridin-3-yl)piperidin-4-ol (tetra-hydro-2H-pyran-4-yl)zincobromate I-37 2-(1-hydroxyetil)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one Method 10cm_Bicarb_AQ, m/z = 382 [M + H]+, Ret. Time = 2.56 min. 1RMN-H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,53 (d, J = 5,0 Hz, 1H), 8,29 (s, 1H), 8,22 (s, 1H), 8,09 (d, J = 3,0 Hz). , 1H), 7,48 (dd, J = 3,1, 9,0 Hz, 1H), 7,35 (dd, J = 5,5, 7,0 Hz, 1H), 7,02 (d, J = 8,9 Hz, 1H), 6,48 (d, J = 7,3 Hz, 1H), 5,36 (s, 1H), 4,65 (q, J = 6,5 Hz, 1H) , 3,69–3,51 (m, 3H), 2,93–2,84 (m, 2H), 1,89–1,81 (m, 2H). ), 1,57–1,46 (m, 2H), 1,37 (d, J = 6,7 Hz, 3H). M 1-(6-Aminopyridin-3-yl)piperidin-4-ol I-38 2-(1-hydroxy-1-methylethyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridina 5-one 10cm_Formic_AQ method, m/z = 396 [M + H]+, Ret. Time = 2.29 min. 1RMN H (400 MHz, DMSO) δ 12,32 (s, 1H), 11,59–11,54 (m, 1H), 8,44 (s, 1H), 8,13 (d, J = 2, 5 Hz, 1H), 7,51 (dd, J = 2,9, 9,0 Hz, 1H), 7,38 (d, J = 7,3 Hz, 1H), 7,10-7,05 ( m, 1H), 6,53 (d, J = 7,3 Hz, 1H), 5,26 (s, 1H), 4,75 (d, J = 3,8 Hz, 1H), 3,74– 3,67 (m, 1H), 3,61–3,53 (m, 2H), 2,97–2,88 (m, 2H), 1,93–1,87 (m, 2H), 1, 62–1,52 (m, 2H), 1,50 (s, 6H). M 1-(6-Aminopyridin-3-yl)piperidin-4-ol I-39 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naftiridina-8 - Carbonsäure Method 10cm_Formic_AQ, m/z = 494 [M + H]+, Ret. Time = 2.68 min. 1RMN-H (400 MHz, DMSO) δ 12,83–12,77 (m, 1H), 8,65 (s, 1H), 8,38 (s, 1H), 8,15 (d, J = 2 ,8 Hz, 1H), 7,75–7,67 (m, 1H), 7,54 (dd, J = 2,8, 8,8 Hz, 1H), 7,40 (dd, J = 8, 3, 8,3 Hz, 2H), 7,15 (d, J = 8,8 Hz, 1H), 4,74 (s, 1H), 4,08 (s, 1H), 3,72-3, 67 (m, 1H), 3,60 (dd, J = 4,4, 8,2 Hz, 2H), 2,98-2,91 (m, 2H), 1,87 (dd, J = 3, 2, 12,8 Hz, 2H), 1,58–1,47 (m, 2H). 1 Austauschbares Proton nicht beobachtet I 1-(6-Amino-piridin-3-il)piperidin-4-ol 2-Brom-1,3-difluorbenzol I-40 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-norbornan-2-yl-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ Method, m/z = 432 [M + H]+, Ret. Time = 2.6 min. 1RMN-H (400 MHz, DMSO) δ 12,27 (s, 1H), 11,51–11,48 (m, 1H), 8,27 (s, 1H), 8,08 (d, J = 3 ,0 Hz, 1H), 7,49–7,44 (m, 1H), 7,33 (d, J = 7,3 Hz, 1H), 6,98 (d, J = 9,0 Hz, 1H ), 6,50-6,48 (m, 1H), 4,71 (d, J = 4,0 Hz, 1H), 3,68- 3,61 (m, 1H), 3,56-3, 48 (m, 2H), 3,30-3,24 (m, 1H), 2,91-2,83 (m, 2H), 2,35-2,30 (m, 1H), 1,93- 1,79 (metro, 4H). ), 1,62–1,37 (m, 6H), 1,31–1,20 (m, 3H). B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol Exo-2-norborn-ylzinc bromide I-41 2-(2-Fluoro-2-methylphenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 446 [M + H]+, Ret. Time = 3.01 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,65 (d, J = 5,4 Hz, 1 H), 8,69 (d, J = 1,6 Hz , 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,76 (dd, J = 2,0, 7,4 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,44–7,39 (m, 1 H), 7,34–7,30 (m, 1 H), 7,26-7,20 ( m, 1 H), 7,02 (d, J = 8,9 Hz, 1 H), 6,60 (d, J = 7,2 Hz, 1 H), 4,70 (brs, 1 H) , 3,68-3,60 (m, 1 H), 3,58-3,48 (m, 2 H), 2,92-2,83 (m, 2 H), 2,38 (s, 3 H ), 1,88-1,80 (m, 2 H), 1,56 -1,45 (m, 2 Std.). B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 2-Bromo-1-fluoro-4-methylbenzene I-42 2-(2-Fluoro-5-methoxy-fenil)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one 10cm_Formic_AQ method, m/z = 462 [M + H]+, Ret. Time = 2.53 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,68-11,62 (m, 1 H), 8,72 (d, J = 1,4 Hz, 1 H ), 8,07 (d, J = 3,0 Hz, 1 H), 7,50-7,46 (m, 2 H), 7,42 (d, J = 7,3 Hz, 1 H), 7,29 (dd, J = 9,0, 10,5 Hz, 1 H), 7,11-7,01 (m, 2 H ), 6,60 (d, J = 7,3 Hz, 1 H ), 4,70 (d, J = 4,3 Hz, 1 H), 3,83 (s, 3 H), 3,68-3,61 (m, 1 H), 3,57-3,49 ( m, 2 h), 2,92–2,83 (m, 2 h), 1,88–1,80 (m, 2 h), 1,56–1,45 (m, 2 h). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-methoxyphenyl)boronic acid I-43 2-(5-Cyclopropyl-2-fluoro-phenyl)-4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]-amino]-6H-1,6-nafthyridin-5-one Method 10cm_Formic_AQ, m/z = 0 [M + H]+, Ret. Time = 2.67 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,65 (s, 1 H), 8,67 (d, J = 1,6 Hz, 1 H), 8, 07 (d, J = 3,0 Hz, 1 H). ), 7,67 (d, J = 2,3, 7,4 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,42 (d, J = 7,4 Hz, 1 H), 7,24 (d, J = 7,4 Hz, 1 H), 7,21-7,18 (m, 1 H), 7,02 (d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz, 1 H), 3,67–3,61 (m, 1 H), 3,57–3,48 (m, 2 H), 2,91–2,83 (m, 2 H), 2,08–2,00 (m, 1 H), 1 ,88–1,80 (m, 2H), 1,56–1,44 (m, 2H), 1,02–0,96 (m, 2H), 0,73–0,68 (m, 2H) . A 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-(5-cyclo-propyl-2-fluoro-phenyl)-boronic acid I-44 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 338 [M + H]+, Ret. Time = 2.22 min. 1H-NMR (400 MHz, DMSO) δ 8.38 (d, J = 6.0 Hz, 1H), 8.19 (s, 1H), 8.15 (d, J = 5.8 Hz, 1H) , 8.05 (d, J=2.9Hz, 1H). ), 7.46 (dia, J=3.0, 9.0 Hz, 1H), 7.38 (dia, J=7.3 Hz, 1H), 7.03 (dia, J=8.9 Hz , 1H), 6.55 (dia, J=7.4 Hz, 1H) ). ), 3.68–3.60 (m, 1H), 3.54–3.46 (m, 2H), 2.89–2.81 (m, 2H), 1.87–1.80 (m , 2H), 1.54–1.44 (m, 2H). (1 Äq. Formiatsalz, 3 austauschbare Protonen nicht A (Subproduct) 1-(6-amino-pyridin-3-yl)piperidin-4-ol bis(pinacolate)diboron observed) I-45 4-Fluoro-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]benzonitrilo 10cm_Formic_AQ method, m/z = 457 [M + H]+, Ret. Time = 2.5 min. 1RMN-H (400 MHz, DMSO) δ 12,42 (s, 1 H), 11,65 (s, 1 H), 8,67 (d, J = 1,6 Hz, 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,67 (dd, J = 2,3, 7,4 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,42 (d, J = 7,4 Hz, 1 H), 7,24 (d, J = 7,4 Hz, 1 H), 7,21-7,18 (m, 1 H), 7,02 (d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz , 1 H), 3,67–3,61 (m, 1 H), 3,57–3,48 (m, 2 H), 2,91–2,83 (m, 2 H), 1,88 –1,80 (m, 2H), 1,56–1,44 (M, 2H). 1-(6-amino-pyridin-3-yl)-piperidin-4-ol(5-cyano-2-fluoro-phenyl)-boronic acid I-46 2-[6-[[2-(2,6-Difluoro-phenyl)-5-oxo-6H-1,6-naphthyridin-4-yl]amino]-3-pyridyl]-N-ethyl-2-methyl - Propanamide 10cm_Formic_AQ method, m/z = 464 [M + H]+, Ret. Time = 2.67 min. 1RMN H (400 MHz, DMSO) δ 12,89 (s, 1H), 11,84 (d, J = 1,0 Hz, 1H), 8,80 (s, 1H), 8,36 (d, J = 2,3 Hz, 1H), 7,71 (dd , J = 2,5, 8,6 Hz, 1H), 7,68–7,59 (m, 1H), 7,52 (d, J = 7,3 Hz, 1H), 7,45 (dd, J = 5,3, 5,3 Hz, 1H), 7,32 (dd, J = 8,0, 8,0 Hz, 2H), 7, 11 (d, J = 8,6 Hz, 1H), 6,65 (d, J = 7,1 Hz, 1H), 3,14-3,05 (m, 2H), 1,50 (s, 6H ), 1,00 (t, J = 7,2 Hz, 3H). B CA1 2-Brom-1,3-difluorbenzol I-47 2-Acetyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on Method 10cm_Formic_AQ, m/z = 380 [M + H]+, Ret. Time = 2.33 min. 1RMN-H (400 MHz, DMSO) δ 12,46 (s, 1H), 11,78 (s, 1H), 8,71 (s, 1H), 8,15–8,10 (m, 1H), 7,53–7,42 (m, 2H), 7,02 (d, J = 8,8 Hz, 1H), 6,63 (d, J = 7,3 Hz, 1H), 4,71 (s , 1H), 3,71–3,49 (m, 3H), 2,96–2,84 (m, 2H), 2,65 (s , 3H), 1,85 (s, 2H), 1, 53 (t, J = 9,3 Hz, 2H). M 1-(6-Aminopyridin-3-yl)piperidin-4-ol I-48 2-(2-Fluoro-5-isopropylphenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 474 [M + H]+, Ret. Time = 2.75 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,68-11,62 (m, 1 H), 8,68 (d, J = 1,6 Hz, 1 H ), 8,06 (d, J = 3,0 Hz, 1 H), 7,78 (dd, J = 2,4, 7,5 Hz, 1 H), 7,48 (dd, J = 3, 1, 9,0 Hz, 1 H), 7,43-7,38 (m, 2 H), 7,27 (dd, J = 8,5, 11,0 Hz, 1 H), 7,03 ( d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz, 1 H), 3,67 - 3,61 (m, 1 H), 3,56-3,48 (m, 2 H), 3,06-2,95 (m, 1 H), 2,91-2,83 (m, 2 H), 1,88-1,80 (m, 2 H), 1,55- 1,44 (m, 2 H), 1,26 (d, J = 6,9 Hz, 6 H). 1-(6-amino-pyridin-3-yl)-piperidin-4-ol-[2-fluoro-5-(propan-2-yl)-phenyl]-boronic acid I-49 2-(2,6-difluorophenyl)-8-ethyl-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z 478 [M + H]+, Ret. Time = 2.8 min. 1RMN-H (400 MHz, DMSO) δ 12,78 (s, 1H), 11,71–11,65 (m, 1H), 8,60–8,53 (m, 1H), 8,09 (d , J = 2,8 Hz, 1H), 7,67–7,57 (m, 1H), 7,51 (dd, J = 2,8, 9,1 Hz, 1H), 7,35–7, 27 (m, 3H), 7,06–7,02 (m, 1H), 4,72 (s, 1H), 3,72–3,63 (m, 1H), 3,59–3,51 ( m, 2H), 2,93–2,87 (m, 2H), 2,71 (q, J = 7,2 Hz, 2H), 1,86 (d, J = 9,6 Hz, 2H), 1,58–1,47 (m , 2H), 1,22 (t, J = 7,5 Hz, 3H). K 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-2-bromo-1,3-difluorobenzene I-50 2-(3-hydroxy-1-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one 10cm_Formic_AQ method, m/z = 437 [M + H]+, Ret. Time = 2.32 min. 1RMN-H (400 MHz, DMSO) δ 12,25 (s, 1H), 11,15 (s, 1H), 8,04 (s, 1H), 7,92 (s, 1H), 7,47– 7,41 (m, 1H), 7,21 (s, 1H), 6,92–6,87 (m, 1H), 6,22 (d, J = 6,9 Hz, 1H), 4,30 (d, J = 11,8 Hz, 1H), 4,14–4,06 (m, 1H), 3,62–3,50 (m , 4H), 3,04–2,98 (m, 1H ), 2,89–2,79 (m, 3H), 1,94–1,75 (m, 4H), 1,55–1,41 (m, 4H). E 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-3-hydroxypiperidina I-51 4-Fluoro-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]benzamide Method 10cm_Formic_AQ, m/z = 475 [M + H]+, Ret. Time = 2.35 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,67 (s, 1 H), 8,73 (d, J = 1,6 Hz, 1 H), 8, 47 (dd, J = 2,4, 7,5 Hz, 1 H), 8,13 (s, 1 H), 8,09-8,00 (m, 2 H), 7,51-7,42 (m, 4 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4 ,3 Hz, 1 H), 3,68–3,61 (m, 1 H), 3,57–3,49 (m, 2 H), 2,92–2,83 (m, 2 H) , 1,88–1,80 (m, 2 H), 1,56–1,45 (m, 2 H). A 1-(6- Amino-pyridin-3-yl) piperidin- 4-ol 5- Carbamoyl- 2-fluoro- phenyl- borosaurium I-52 2-[6-[[2-(2,6-Difluoro-phenyl)-5-oxo-6H-1,6-naphthyridin-4-yl]amino]-3-pyridyl]-N-ethyl-N,2 - Dimethylpropanamide Method 10cm_Bicarb_AQ, m/z = 478 [M + H]+, Ret. Time = 3.3 min. 1RMN-H (400 MHz, DMSO) δ 12,73 (s, 1H), 11,50–11,47 (m, 1H), 8,60 (s, 1H), 8,21 (d, J = 2 ,5 Hz, 1H), 7,56–7,50 (m, 2H), 7,39 (d, J = 7,3 Hz, 1H), 7,19 (dd, J = 8,0, 8, 0 Hz, 2H), 7,07 (d, J = 8,5 Hz, 1H), 6,57 (d, J = 7,3 Hz, 1H), 3,19–3,13 (m, 2H) , 2,60 (s, 3H), 1,46 (s, 6H), 0,89 (dd, J = 7,0, 7,0 Hz, 3H). B CA2 2- Bromo-1,3-difluorbenzol I-53 Ethyl-2-[6-[[2-(2,6-difluoro-phenyl)-5-oxo-6H-1,6-naphthiridin-4-yl]amino]-3-pyridyl]-2-methyl-propanoato Method 10cm_Bicarb_AQ, m/z = 465 [M + H]+, Ret. Time = 3.56 min. 1RMN-H (400 MHz, DMSO) δ 12,92 (s, 1H), 11,87 (s, 1H), 8,80 (s, 1H), 8,39 (d, J = 2,3 Hz, 1H), 7,78 (dd, J = 2,5, 8,6 Hz, 1H), 7,67-7,60 (m, 1H), 7,52 (d, J = 7,3 Hz, 1H) ), 7,32 (dd, J = 8,0, 8,0 Hz, 2H), 7,12 (d, J = 8,6 Hz, 1H), 6,65 (d, J = 7,1 Hz , 1H), 4,12 (q, J = 7,1 Hz, 2H), 1,57 (s, 6H), 1,17 (dd, J = 7,1, 7,1 Hz, 3H). B CA1.1 2-bromo-1,3-difluorbenzol I-54 4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-2-[(3-metil-oxetan-3-yl)metil]-6H-1,6-naphthiridina- 5- one Method 10cm_Formic_AQ, m/z = 422 [M + H]+, Zeit = 2.38 min. 1H-NMR (400 MHz, DMSO) δ 15.30 (s, 1H), 8.38 (s, 1H), 8.16 (d, J = 3.1 Hz, 1H), 8.14 (s, 1H), 8.03 (d, J=6.3Hz). , 1H), 7.52 (dd, J=3.1, 9.0Hz, 1H), 7.10 (d, J=8.9Hz, 1H), 6.42 (d, J=6, 3Hz, 1H), 4.26 (d, J=11.7Hz). , 2H), 4.01 (d, J=11.7Hz, 2H), 3.72–3.64 (m, 1H), 3.63–3.55 (m, 2H), 3.24 ( d, J=17.9Hz, 1H), 3.05 (d, J=18.3Hz, 1H), 2.99-2.91 (m, 2H), 1.89- N 1-(6-amino-pyridin-3-yl)piperidin-4-ol((3-methyl-oxetan-3-yl)methyl)cincobromet 1.80 (m, 2H), 1.55-1.45 (m, 2H), 1,20 (s, 3H). (1 eq. format salt, 2 non-exchangeable proton observed) I-55 2-(2- Fluor-4-methoxy-5-metil-fenil)-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- naphthiridina- 5 -eins 10cm_Formic_AQ method, m/z = 476 [M + H]+, Ret. Time = 2.64 min. 1RMN-H (400 MHz, DMSO): δ 12,36 (s, 1 H), 11,59 (s, 1 H), 8,70 (d, J = 1,1 Hz, 1 H), 8, 07 (d, J = 3,0 Hz, 1 H). ), 7,83 (d, J = 8,9 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,39 (d, J = 7, 3 Hz, 1 H), 7,02 (d, J = 8,9 Hz, 1 H), 6,98 (s, 1 H), 6,59 (d, J = 7,3 Hz, 1 H) , 4,70 (d, J = 4,3 Hz, 1 H), 3,89 (s, 3 H), 3,68- 3,61 (m, 1 H), 3,57–3,49 ( m, 2 H), 2,92–2,84 (m, 2 H), 2,20 (s, 3 H), 1,88–1,81 (m, 2 H), 1,56–1, 45 (M, 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB3 I-56 2-(2,6-difluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-8-phenyl]-6H- 1,6-naphthyridin-5-ein 10cm_Formic_AQ method, m/z = 526 [M + H]+, Ret. Time = 2.96 min. 1RMN-H (400 MHz, DMSO) δ 12,79 (s, 1H), 12,06 (s, 1H), 8,59 (s, 1H), 8,11 (d, J = 2,5 Hz, 1H), 7,67–7,49 (m, 5H), 7,44-7,24 (m, 5H), 7,08 (d, J = 8,8 Hz, 1H), 4,73 (s , 1H), 3,69-3,64 (m, 1H), 3,56 (d, J = 12,6 Hz, 2H), 2,95–2,88 (m, 2H), 1,86 ( d, J = 9,6 Hz, 2H), 1,58–1,48 (m, 2H). J 1-(6-amino-pyridin-3-yl)piperidin-4-ol phenylboronic acid (step 2) I-57 2-isopropyl-4-[[5-(4-methyl-piperazin-1-yl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one Method 10cm_Formic_AQ, m/z = 379 [M + H]+, Ret. Time = 2.14 min. 1RMN-H (400 MHz, DMSO) δ 8,16 (d, J = 6,3 Hz, 1H), 8,07 (d, J = 2,7 Hz, 1H), 7,67–7,54 ( m, 2H), 7,47 (dd, J = 3,1, 9,0 Hz, 1H), 7,34 (d, J = 7,7 Hz, 1H), 7,02 (d, J = 8 ,5 Hz, 1H), 6,54 (d, J = 7,3 Hz, 1H), 3,20–3,16 (m, 4H), 2,96–2,88 (m, 1H), 2 ,60–2,56 (m, 4H), 2,28 (s, 3H), 1,23 (d, J = 6,9 Hz, 6H). B 5-(4-methyl-piperazin-1-yl)pyridin-2-amina 2-propylcincbromuro I-58 N-[3-Fluor-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ] Cyclopropanecarboxamide Method 10cm_Bicarb_AQ, m/z = 515 [M + H]+, Ret. Time = 3.01 min. 1HR-NMR (400 MHz, DMSO): δ 12.38 (s, 1H), 11.61 (d, J = 4.4 Hz, 1H), 10.60 (s, 1H), 8, 73 (s, 1H), 8.08 (d, J = 3.0 Hz, 1H), 8.01 (dd, J = 8.8, 8.8 Hz, 1H), 7.79– 7.69 (m, 1H), 7.50–7.37 (m, 3H), 7.02 (d, J = 8.9 Hz, 1H), 6.58 (d, J = 7 .2 Hz, 1H), 4.70 (d, J = 4.1 Hz, 1H), 3.68-3.60 (m, 1H), 3.58-3.51 (m, 2 standard), 2.92–2.83 (m, 2 standard), 1.88–1.82 (m, 3 standard), 1.57–1.45 (m, 2 standard), 0.89–0 .84 (m, 4 standard). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB2.1 I-59 5-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-7-isopropyl-3H-pyrido[2,3-d]pyrimidin-4-one Method 10cm_Formic_AQ, m/z = 381 [M + H]+, Ret. Time = 2.38 min. 1RMN H (400 MHz, DMSO) δ 12,06-11,99 (m, 1H), 8,26 (s, 1H), 8,21 (s, 1H), 8,14 (d, J=2, 8 Hz, 1H), 7,51 (dd, J =2,9, 9,0 Hz, 1H), 7,05 (d, J=9,1 Hz, 1H), 4,74 (s, 1H ) , 3,72–3,66 (m, 1H), 3,62–3,53 (m, 2H), 3,02–2,88 (m, 3H), 1,93–1,87 (m, 2H), 1,61-1,50 (m, 2H), 1,30 (d, J = 7,1 Hz, 6H). 1-Protón trocável não observado U 1-(6-Amino-pyridin-3-yl)piperidin-4-ol I-60 5-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-7-propyl-3H-pyrido[2,3-d]pyrimidin-4-one Method 10cm_Formic_AQ, m/z = 381 [M + H]+, Ret. Time = 2.39 min. 1RMN-H (400 MHz, DMSO) δ 12,55 (s, 1H), 11,78 (s, 1H), 8,27 (s, 1H), 8,18–8,13 (m, 2H), 7,51 (dd, J = 2,9, 9,0 Hz, 1H), 7,07 (d, J = 8,8 Hz, 1H), 4,74 (d, J = 3,5 Hz, 1H) ), 3,71–3,67 (m, 1H), 3,62–3,53 (m, 2H), 2,97–2,87 (m , 2H), 2,70 (dd, J = 7 ,6, 7,6 Hz, 2H), 1,93– 1,87 (m, 2H), 1,76 (dd, J = 7,3, 14,9 Hz, 2H), 1,61– 1, 49 (m, 2H), 0,99 (dd, J = 7,5, 7,5 Hz, 3H). U 1-(6-Amino-pyridin-3-yl)piperidin-4-ol I-61 2-[2- Fluor-4-(4-pyridyl-methoxy)fenil]-4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-6H-1,6- naftiridina- 5-ein 10cm_Formic_AQ method, m/z = 539 [M + H]+, Ret. Time = 2.34 min. 1RMN-H (400 MHz, DMSO): γ; 12,39 (s, 1 H), 11,61 (s, 1 H), 8,71 (d, J = 1,0 Hz, 1 H), 8,63–8,61 (m, 2 H) , 8,07 (d, J = 3,0 Hz, 1 H), 8,01 (dd, J = 9,2, 9,2 Hz, 1 H), 7,50-7,45 (m, 3 H), 7,40 (d, J = 7,3 Hz, 1 H), 7,09 (d, J = 7,3 Hz, 1 H), 7,07–7,00 (m, 2 H) , 6,57 (d, J = 7,3 Hz, 1 H), 5,32 (s, 2 H), 4,71 (d, J = 4,1 Hz, 1 H), 3,68–3 ,61 (m, 1 H), 3,57–3,49 (m, 2 H), 2,92–2,83 (m, 2 H), 1,88–1,80 (m, 2 H) , 1,56–1,45 (M, 2H). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB5 I-62 3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]-N,N - Dimetil-benzamdie Method 10cm_Bicarb_AQ, m/z = 503 [M + H]+, Ret. Time = 2.84 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,71 (s, 1 H), 8,80 (s, 1 H), 8,12 (d, J = 2 ,5 Hz, 1 H), 8,07 (dd , J = 7,8, 7,8 Hz, 1 H), 7,52 (dd, J = 2,8, 8,8 Hz, 1 H), 7,50–7,40 (m, 3 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4, 74 (d, J = 4,0 Hz, 1 H), 3,72–3,66 (m, 1 H), 3,56 (d, J = 12,4 Hz, 2 H), 3,07 ( s, 3 H), 3,02 (s, 3 H), 2,92 (dd, J = 10,0, 10,0 Hz, 2 H), 1,88 (d, J = 9,6 Hz, 2 H), 1,59–1,49 (m, 2 H). A 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-(4-(dimethyl-carbamoyl)-2-fluoro-phenyl)-boronic acid I-63 2-(1-Acetyl-4-piperidyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on 10cm_Formic_AQ method, m/z = 463 [M + H]+, Ret. Time = 2.31 min. 1H-NMR (400 MHz, DMSO) &dr; 12,34 (s, 1H), 11,55 (d, J = 2,3 Hz, 1H), 8,37–8,31 (m, 1H), 8,11 (d, J = 6,5 Hz , 1H), 8,06 (d, J = 3,3 Hz, 1H), 7,46 (dd, J = 3,0, 9,0 Hz, 1H), 7,34 (d, J = 7, 3 Hz, 1H), 7,00 (d, J = 8,8 Hz, 1H), 6,52 (d, J = 7,3 Hz, 1H), 4,52–4,47 (m, 1H) , 3,95–3,91 (m, 1H), 3,68–3,61 (m, 1H), 3,54–3,46 (m, 2H), 3,18–3,10 (m, 1H), 2,89–2,80 (m, 3H), 2,68–2,60 (m, 1H), 2,04 (s, 3H), 1,89–1,80 (m, 4H) , 1,71–1,44 (m, 4H). (1 Äq. Formiatsalz, 1 austauschbares Proton nicht beobachtet) O 1-(6-Amino-pyridin-3-yl)piperidin-4-ol (1-(tert-Butoxy-carbonyl)piperidin-4-yl)zincobromite I-64 2-(2-fluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 432 [M + H]+, Ret. Time = 3.09 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,69 (s, 1 H), 8,76 (s, 1 H), 8,11 (d, J = 2 ,8 Hz, 1 H), 8,01 (dd , J = 7,2, 7,2 Hz, 1 H), 7,62-7,46 (m, 2 H) 7,44-7,36 ( m, 2 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 4, 0 Hz, 1 H), 3,73–3,66 (m, 1 H), 3,57 (d, J = 12,4 Hz, 2 H), 2,92 (dd, J = 10,0 , 10,0 Hz, 2 H), 1,92–1,84 (m, 2 H), 1,60–1,48 (m, 2 H), 1,14 (dd, J = 6,9, 6 ,9Hz, 1H). 1-(6-amino-pyridin-3-yl)-piperidin-4-ol-2-fluoro-phenyl-boronic acid I-65 2-(2-Fluor-3-methoxy-fenil)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one Method 10cm_Bicarb_AQ, m/z = 462 [M + H]+, Ret. Time = 3.05 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,68 (s, 1 H), 8,69 (s, 1 H), 8,11 (d, J = 2 ,5 Hz, 1 H), 7,76 (dd , J - 7,1, 7,1 Hz, 1 H), 7,52 (dd, J = 2,9 9,0 Hz, 1 H), 7 ,44 (dd, J = 7,1, 7,1 Hz, 2 H), 7,27 (dd, J = 7,6, 7,6 Hz, 1 H), 7,07 (d, J = 8 ,8 Hz, 1 H), 6,62 (d, J = 7,1 Hz, 1 H), 4,73 (d, J = 4,0 Hz, 1 H), 4,38 (s, 3 H ), 3,73–3,65 (m, 1 H), 3,60–3,52 (m, 2 H), 2,91 (dd, J = 9,9, 9,9 Hz, 2 H) , 1,89 (d, J = 9,3 Hz, 2H), 1,60-1,48 (m, 2 H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol-2-fluoro-3-methoxyphenylboronic acid I-66 2-(5-ethoxy-2-fluoro-phenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]-amino]-6H-1,6-naphthyridin-5-uno Method 10cm_Bicarb_AQ, m/z = 476 [M + H]+, Ret. Time = 3.22 min. 1RMN-H (400 MHz, DMSO): γ; 12,46 (s, 1 H), 11,68 (s, 1 H), 8,76 (s, 1 H), 8,11 (d, J = 2,8 Hz, 1 H), 7,55 –7,48 (m, 2 H), 7,46 (d, J = 7,3 Hz, 1 H), 7,32 (d, J = 9,3 Hz, 1 H), 7,14-7 ,04 (m, 2 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,73 (d, J = 3,8 Hz, 1 H), 4,12 (q, J = 6,9 Hz, 2 H), 3,73–3,64 (m, 1 H), 3,57 (d, J = 12,6 Hz). , 2 H), 2,91 (dd, J = 10,0, 10,0 Hz, 2 H), 1,89 (d, J = 9,3 Hz, 2 H), 1,60-1,50 (m, 2 H), 1,40 (dd, J = 6,9, 6,9 Hz, 3 H). 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-5-ethoxy-2-fluoro-phenyl-boronic acid I-67 2-(2-Fluoro-5-isopropoxyphenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one Method 10cm_Bicarb_AQ, m/z = 490 [M + H]+, Ret. Time = 3.31 min. 1RMN-H (400 MHz, DMSO): γ; 12,46 (s, 1 H), 11,68 (s, 1 H), 8,76 (s, 1 H), 8,11 (d, J = 2,5 Hz, 1 H), 7,55 –7,43 (m, 3 H), 7,31 (d, J = 9,3 Hz, 1 H), 7,14–7,05 (m, 2 H), 6,64 (d, J = 7,1 Hz, 1 H), 4,75–4,63 (m, 2 H), 3,73-3,66 (m, 1 H), 3,61-3,53 (m, 2 H) , 2,96-2,87 (m, 2 H), 1,88 (d, J = 9,3 Hz, 2 H), 1,60-1,48 ( m, 2 H), 1,34 ( d, J = 5,8 Hz, 6 H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-isopropoxy-phenyl)boronic acid I-68 2-(2-Fluoro-3-methylphenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 446 [M + H]+, Ret. Time = 2.68 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,68 (s, 1 H), 8,69 (s, 1 H), 8,11 (d, J = 2 ,5 Hz, 1 H), 7,76 (dd , J = 7,1, 7,1 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,44 (dd, J = 7,1, 7,1 Hz, 2 H), 7,27 (dd, J = 7,6, 7,6 Hz, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7,1 Hz, 1 H), 4,73 (d, J = 4,0 Hz, 1 H), 3,73–3,65 (m, 1 H), 3,60-3,52 (m, 2 H), 2,91 (dd, J = 9,9, 9,9 Hz, 2 H), 2,38 (s, 3 H ), 1,89 (d, J = 9,3 Hz, 2 H ), 1,60–1,48 (m, 2 H). A 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-2-fluoro-3-methyl-phenylborosaur I-69 2-[2-Fluoro-4-(1-hydroxy-1-methylethyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6- Naphthyridin-5-one 10cm_Formic_AQ method, m/z = 490 [M + H]+, Ret. Time = 2.63 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,66 (d, J = 5,3 Hz, 1 H), 8,89 (s, 1 H), 8, 20 (d, J = 2,8 Hz, 1 H). ), 7,89 (dd, J = 1,4, 8,2 Hz, 1 H), 7,86–7,78 (m, 2 H), 7,53 (dd, J = 3,0, 8 ,8 Hz, 1 H), 7,49–7,42 (m, 1 H ), 7,09 (d, J = 8,8 Hz, 1 H), 6,66–6,63 (m, 1 H), 5,42 (s, 1 H), 4,75 (d, J = 4,0 Hz, 1 H), 3,73–3,66 (m, 1 H), 3,63–3, 57 (m, 2 H), 2,97–2,89 (m, 2 H), 1,90 (dd, J = 3,2, 12,8 Hz, 2 H), 1,58 (s, 6 H), 1,56 -1,52 (m, 2 Std.). A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-(3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2). ). ). -yl)phenyl)propan-2-ol I-70 3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]-N-methyl - Benzamide 10cm_Formic_AQ method, m/z = 489 [M + H]+, Ret. Time = 2.48 min. 1RMN-H (400 MHz, DMSO): γ; 12,50 (s, 1 H), 11,74–11,67 (m, 1 H), 8,79 (s, 1 H), 8,69 (d, J = 4,5 Hz, 1 H) , 8,13 -8,07 (m, 2 H), 7,86 (d, J = 8,3 Hz, 1 H), 7,80 (s, 1 H), 7,55-7,45 ( m, 2 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 1, 5 Hz, 1 H), 3,68 (d, J = 2,5 Hz, 1 H), 3,57 (d, J = 12,4 Hz). , 2 H), 2,96 (d, J = 12,4 Hz, 2 H), 2,94-2,86 (m, 3 H), 1,88 (d, J = 9,6 Hz, 2 H), 1,60-1,49 (m, 2H) . 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-4-(methyl-carbamoyl)phenyl)boronic acid I-71 2-[2-Fluoro-4-(piperidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine 5 -eins 10cm_Formic_AQ method, m/z = 543 [M + H]+, Ret. Time = 2.68 min. 1RMN-H (400 MHz, DMSO): γ; 12,49 (s, 1 H), 11,74–11,69 (m, 1 H), 8,79 (s, 1 H), 8,12 (d, J = 2,5 Hz, 1 H) , 8,07 (dd, J = 7,8, 7,8 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,47 (d, J = 7,3 Hz, 1 H), 7,42–7,37 (m, 2 H ), 7,08 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7 ,1 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 3,72–3,63 (m, 3 H), 3,61–3,53 (m, 2H ), 2,92 (dd, J = 9,9, 9,9 Hz, 2H), 1,89–1,86 (m, 2H), 1,73–1,50 (m, 10H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-4-(piperidine-1-carbonyl)phenyl)boronic acid I-72 3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]benzamide Method 10cm_Formic_AQ, m/z = 475 [M + H]+, Ret. Time = 2.71 min. 1RMN-H (400 MHz, DMSO): δ 12,32 (s, 1 H), 11,62 (s, 1 H), 10,90 (s, 1 H), 8,97 (s, 1 H) , 8,13 (d, J = 2,8 Hz). , 1 H), 7,95–7,87 (m, 2 H), 7,65–7,58 (m, 1 H), 7,56–7,47 (m, 2 H), 7,42 (d, J = 7,3 Hz, 1 H), 7,13 (d, J = 8,8 Hz, 1 H), 6,47 (d, J = 7,3 Hz, 1 H), 4, 74 (s, 1 H), 3,69 (s, 1 H), 3,61-3,55 (m, 2 H), 2,98–2,89 (m, 2H), 1,94–1 ,88 (m, 2H), 1,62–1,50 (m, 2H). 1-(6-amino-pyridin-3-yl)-piperidin-4-ol(4-carbamoyl-2-fluoro-phenyl)-boronic acid I-73 2-(4-Benzyloxi-2-fluoro-phenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 538 [M + H]+, Ret. Time = 2.83 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,61 (s, 1 H), 8,81 (s, 1 H), 8,16 (d, J = 3 ,0 Hz, 1 H), 7,86 (d , J = 8,7 Hz, 1 H), 7,53-7,38 (m, 9 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 5,29 (s, 2 H), 4,72 (d, J = 4,1 Hz, 1 H), 3,69–3,62 (m, 1 H), 3,59–3,51 (m, 2 H), 2,94–2,85 (m, 2 H), 1,90–1,82 ( m, 2 H), 1,58-1,47 (m, 2 H). 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-4-benzyloxy-2-fluoro-phenyl-boronic acid I-74 2-[4- [(Dimetilamino)metil]-2-fluorophenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- Naftiridina-5- sober 10cm_Formic_AQ method, m/z = 489 [M + H]+, Ret. Time = 2.3 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,68 (d, J = 5,6 Hz, 1 H), 8,75 (s, 1 H), 8, 11 (d, J = 2,5 Hz, 1 H). ), 7,97 (dd, J = 8,1, 8,1 Hz, 1 H), 7,52 (dd, J = 2,8, 9,1 Hz, 1 H), 7,45 (dd, J = 6,4, 6,4 Hz, 1 H), 7,32 (d , J = 8,8 Hz, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6 ,62 (d, J = 7,1 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 3,72 -3,65 (m, 1 H), 3,61 -3,51 (m, 5 H), 2,96-2,87 (m, 2 H), 2,25 (s, 6 H), 1,88 (d, J = 9,9 Hz, 2 H ), A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-(3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2 ).-yl)phenyl)- N,N- 1,60-1,49 (m, 2 H). Dimetil I-75 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(1-metil-4-piperidyl)-6H-1,6-naphthiridin-5-one Method 10cm_Formic_AQ, m/z = 435 [M + H]+, Ret. Time = 2.11 min. 1RMN-H (400MHz, DMSO) δ 13.08 (s, 1H), 12.49 (s, 1H), 9.71-9.70 (m, 1H), 8.32-8.28 (m , 1H), 8.15-8.13 (m, 1H), 7.77 (s, 1H), 7.58-7.53 (m, 1H), 7.26-7.20 (m, 1H ), 6.70 (s, 1H), 3.73-3.56 (m, 5H), 3.17-3.08 (m, 3H), 3.02-2.94 (m, 2H), 2.87 (s, 3H), 2.24-2.20 (m, 2H), 2.01-1.82 (m, 4H), 1.55-1.45 (m, 2H). (1 eq. formate salt, 1 exchangeable proton not observed) O 1-(6-amino-pyridin-3-yl)piperidin-4-ol (1-(tert-Butoxy-carbonyl)piperidin-4-yl)zincobromite I-76 N-[4-Fluoro-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl]phenyl ] Acetamide 10cm_Formic_AQ method, m/z = 489 [M + H]+, Ret. Time = 2.39 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,70 (s, 1 H), 10,18 (s, 1 H), 8,76 (s, 1 H) , 8,16–8,10 (m, 2H). ), 7,88–7,83 (m, 1 H), 7,52 (dd, J = 2,8, 8,8 Hz, 1 H), 7,47 (d, J = 7,3 Hz, 1 H), 7,33 (dd, J = 9,2, 10,5 Hz). , 1 H), 7,07 (d, J = 9,1 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 4,3 Hz, 1 H), 3,72-3,66 (m, 1 H), 3,57 (d, J = 12,6 Hz, 2 H), 2,95–2,87 (m, 2 H) , 2,11 (s, 3 H), 1,92–1,86 (m, 2 H), 1,60–1,48 (m , 2 Std.). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol N-(4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2 ). ). - yl)phenyl)acetamid I-77 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(2,2,2-trifluoro-1-hydroxy-1-methylethyl)-6H-1,6 - Naphthyridine -5-uno Method 10cm_Formic_AQ, m/z = 450 [M + H]+, Ret. Time = 2.48 min. 1RMN-H (400 MHz, DMSO) δ 12,43 (s, 1H), 11,71 (s, 1H), 8,64 (s, 1H), 8,14 (d, J = 2,8 Hz, 1H), 7,53 (dd, J = 2,9, 9,0 Hz, 1H), 7,45 (d, J = 7,3 Hz, 1H), 7,06 (d, J = 8,8 Hz, 1H), 6,75 (s, 1H), 6,58 (d, J = 7,1 Hz, 1H), 4,74 (d, J = 4,3 Hz, 1H), 3,73– 3,67 (m, 1H), 3,63–3,55 (m, 2H), 2,98–2,90 (m, 2H), 1,88 (d, J = 9,9 Hz, 2H) , 1,75 (s, 3H), 1,61-1,49 (m, 2H). M 1-(6-Aminopyridin-3-yl)piperidin-4-ol I-78 N-[1-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]-3-piperidyl] acetamide 10cm_Formic_AQ Method, m/z = 478 [M + H]+, Ret. Time = 2.27 min. 1H-NMR (400 MHz, DMSO) &dr; 12,24 (s, 1H), 11,15 (d, J = 5,4 Hz, 1H), 8,02–7,97 (m, 2H), 7,81 (d, J = 5,4 Hz , 1H), 7,43 (dd, J = 2,9, 9,0 Hz, 1H), 7,22 (d, J = 7,4 Hz, 1H), 6,93 (d, J = 9, 0 Hz, 1H), 6,28 (d, J = 7,3 Hz, 1H), 4,69 (s, 1H), 4,18-4,14 ​​​​(m, 1H), 3, 99 ( 1H, d, J = 13,7 Hz), 3,67-3,59 (m, 2H), 3,48-3,42 (m, 2H), 3,22-3,16 (m, 1H), 2,99 (dd, J = 9,0, 12,6 Hz, E 1-(6-amino-pyridin-3-yl) piperidin-4-ol N-(piperidin-3-yl) acetamida (etapa 2) 1H), 2,85-2,77 (M, 2H), 1,82-1,80 (m, 7H), 1,55- 1.44 (metro, 4H). I-79 4-Fluoro-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]-N,N - Dimetilbenzamide 10cm_Formic_AQ method, m/z = 503 [M + H]+, Ret. Time = 2.4 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,68 (s, 1 H), 8,81 (s, 1 H), 8,11 (d, J = 2 ,8 Hz, 1 H), 8,06 (dd , J = 1,8, 7,3 Hz, 1 H), 7,66–7,61 (m, 1 H), 7,55–7,43 (m, 3 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 4 ,3 Hz, 1 H), 3,72-3,66 (m, 1 H), 3,62-3,53 (m, 2 H), 3,04 (d, J = 12,9 Hz, 6 H), 2,92 (dd, J = 10,0, 10,0 Hz, 2 H), 1,88 (d, J = 9,6 Hz, 2 H), 1,60–1,49 (m , 2H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(5-dimethyl-carbamoyl)-2-fluoro-phenyl)boronic acid I-80 2-[2-Fluoro-5-(morpholine-4-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine- 5-eins Method 10cm_Formic_AQ, m/z = 545 [M + H]+, Ret. Time = 2.39 min. 1RMN-H (400 MHz, DMSO) δ 12,37 (s, 1H), 11,60 (s, 1H), 8,69 (s, 1H), 8,00–7,93 (m, 2H), 7,54–7,49 (m, 1H), 7,43– 7,32 (m, 3H), 6,96 (d, J = 9,1 Hz, 1H), 6,53 (d, J = 7,3 Hz, 1H), 4,62 (d, J = 3,8 Hz, 1H), 3,56 (d, J = 3,8 Hz, 6H), 3,54–3,38 (m, 5H), 2,83–2,75 (m, 2H), 1,75 (d, J = 9,9 Hz, 2H), 1,48–1,36 (m, 2H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-morpholine-4-carbonyl)phenyl)boronic acid I-81 2-[(3S)-3-Hydroxy-1-piperidyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one 10cm_Formic_AQ method, m/z = 437 [M + H]+, Ret. Time = 2.26 min. 1RMN-H (400 MHz, DMSO) δ 12,26 (s, 1H), 11,14 (d, J = 5,6 Hz, 1H), 8,04 (d, J = 3,0 Hz, 1H) , 7,92 (s, 1H), 7,44 (dd , J = 3,1, 9,0 Hz, 1H), 7,21 (dd, J = 6,0, 7,1 Hz, 1H), 6,89 (d, J = 8,9 Hz, 1H), 6,23 (dd, J = 0,7, 7,2 Hz, 1H), 4,94–4,71 (m, 2H), 4 ,32–4,26 (m, 1H), 4,12 (d, J = 12,9 Hz, 1H), 3,67–3,60 (m, 1H), 3,53–3,47 (m , 3H), 3,05 -2,98 (m, 1H), 2,89-2,78 (m, 3H), E 1-(6-amino-pyridin-3-yl)piperidin-4-ol (S)-piperidin-3-ol (stage 2) 1,95-1,73 (m, 4H), 1,56- 1.38 (metro, 4H). I-82 2-[(3R)-3-Hidroxi-1-piperidil]-4-[[5-(4-hidroxi-1-piperidil)-2-piridil]amino]-6H-1,6-naftiridin-5-on 10cm_Formic_AQ method, m/z = 437 [M + H]+, Ret. Time = 2.27 min. 1RMN-H (400 MHz, DMSO) δ 12,26 (s, 1H), 11,14 (d, J = 5,6 Hz, 1H), 8,29 (1H, s), 8,04 (d, J = 3,0 Hz, 1H), 7,92 (s, 1H), 7,44 (dd, J = 3,1, 9,0 Hz, 1H), 7,21 (dd, J = 6,0 , 7,1 Hz, 1H), 6,91-6,87 (m, 1H), 6,23-6,21 (m, 1H), 4,83–4,83 (m, 2H), 4, 32–4,26 (m, 1H), 4,12 (d, J = 12,4 Hz, 1H), 3,67–3,60 (m, 1H), 3,53–3,48 (m, 3H), 3,06– 2,98 (m, 1H), 2,89-2,78 (m, 3H), 1,94-1,75 (m, 4H), 1,57- E 1-(6-amino-pyridin-3-yl)piperidin-4-ol (R)-piperidin-3-ol (Stage 2) 1.38 (meter, 4H). (1 aquiv Formiatsalz) I-83 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(1-piperidyl)-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 421 [M + H]+, Ret. Time = 2.45 min. 1RMN-H (400 MHz, DMSO) δ 12,26 (s, 1H), 11,12 (d, J = 5,9 Hz, 1H), 8,05 (d, J = 2,9 Hz, 1H) , 7,93 (s, 1H), 7,44 (dd , J = 3,3, 9,5 Hz 1H), 7,21 (dd, J = 6,0, 6,8 Hz, 1H), 6 ,89 (d, J = 8,8 Hz, 1H), 6,23 (d, J = 7,5 Hz, 1H), 4,69 (d, J = 4,1 Hz, 1H), 3,67 –3,59 (m, 5H), 3,53–3,45 (m, 2H), 2,88–2,80 (m, 2H), 1,88–1,80 (m, 2H), 1 ,68–1,47 (M, 8H). E 1-(6-amino-pyridin-3-yl)piperidin-4-olpiperidine (paso 2) I-84 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-pyrrolidin-yl-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 407 [M + H]+, Ret. Time = 2.38 min. 1RMN-H (400 MHz, DMSO) δ 12,28 (s, 1H), 11,08 (s, 1H), 8,03 (d, J = 3,0 Hz, 1H), 7,55 (s, 1H), 7,43 (dd, J = 3,1, 9,0 Hz, 1H), 7,20 (d, J = 7,3 Hz, 1H), 6,93-6,90 (m, 1H ), 6,23 (d, J = 7,3 Hz, 1H), 4,70 (d, J = 4,3 Hz, 1H), 3,68-3,60 (m, 1H), 3,52 -3,43 (m, 6H), 2,88-2,80 (m, 2H), 1,97-1,94 (m, 4H), 1,88-1,80 (m, 2H), 1 ,56-1,46 (M, 2H). E 1-(6-amino-pyridin-3-yl)piperidin-4-ol-pyrrolidine (Stage 2) I-85 4-Fluor-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]-N-methyl - Benzamide 10cm_Formic_AQ method, m/z = 489 [M + H]+, Ret. Time = 2.36 min. 1RMN-H (400 MHz, DMSO): γ; 12,38 (s, 1 H), 11,66–11,59 (m, 1 H), 8,65 (s, 1 H), 8,51 (d, J = 4,3 Hz, 1 H) , 8,35 (dd, J = 1,9, 7,2 Hz, 1 H), 7,99 (d, J = 2,8 Hz, 1 H), 7,93-7,87 (m, 1 H), 7,43-7,33 (m, 3 H), 6,95 (d , J = 8,8 Hz, 1 H), 6,55 (d, J = 7,3 Hz, 1 H) , 4,62–4,62 (m, 1 H), 3,59–3,53 (m, 1 H), 3,49–3,40 (m, 2 H), 2,81-2,71 (m, 2 H), 2,75-2,69 (m, 3 H), 1,75 (dd, J = 8,7, 8,7 Hz, 2 H), 1,48-1,36 ( m, 2H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-(methyl-carbamoyl)phenyl)boronic acid I-86 2-(2-Fluoro-4,5-dimethyl-phenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 460 [M + H]+, Ret. Time = 2.65 min. 1RMN-H (400 MHz, DMSO): δ 12,39 (s, 1 H), 11,62 (d, J = 1,8 Hz, 1 H), 8,68 (d, J = 1,4 Hz , 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,77-7,74 (m, 1 H), 7,50-7,38 (m, 2 H), 7,14 (d, J = 12,3 Hz, 1 H), 7,04-7,00 (m, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 4, 70 (d, J = 3,5 Hz, 1 H), 3,67–3,63 (m, 1 H), 3,57–3,49 (m, 2 H), 2,91–2,83 (m, 2 h), 2,31–2,28 (m, 6 h), 1,87–1,80 (m, 2 h), 1,56–1,45 (m, 2 h), A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-(2-fluoro-4,5-dimethyl-phenyl)-4,4,5,5-tetramethyl- 1,3,2 - Dioxa-Borolan See more I-87 4-[[5-(4- Hidroxi-1-piperidil)-2- piridil]amino]-2- [(3R)-3- hidroxi-pirrolidin-1-il]-6H-1,6- naftiridin- 5 -eins Method 10cm_Formic_AQ, m/z = 423 [M + H]+, Ret. Time = 2.26 min. 1RMN H (400 MHz, DMSO) δ 12,29 (s, 1H), 11,10 (d, J = 5,5 Hz, 1H), 8,40 (s, 1H), 8,03 (d, J = 3,0 Hz, 1H), 7,54 (s, 1H), 7,44 (dd, J = 3,1, 9,0 Hz, 1H), 7,20 (dd, J = 6,0, 7,2 Hz, 1H), 6,92 (d, J = 9,0 Hz, 1H), 6,24 (d, J = 6,3 Hz, 1H), 4,92 (s, 1H), 4 ,40 (s, 1H), 3,67–3,45 (m, 8H), 2,89–2,80 (m, 2H), 2,08–1,98 (m, 1H), 1,93 –1,81 (m, 3H), 1,57-1,46 (m, 2H). (1 Äq. Formato E 1-(6-amino-pyridin-3-yl)piperidin-4-ol (R)-pyrrolidin-3-ol (stage 2) Sal) I-88 4-[[5-(4-Hydroxy-1-piperidyl)-2- pyridyl]amino]-2- [(3S)-3- hydroxypyrrolidin-1-yl]-6H-1,6- naphthiridin- 5 -einas Method 10cm_Formic_AQ, m/z = 423 [M + H]+, Ret. Time = 2.27 min. 1RMN-H (400 MHz, DMSO) δ 12,29 (s, 1H), 11,09 (d, J = 5,5 Hz, 1H), 8,29 (s, 1H), 8,03 (d, J = 3,0 Hz, 1H), 7,54 (s , 1H), 7,44 (dd, J = 3,1, 9,0 Hz, 1H), 7,20 (dd, J = 6,0 , 7,2 Hz, 1H), 6,91 (d, J = 8,9 Hz, 1H), 6,24 (dd, J = 1,4, 7,3 Hz, 1H), 4,96 (s , 1H), 4,40 (s, 1H), 3,67–3,43 (m, 8H), 2,88–2,80 (m, 2H), 2,09–2,00 (m, 1H ), 1,93 -1,81 (m, 3H), 1,57-1,46 (m, 2H). (1 Äq. Formiatsalz) E 1-(6-amino-pyridin-3-yl)piperidin-4-ol (S)-pyrrolidin-3-ol (stage 2) I-89 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-isopropyl-8-methyl-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 394 [M + H]+, Ret. Time = 2.43 min. 1RMN-H (400 MHz, DMSO) δ 12,54 (s, 1H), 11,39 (s, 1H), 8,28 (s, 1H), 8,13 (d, J = 3,0 Hz, 1H), 7,51 (dd, J = 2,8, 2,8 Hz, 1H), 7,27 (s, 1H), 7,01 (d, J = 8,8 Hz, 1H), 4, 74 (d, J = 4,0 Hz, 1H), 3,72–3,66 (m, 1H), 3,58–3,52 (m, 2H), 3,05-2,97 (m, 1H), 2,96-2,88 (m, 2H), 2,25 (s, 3H), 1,93-1,87 (m, 2H), 1,61-1,50 (m, 2H) , 1,32 (d, J = 7,1 Hz, 6H). L 1-(6-amino-pyridin-3-yl)piperidin-4-ol isopropyl-magnesiumchloreth I-90 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1,6-naphthiridin-5-one Method 10cm_Formic_AQ, m/z = 352 [M + H]+, Ret. Time = 2.24 min. 1RMN-H (400 MHz, DMSO) δ 12,62 (s, 1H), 11,51 (d, J = 4,7 Hz, 1H), 8,55 (d, J = 5,8 Hz, 1H) , 8,32 (d, J = 6,0 Hz, 1H). ), 8,12 (d, J = 2,8 Hz, 1H), 7,51 (dd, J = 2,8, 8,8 Hz, 1H), 7,30 (d, J = 4,8 Hz , 1H), 7,03 (d, J = 8,8 Hz, 1H). ), 4,77 (s, 1H), 3,72–3,65 (m, 1H), 3,59–3,51 (m, 2H), 2,94–2,87 (m, 2H), 2,24 (s, 3H), 1,87 (dd, J = 4,5). , 7,9 Hz, 2H), 1,61-1,50 (m, 2H). L 1-(6-Amino-pyridin-3-yl)piperidin-4-ol I-91 2-(2,6-Difluoro-3-methoxy-phenyl)-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 0 [M + H]+, Ret. Time = 2.2 min. 1H-NMR (400 MHz, DMSO): δ 12,59 (s, 1 H), 11,70 (brs, 1 H), 8,49 (s, 1 H), 8,08 (d, J = 2 ,5 Hz, 1 H), 7,53–7,46 (m, 2 H), 7,40-7,31 (m, 1 H), 7,23 (dd, J = 9,1, 9, 1 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 3,94 (s, 3 H) , 3,12-3,08 (m, 4 H), 3,08-3,06 (m, 1 H), 2,88 (dd, J = 4,9, 4,9 Hz, 4 H). B und G tert-butyl-4-(6-amino-pyridin-3-yl)piperazinecarboxylate-2-bromo-1,3-difluoro-4-methoxy I-92 2-(2,6-Difluor-3-methoxy-fenil)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one Method 10cm_Bicarb_AQ, m/z = 480 [M + H]+, Ret. Time = 2.84 min. 1RMN-H (400 MHz, DMSO): γ; 12,45 (s, 1 H), 11,64 (s, 1 H), 8,35 (s, 1 H), 7,97 (d, J = 2,8 Hz, 1 H), 7,41 –7,33 (m, 2 H), 7,27–7,18 (m, 1 H), 7,10 (dd, J = 9,0, 9,0 Hz, 1 H), 6,92 ( d, J = 9,1 Hz, 1 H), 6,47 (d, J = 7,3 Hz, 1 H), 4,60 (d, J = 4,0 Hz, 1 H), 3,82 (s, 3 H), 3,60–3,50 (m, 2 H), 3,15–3,08 (m, 1 H), 2,82 -2,73 (m, 2 H), 1 ,72 (d, J = 9,6 Hz, 2 H), 1,46-1,34 (m, 2 H). B 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-2-bromo-1,3-difluoro-4-methoxybenzol 1-93 2-[2-Fluoro-5-(pyrrolidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine- 5-eins Method 10cm_Formic_AQ, m/z = 529 [M + H]+, Ret. Time = 2.48 min. 1RMN-H (400 MHz, DMSO): δ 12,98–12,98 (m, 1 H), 12,32–12,31 (m, 1 H), 8,57 (s, 1 H), 8 ,09–8,01 (m, 2 H), 7,86– 7,82 (m, 1 H), 7,66 (s, 1 H), 7,52–7,43 (m, 2 H) , 7,13 (d, J = 8,9 Hz, 1 H), 6,65 (d, J = 7,2 Hz, 1 H). ), 4,29 (dd, J = 7,5, 7,5 Hz, 2 H), 4,01 (dd, J = 7,5, 7,5 Hz, 2 H), 3,60–3, 46 (m, 6 H), 2,92–2,82 (m, 2 H), 2,27-2,17 (m, 2 H), 1,79-1,70 (m, 2 H), 1,46-1,35 (m, 2H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-(pyrrolidin-1-carbonyl)phenyl)boronic acid I-94 2-[2- Fluoro-5-(piperidine- 1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H- 1,6-naphthyridine- 5-eins 10cm_Formic_AQ method, m/z = 543 [M + H]+, Ret. Time = 2.55 min. 1RMN-H (400 MHz, DMSO): δ 12,98–12,98 (m, 1 H), 12,32–12,31 (m, 1 H), 8,57 (s, 1 H), 8 ,09–8,01 (m, 2 H), 7,86– 7,82 (m, 1 H), 7,66 (s, 1 H), 7,52–7,43 (m, 2 H) , 7,13 (d, J = 8,9 Hz, 1 H), 6,65 (d, J = 7,2 Hz, 1 H). ), 4,29 (dd, J = 7,5, 7,5 Hz, 4 H), 4,01 (dd, J = 7,5, 7,5 Hz, 4 H), 3,67-3, 54 (m, 4H), 2,92-2,82 (m, 2H), 2,27–2,17 (m, 2H), 1,79–1,70 (m, 2H), 1, 46–1,35 (m, 2H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-(piperidine-1-carbonyl)phenyl)boronic acid I-95 2-[5-(Azetidine-1-carbonyl)-2-fluoro-phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine - 5-eins Method 10cm_Formic_AQ, m/z = 515 [M + H]+, Ret. Time = 2.43 min. 1RMN-H (400 MHz, DMSO): γ; 12,43 (s, 1 H), 11,60–11,60 (m, 1 H), 8,68 (s, 1 H), 7,99 (d, J = 2,5 Hz, 1 H) , 7,91 (dd, J = 1,9, 7,2 Hz, 1 H), 7,50–7,43 (m, 1 H), 7,43–7,31 (m, 3 H), 6,95 (d, J = 9,1 Hz, 1 H), 6,52 (d, J = 7,3 Hz, 1 H), 4,65–4,58 (m, 1 H), 3, 62–3,50 (m, 2 H), 3,49–3,40 (m, 3 H), 2,84–2,77 (m, 2 H), 1,76 (d, J = 10, 1 Hz, 2 H), 1,56 (s, 2 H), 1,46–1,38 (m, 4 H). Ein 1-(6-Aminopiridin-3-il)piperidin-4-ol CB15 I-96 2-(5-tert-butyl-2-fluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Bicarb_AQ, m/z = 488 [M + H]+, Ret. Time = 3.37 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,65-11,65 (m, 1 H), 8,68 (d, J = 1,8 Hz, 1 H ), 8,06 (d J = 3,0 Hz, 1 H), 7,92 (dd, J = 2,6, 7,5 Hz, 1 H), 7,58-7,53 (m, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,42 (d, J = 7,3 Hz, 1 H), 7,27 (dd, J = 8 ,7, 10,9 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 ( d , J = 4,0 Hz, 1 H), 3,68–3,61 (m, 1 H), 3,56–3,48 (m, 2 H), 2,91–2,83 (m , 2 H), 1,88–1,80 (m, 2 H), 1,55-1,45 (m, 2 H), 1,35 (s, 9 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB16 I-97 2-(5-ciclobutil-2-fluorophenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-naphthiridin-5-one Method 10cm_Bicarb_AQ, m/z = 486 [M + H]+, Ret. Time = 3.38 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,69 (s, 1 H), 8,73 (s, 1 H), 8,10 (d, J = 2 ,8 Hz, 1 H), 7,79 (dd , J = 1,9, 7,5 Hz, 1 H), 7,52 (dd, J = 3,0, 8,8 Hz, 1 H), 7,49–7,41 (m, 2 H), 7,31 (dd, J = 8,6, 10,9 Hz, 1 H). ), 7,07 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7,1 Hz, 1 H), 4,77-4,70 (m, 1 H), 3,70-3,52 (m, 4 H), 2,95- 2,86 (m, 2 H), 2,43-2,33 (m, 2 H), 2,21-1,99 ( m, 3 H), 1,93-1,83 (m, 3 H), 1,60-1,48 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB6 I-98 2-[4-Fluoro-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]fenilo ] acetonitrile 10cm_Formic_AQ method, m/z = 471 [M + H]+, Ret. Time = 2.47 min. 1RMN-H (400 MHz, DMSO): δ 12,36 (s, 1 H), 11,59 (s, 1 H), 8,66 (s, 1 H), 7,99 (d, J = 2 ,5 Hz, 1 H), 7,89 (dd , J = 1,9, 6,9 Hz, 1 H), 7,44–7,37 (m, 2 H), 7,37–7,29 (m, 2 H), 6,95 (d, J = 9,1 Hz, 1 H), 6,52 (d, J = 7,3 Hz, 1 H), 4,62 (d, J = 4 ,0 Hz, 1 H), 4,08 (s, 2 H), 3,61–3,54 (m, 1 H), 3,51–3,40 (m, 2 H), 2,84 - 2,75 (m, 2 H), 1,76 (d, J = 9,3 Hz, 2 H), 1,48-1,36 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB17 I-99 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(3-isopropylphenyl)-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 456 [M + H]+, Ret. Time = 2.73 min. 1RMN-H (400 MHz, DMSO): δ 12,54 (s, 1 H), 11,64 (d, J = 4,8 Hz, 1 H), 8,90 (s, 1 H), 8, 18 (d, J = 2,8 Hz, 1 H). ), 8,01 (s, 1 H), 7,89 (d, J = 7,3 Hz, 1 H), 7,56-7,42 (m, 4 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,66 (d, J = 7,1 Hz, 1 H), 4,80 (brs, 1 H), 3,72–3,66 (m, 1 H) , 3,62–3,53 (m, 2 H), 3,12–3,03 (m, 1 H), 2,97–2,88 (m, 2 H), 1,90 (d, J = 9,3 Hz, 2 H), 1,62-1,50 (m, 2 H), 1,33 (d, J = 6,8 Hz, 6 H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol-3-isopropyl-benzeneboronic acid I-100 2-(3,5-dimethoxyphenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 474 [M + H]+, Ret. Time = 2.58 min. 1RMN-H (400 MHz, DMSO): δ 12,40 (s, 1 H), 8,81-8,78 (m, 1 H), 8,13 (d, J = 3,0 Hz, 1 H ), 7,49 (dd, J = 2,9, 9,0 Hz, 1 H), 7,40 (d, J = 7,4 Hz, 1 H), 7,17 (dd, J = 2, 4, 2,4 Hz, 2 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,67-6,62 (m, 2 H), 3,84 (s, 3 H ), 3,67-3,63 (m, 2 H), 3,62-3,40 (m, 6 H), 2,91-2,83 (m, 2 H), 1,86-1, 83 (m, 2 H), 1,56–1,46 (m, 2 H). 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-3,5-dimethoxy-phenyl-boronic acid I-101 2-(2-Chloro-5-isopropyl-phenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-on 10cm_Formic_AQ method, m/z = 490 [M + H]+, Ret. Time = 2.75 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,69 (d, J = 5,1 Hz, 1 H), 8,49 (s, 1 H), 8, 04 (d, J = 3,0 Hz, 1 H). ), 7,51-7,35 (m, 6 H), 7,01 (d, J = 8,9 Hz, 1 H), 6,58 (d, J = 7,3 Hz, 1 H), 3,67-3,58 (m, 1 H), 3,54- 3,46 (m, 2 H), 3,04-2,92 (m, 1 H), 2,89-2,80 ( m, 2 H), 1,86-1,78 (m, 2 H), 1,53-1,42 (m, 2 H), 1,24 (d, J = 6,9 Hz, 6 H) . A 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-5-isopropyl-2-chloro-phenyl boronic I-102 2-[2- Fluor-5-(1- methylciclo-propyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- naphthyridin- 5-ein Method 10cm_Formic_AQ, m/z = 486 [M + H]+, Ret. Time = 2.92 min. 1RMN-H (400 MHz, DMSO): γ; 12,46 (s, 1 H), 11,73–11,65 (m, 1 H), 8,71 (s, 1 H), 8,10 (d, J = 2,8 Hz, 1 H) , 7,84 (dd, J = 2,3, 7,3 Hz, 1 H), 7,51 (dd, J = 3,0, 8,8 Hz, 1 H), 7,48–7,37 (m, 2 H), 7,29 (dd, J = 8,7, 10,7 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,65 (d , J = 7,1 Hz, 1 H), 4,73 (d, J = 3,0 Hz, 1 H), 3,69 (d, J = 3,3 Hz, 1 H), 3,60– 3,52 (m, 2 H), 2,96–2,88 (m, 2 H), 1,89 (d, J = 9,3 Hz, 2 H), 1,60–1,49 (m , 2 H), 1,46 (s, 3H), 0,93–0,89 (m, 2H), 0,86–0,80 (m, 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB7 I-103 2-[2-Fluoro-5-(oxetan-3-yl)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridina- 5-ein Method 10cm_Bicarb_AQ, m/z = 488 [M + H]+, Ret. Time = 2.8 min. 1RMN-H (400 MHz, DMSO): γ; 12,30 (s, 1 H), 11,51–11,50 (m, 1 H), 8,59 (s, 1 H), 7,94 (d, J = 2,5 Hz, 1 H) , 7,81 (dd, J = 2,0, 7,3 Hz, 1 H), 7,51–7,44 (m, 1 H), 7,35 (dd, J = 2,8, 8, 8 Hz, 1 H), 7,32–7,20 (m, 2 H), 6,90 (d, J = 8,8 Hz, 1 H), 6,49 (d, J = 7,3 Hz , 1 H), 4,86 ​​(dd, J = 6,1, 8,3 Hz, 2 H), 4,61–4,50 (m, 3 H), 4,29-4, 19 (m, 1 H), 3,58-3,47 (m, 1 H), 3,47-3,32 (m, 2 H), 2,75 (dd, J = 9,9, 9, 9 Hz , 2 H), 1,69 (dd, J = 8,5, 8,5 Hz, 2 H), 1,43-1,32 (m, 2 H). A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-(2-fluoro-5-(oxetan-3-yl)phenyl)-4,4,5,5-tetramethyl- 1,1. 1. 3,2-dioxa-borolene I-104 2-[4-Fluoro-3-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl]phenyl ]-2-Methyl-propane-nitrile 10cm_Formic_AQ method, m/z = 499 [M + H]+, Ret. Time = 2.58 min. 1RMN-H (400 MHz, DMSO): γ; 12,49 (s, 1 H), 11,72 (s, 1 H), 8,77 (d, J = 1,3 Hz, 1 H), 8,16–8,08 (m, 2 H) , 7,76 -7,69 (m, 1 H), 7,55-7,46 (m, 3 H), 7,07 (d, J = 8,8 Hz 1 H), 6,65 (d , J = 7,1 Hz, 1 H), 4,73 (d, J = 3,5 Hz, 1 H), 3,68 (dd, J = 4,3, 8,1 Hz, 1 H), 3,61–3,52 (m, 2 H), 2,96–2,86 (m, 2 H), 1,90–1,83 (m, 2 H ), 1,80 (s, 6 H ), 1,60-1,48 (m, 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB18 I-105 1-[4-Fluor-3-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ] cyclopropanecarbonitrila Method 10cm_Bicarb_AQ, m/z = 497 [M + H]+, Ret. Time = 2.96 min. 1RMN-H (400 MHz, DMSO): δ 12,37 (s, 1 H), 11,60 (s, 1 H), 8,64 (d, J = 1,0 Hz, 1 H), 7, 98 (d, J = 2,8 Hz, 1 H). ), 7,89 (dd, J = 2,4, 6,9 Hz, 1 H), 7,43-7,32 (m, 3 H), 7,30 (d, J = 8,8 Hz, 1 H), 6,95 (d, J = 8,8 Hz, 1 H), 6,54 (d, J = 7,3 Hz, 1 H), 4,61 (d, J = 3,5 Hz , 1 H), 3,59–3,51 (m, 1 H), 3,50–3,37 (m, 2 H), 2,84 -2,72 (m, 2 H), 1,80 -1,67 (m, 4 H), 1,52-1,39 (m, 4 H). A 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-(5-(1-cyanocyclo-propyl)-2-fluoro-phenyl)-boronic acid I-106 2-[2-Fluoro-5-(trifluoromethoxy)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-ein Method 10cm_Bicarb_AQ, m/z = 516[M+H]+, Ret. Time = 3.28 min. 1RMN-H (400 MHz, DMSO): γ; 12,40 (s, 1 H), 11,64–11,63 (m, 1 H), 8,72 (s, 1 H), 7,99 (d, J = 2,5 Hz, 1 H) , 7,89 (d, J = 3,3 Hz, 1 H), 7,49 (d, J = 3,3 Hz, 1 H), 7,47–7,33 (m, 3 H), 6 ,96 (d, J = 9,1 Hz, 1 H), 6,53 (d, J = 7,3 Hz, 1 H), 4,66–4,62 (m, 1 H), 3,55 (d, J = 3,3 Hz, 1 H), 3,50–3,37 (m, 2 H), 2,83–2,74 (m, 2 H), 1,77 (d, J = 9,6 Hz, 2 H), 1,48–1,36 (m, 2 H). 1-(6-Amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-5-(trifluoromethoxy)phenyl)boronic acid I-107 2-(6-Fluoro-1H-indol-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 471 [M + H]+, Ret. Time = 2.48 min. 1RMN-H (400 MHz, DMSO): δ 12,37 (s, 1 H), 11,58 (d, J = 3,9 Hz, 1 H), 11,29 (s, 1 H), 8, 70 (d, J = 1,9 Hz, 1 H). ), 8,16 (d, J = 7,8 Hz, 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,48 (dd, J = 3,1, 9, 0 Hz, 1 H), 7,43-7,38 (m, 2 H), 7,30 (d, J = 8,9 Hz, 1 H), 7,03 (d, J = 8,9 Hz , 1 H), 6,62–6,54 (m, 2 H), 4,70 (d, J = 4,1 Hz, 1 H). ), 2,91-2,83 (m, 2 H), 1,87-1,81 (m, 5 H), 1,56-1,45 (m, 2 H). A and G 1-(6-Aminopyridin-3-yl)piperidin-4-ol-tert-butyl-6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolane -2-yl)-1H-indole-1-carboxylate I-108 N-[3-Fluor-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ]-N-methyl-cyclopropane-carboxamide Method 10cm_Formic_AQ, m/z = 529 [M + H]+, Ret. Time = 2.51 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,68-11,68 (m, 1 H), 8,75 (d, J = 1,6 Hz, 1 H ), 8,08-8,02 (m, 2 H), 7,52-7,47 (m, 2 H), 7,46-7,37 (m, 2 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 3,67-3,45 (m, 2 H), 3,28 (s, 3 H ), 2,92-2,84 (m, 2 H), 2,54 (s, 1 H), 1,88-1,80 (m, 2 H), 1,58-1,45 (m, 4 H), 0,88–0,83 (m, 2H), 0,75–0,69 (m, 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB2 I-109 2-(2-Fluoro-5-isopropyl-phenyl)-4-[[5-(4-methyl-piperazin-1-yl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 473 [M + H]+, Ret. Time = 2.37 min. 1RMN-H (400 MHz, DMSO): δ 12,40 (s, 1 H), 8,68 (s, 1 H), 8,05 (d, J = 2,6 Hz, 1 H), 7, 76 (d, J = 7,7 Hz, 1 H). ), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,43–7,37 (m, 2 H), 7,29–7,23 (m, 1 H) , 7,04 (d, J = 8,9 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 3,46–3,42 (m, 1 H), 3 ,15 (dd, J = 4,7, 4,7 Hz, 4 H), 3,04–2,96 (m, 1 H), 2,47 (dd, J = 4,9, 4,9 Hz , 4 H), 2,23 (s, 3 H), 1,25 (d, J = 6,9 Hz, 6 H). 5-(4-Methyl-piperazin-1-yl)-pyridin-2-amino-2-fluoro-5-isopropyl-phenyl-boronic acid I-110 2-(2-Fluoro-5-isopropyl-phenyl)-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 459 [M + H]+, Ret. Time = 2.34 min. 1RMN-H (400 MHz, DMSO): δ 12,37 (s, 1 H), 8,61 (s, 1 H), 7,95 (d, J = 2,5 Hz, 1 H), 7, 70 (dd, J = 2,1, 7,5 Hz, 1 H), 7,41-7,30 (m, 3 H), 7,22-7,14 (m, 1 H), 6,95 (d, J = 8,8 Hz, 1 H), 6,53 (d, J = 7,3 Hz, 1 H), 3,01-2,95 (m, 6 H), 2,94-2 ,89 (m, 1 H), 2,76 (dd, J = 4,5, 4,5 Hz, 4 H), 1,17 (d, J = 6,8 Hz, 6 H). A and G tert-butyl 4-(6-amino-pyridin-3-yl)-piperazin-1-carboxylate 2-fluoro-5-isopropyl-phenyl-boronic I-111 N-[3-Fluoro-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ] cyclohexanocarboxamide BicarbB EHC18 method, m/z = 557.3 [M+H]+, Ret. tempo = 4.09 min. 1H-RMN (400 MHz, DMSO): δ 12,38 (s, 1H), 11,61 (d, J = 1,5 Hz, 1H), 10,22 (s, 1H), 8,73 (d , J = 1,0 Hz, 1H), 8,09- 7,98 (m, 2H), 7,76 (dd, J = 1,9, 14,3 Hz, 1H), 7,50-7, 39 (m, 3H), 7,04–7,01 (m, 1H), 6,59–6,56 (m, 1H), 4,70 ( s, 1H), 3,69-3,61 ( m, 1H), 3,56-3,50 (m, 2H), 2,92-2,84 (m, 4H), 2,41-2,33 (m, 1H), 1,87-1, 75 (m, 4H), 1,56-1,19 (m, 8H). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB19 I-112 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-uno Method 10cm_Formic_AQ, m/z = 573 [M + H]+, Ret. Time = 2.6 min. 1RMN-H (400 MHz, DMSO) δ 12,47 (s, 1 H), 11,68-11,65 (m, 1 H), 8,74 (d, J = 1,5 Hz, 1 H) , 8,08 (d, J = 2,9 Hz, 1 H), 7,58 (d, J = 6,0 Hz, 1 H), 7,50–7,42 (m, 2 H), 7 ,24 (d, J = 10,4 Hz, 1 H), 7,05–7,02 (m, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,3 Hz, 1 H), 3,87–3,86 (m, 3 H), 3,68–3,50 (m, 5 H), 3,20–3,14 ( m , 2 std.), 2,91–2,84 (m, 2 std.), 1,88–1,81 (m, 2 std.), 1,64–1,45 (m, 8 std. ). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB1 I-113 2-[2-Fluoro-3-Methyl-4-(Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on Method 10cm_Bicarb_AQ, m/z = 557 [M+H]+, Ret. Time = 2.95 min. 1RMN-H (400 MHz, DMSO): γ; 12,28 (s, 1 H), 11,51–11,49 (m, 1 H), 8,52 (s, 1 H), 7,92 (d, J = 2,8 Hz, 1 H) , 7,62 (dd, J = 7,6, 7,6 Hz, 1 H), 7,34–7,24 (m, 2 H), 6,99 (d, J = 7,8 Hz, 1 H), 6,87 (d, J = 8,8 Hz, 1 H), 6,43-6,37 (m, 1 H), 4,53 (d, J = 4,3 Hz, 1 H) , 3,52-3,46 (m, 3 H), 3,38-3,35 (m, 2 H), 3,06-3,00 (m, 2 H), 2,74–2,67 (m, 2 H), 2,06 (d, J = 1,5 Hz, 3 H), 1,69–1,65 (m, 2 H), 1,48–1,43 (m, 4 H ), 1,33 (dd, J = 10,0, 13,0 Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB20 Hercios, 4H). I-114 2-[2-Fluoro-5-Methyl-4-(Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on Method 10cm_Formic_AQ, m/z = 557 [M + H]+, Ret. Time = 2.59 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 8,76-8,72 (m, 1 H), 8,11 (d, J = 2,8 Hz, 1 H ), 7,87 (d, J = 7,6 Hz, 1 H), 7,52 (dd, J = 2,8, 2,8 Hz, 1 H), 7,46 (d, J = 7, 3 Hz, 1 H), 7,26 (s, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7,3 Hz, 1 H) , 3,73–3,63 (m, 3 H), 3,57 (d, J = 12,4 Hz, 3 H), 3,02 (dd, J = 5,3, 5,3 Hz, 2 H), 2,96-2,87 (m, 2 H), 2,60 (s, 1 H), 2,30 (s, 3 H), 1,91-1,86 (m, 2 H) , 1,67-1,63 (m, 4 H), 1,59-1,48 (m, 4 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB21 I-115 2-[2-Fluoro-4-(pyrrolidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine- 5-eins Method 10cm_Formic_AQ, m/z = 529 [M + H]+, Ret. Time = 2.46 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,73 (s, 1 H), 8,79 (s, 1 H), 8,13–8,04 (m , 2 H), 7,57–7,50 (m, 3 H), 7,47 (d, J = 7,1 Hz, 1 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 1,3 Hz, 1 H), 3,68 (d, J = 1,3 Hz, 1 H), 3,61–3,46 (m, 6 H), 2,95–2,87 (m, 2 H), 1,98–1,86 (m, 6 H), 1,60 - 1,49 (m, 2 Est.). F 1-(6-Aminopyridin-3-yl)piperidin-4-ol I-116 2-[2-Fluoro-5-(1-hydroxy-1-methylethyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6- Naphthyridin-5-one 10cm_Formic_AQ method, m/z = 490 [M + H]+, Ret. Time = 2.48 min. 1RMN H (400 MHz, DMSO): δ 12,33 (s, 1 H), 11,56 (d, J = 4,5 Hz 1 H), 8,59 (s, 1 H), 7,96 (dd, J = 2,7, 10,5 Hz, 2 H), 7,52–7,45 (m, 1 H), 7,40 (dd, J = 2,9, 9,0 Hz, 1 H), 7,33 (dd, J = 6,3, 6,3 Hz, 1 H), 7,18 (dd, J = 8,8, 10,9 Hz, 1 H), 6,94 (d , J = 8,8 Hz, 1 H), 6,52 (d, J = 7,3 Hz, 1 H), 5,09 (s, 1 H), 4,61-4,58 (m, 1 H ), 3,60–3,54 (m, 1 H), 3,25 (s, 2 H), 2,82–2,72 (m, 2 H), 1,77 (d, J = 9 ,6 Hz, 2 H), 1,51–1,40 (m, 8 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB22 I-117 2-[5- [(Dimetilamino)metil]-2-fluorophenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- Naftiridina-5- sober 10cm_Formic_AQ method, m/z = 489 [M + H]+, Ret. Time = 2.21 min. 1RMN-H (400 MHz, DMSO): γ; 12,46 (s, 1 H), 11,69 (s, 1 H), 8,75 (s, 1 H), 8,10 (d, J = 2,5 Hz, 1 H), 7,93 –7,89 (m, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,46 (d, J = 7,3 Hz, 2 H), 7 ,37-7,30 (m, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 3,73-3,65 (m, 1 H), 3,61-3,52 (m, 2 H), 3,49 (s, 2 H ), 2,94-2,86 (m, 2 H), 2,22 (s, 6 H), 1,88 (d, J = 9,3 Hz, 2 H), 1,60-1,49 (m, 2H). A 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 1-(4-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2 ). ). - yl)phenyl)-N,N-dimethylmethanamine I-118 2-(5-Fluoro-1H-indol-6-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 471 [M + H]+, Ret. Time = 2.56 min. 1RMN-H (400 MHz, DMSO): γ; 12,37 (s, 1 H), 11,59–11,59 (m, 1 H), 11,36 (s, 1 H), 8,76 (d, J = 1,6 Hz, 1 H) , 8,10 -8,07 (m, 2 H), 7,56 (dd, J = 2,8, 2,8 Hz, 1 H), 7,51-7,45 (m, 2 H), 7,43-7,39 (m, 1 H), 7,04 (d, J = 8,8 Hz, 1 H), 6,60 (d, J = 7,2 Hz, 1 H), 6, 50 (dd, J = 2,5, 2,5 Hz, 1 H), 4,70 (d, J = 4,3 Hz, 1 H), 3,67- 3,61 (m, 1 H), 3,57-3,50 (m, 2 H), 2,92-2,84 (m, 2 H), 1,87- A and G 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB23 1,81 (metro, 2H), 1,56-1,46 (M, 2H). I-119 2-[2- Fluoro-4-(piperazine- 1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H- 1,6-naphthyridine- 5-eins 10cm_Formic_AQ method, m/z = 544 [M + H]+, Ret. Time = 2.18 min. 1RMN-H (400 MHz, DMSO): γ; 12,51 (s, 1 H), 11,72–11,68 (m, 1 H), 8,79 (s, 1 H), 8,12 (d, J = 2,5 Hz, 1 H) , 8,06 (dd, J = 7,7, 7,7 Hz, 1 H), 7,53 (dd, J = 2,7, 9,0 Hz, 1 H), 7,50–7,44 (m, 1 H), 7,43–7,38 (m, 2 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7,3 Hz , 1 H), 4,73 (d, J = 2,0 Hz, 1 H), 3,68 (s, 1 H), 3,56 (dd, J = 4,7, 8,0 Hz, 4 H), 3,34 (s, 3 H), 2,92 (dd, J = 10,0, 10,0 Hz, 2 H), 2,80 (s, 2 H), 2,72 (s, 2H), 1,89-1,86 (m, 2H), 1,59-1,50 (m, 2H). F and G 1-(6-amino-pyridin-3-yl)piperidin-4-ol tert-butylpiperazine-1-carboxylate (Stage 2) I-120 2-[2-Fluoro-4-(morpholine-4-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino-6H-1,6-naphthyridine-5 -eins Método 10cm_Formic_ACE-AR_AQ, m/z = 545 [M + H]+, Ret. Zeit = 2,53 min. 1RMN-H (400 MHz, DMSO): δ 12,09 (s, 1 H), 11,72 (s, 1 H), 8,78 (s, 1 H), 8,13–8,04 (m , 2 H), 7,55–7,43 (m, 4 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,75–4,74 (ss, 1 H), 3,77–3,61 (m, 8 H), 3,60-3,53 (m, 3 H), 2,96-2 ,87 (m, 2 H), 1,89-1,86 (m, 2 H), 1,60-1,48 (m, 2 H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol morpholine (stage 2) I-121 2-(2-Fluoro-5-isopropyl-phenyl)-4-[[5-(3-oxa-9-azaspiro[5.5]undecan-9-yl)-2-pyridyl]amino]-6H-1,6 - Naphthyridine-5-one Method 10cm_Formic_AQ, m/z = 528 [M + H]+, Ret. Time = 3.06 min. 1RMN-H (400 MHz, DMSO): δ 12,28 (s, 1 H), 8,56 (s, 1 H), 7,98 (d, J = 2,5 Hz, 1 H), 7, 68 (d, J = 7,1 Hz, 1 H). ), 7,39 (dd, J = 2,9, 9,0 Hz, 1 H), 7,32 (dd, J = 7,3, 7,3 Hz, 2 H), 7,17 (dd, J = 8,6, 10,9 Hz, 1 H), 6,95 (d , J = 8,8 Hz, 1 H), 6,54 (d, J = 7,3 Hz, 1 H), 3 ,49 (dd, J = 5,1, 5,1 Hz, 4 H), 3,36-3,32 (m, 1 H), 3,07 (dd, J = 5,4, 5,4 Hz , 4 H), 2,95-2,86 (m, 1 H), 1,53 (dd, J = 5,2, 5,2 Hz, 4 H), 1,38 (dd, J = 4, 9, 4,9 Hz, 4 H), 1,16 (d, J = 6,8 Hz, 6 H). 5-(3-oxa-9-azaspiro[5.5]undecan-9-yl)pyridin-2-amine-2-fluoro-5-isopropyl-phenylboronic acid I-122 4-[[5-[(3R,5R)-3,5-Dimetilmorpholin-4-il]-2-Piridil]Amino]-2-(2-Fluor-5-Isopropil-Fenil)-6H-1,6 -Naftiridina-5-on 10cm_Formic_AQ method, m/z = 488 [M + H]+, Ret. Time = 3.01 min. 1RMN-H (400 MHz, DMSO): δ 12,63 (s, 1 H), 8,90 (s, 1 H), 8,12 (d, J = 2,5 Hz, 1 H), 7, 81 (dd, J = 1,9, 7,7 Hz, 1 H), 7,56 (dd, J = 2,5, 8,8 Hz, 1 H), 7,49–7,44 (m, 2 H), 7,35–7,28 (m, 1 H), 7,12 (d, J = 8,8 Hz, 1 H). ), 6,70 (d, J = 7,3 Hz, 1 H), 3,87 (dd, J = 2,4, 10,7 Hz, 2 H), 3,48-3,41 (m, 4 H), 3,08-2,99 (m, 1 H), 2,39 (s, 1 H), 1,28 (d, J = 7,1 Hz, 6 H), 0,89 (d , J = 6,3 Hz, 6 H). A 5-((3R,5R)-3,5-Dimetilmorfolino)-Piridin-2-Amin-2-Fluor-5-Isopropil-Fenil-Boronsäure I-123 2-[2-Fluoro-5-(2,2,2-Trifluoro-1-hydroxy-1-methylethyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl] amino ]-6H-1,6-naphthyridin-5-one 10cm_Formic_AQ method, m/z = 544 [M + H]+, Ret. Time = 2.59 min. 1RMN-H (400 MHz, DMSO): δ 12,35 (s, 1 H), 11,58 (d, J = 5,3 Hz, 1 H), 8,64 (s, 1 H), 8, 12 (d, J = 6,3 Hz, 1 H). ), 7,98 (d, J = 2,3 Hz, 1 H), 7,63 (d, J = 3,8 Hz, 1 H), 7,43-7,27 (m, 3 H), 6,95 (d, J = 8,8 Hz, 1 H), 6,71 (s, 1 H), 6,53 (d, J = 7,3 Hz, 1 H), 4,61 (d, J = 4,0 Hz, 1 H), 3,61-3,51 (m, 1 H), 3,25 (s, 2 H), 2,79 (dd, J = 10,1, 10,1 Hz, 2 H), 1,76 (d, J = 9,6 Hz, 2 H), 1,67 (s, 3 H), 1,46–1,36 (m, 2 H). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB8 I-124 2-[4-(3-Azabicyclo[2.2.2]octane-3-carbonyl)-2-fluorophenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H -1,6-Nafthyridine-5-on Method 10cm_Formic_AQ, m/z = 569 [M + H]+, Ret. Time = 2.62 min. 1RMN-H (400 MHz, DMSO): δ 12,37 (s, 1 H), 11,60 (s, 1 H), 8,66 (s, 1 H), 8,00–7,97 (m , 1 H), 7,93 (dd, J = 8,0, 8,0 Hz, 1 H), 7,43-7,32 (m, 2 H), 7,26 (d, J = 7, 8 Hz, 1 H), 6,95 (d, J = 8,8 Hz, 1 H), 6,54-6,48 (m , 1 H), 3,59-3,53 (m, 1 H ), 3,31-3,24 (m, 1 H), 2,79 (dd, J = 9,9, 9,9 Hz, 2 H), 2,43 (s, 6 H), 1,97 - 1,90 (m, 1 H), 1,76–1,70 (m, 4 H), 1,63–1,49 (m, 6 H), 1,42 (q, J = 9,2 Hz, 2H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-azabicyclo[2.2.2]octane (Schritt 2) . I-125 Methyl-3-fluor-4-[4-[[5-(4-hidroxi-1-piperidil)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]benzoato 10cm_Formic_AQ method, m/z = 490 [M + H]+, Ret. Time = 2.55 min. 1H-NMR (400 MHz, DMSO): δ 12,51 (s, 1 H), 11,76-11,71 (m, 1 H), 8,81 (s, 1 H), 8,71 (dd , J = 8,0, 8,0 Hz, 1 H), 8,11 (d, J = 2,3 Hz, 1 H), 7,98 (d, J = 8,1 Hz, 1 H), 7,87 (s, 1 H), 7,55-7,45 (m, 2 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7 ,3 Hz, 1 H), 4,74 (d, J = 3,8 Hz, 1 H), 3,97 (s, 3 H), 3,72-3,66 (m, 1 H), 3 ,57 (d, J = 12,1 Hz, 2 H), 2,92 (dd, J = 10,1, 10,1 Hz, 2 H), 1,87 (d, J = 10,1 Hz, 2 H), 1,60–1,49 (m, 2 H). A 1-(6-Amino-pyridin-3-yl)piperidin-4-olmethyl-3-fluor-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl ) benzoate I-126 2-[4-(Azetidine-1-carbonyl)-2-fluoro-phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine - 5-eins 10cm_FormicAQ method, m/z = 515 [M+H]+, Ret. Time = 2.41 min. 1RMN-H (400 MHz, DMSO): δ 12,38 (s, 1 H), 11,60 (s, 1 H), 8,66 (s, 1 H), 8,01–7,93 (m , 2 H), 7,54–7,47 (m, 1 H), 7,40 (dd, J = 2,9, 9,0 Hz, 1 H), 7,39–7,36 (m, 1 H), 7,35 (d, J = 7,3 Hz, 1 H), 6,95 (d, J = 8,8 Hz). , 1 H), 6,51 (d, J = 7,1 Hz, 1 H), 4,61 (d, J = 4,0 Hz, 1 H), 4,31 (dd, J = 7,5 , 7,5 Hz, 2 H), 4,06-3,98 (m, 2 H), 3,60-3,53 (m, 1 H), 3,52-3,46 (m, 2 H ), 2,83-2,75 (m, 2 H), 2,28-2,18 (m, 2 H), 1,78-1,75 (m, 2 H), 1,48–1, 36 (m, 2 H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol azetidine (Stage 2) I-127 2-[2- Fluor-4-(1-piperidyl-methylethyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2-pyridyl]amino]-6H- 1,6- naphthyridina- 5-ein 10cm_Formic_AQ Method, m/z = 265.347 [M + H]+, Ret. Time = 2.23 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,66 (d, J = 4,8 Hz, 1 H), 8,74 (s, 1 H), 8, 11 (d, J = 2,8 Hz, 1 H). ), 7,96 (dd, J = 8,0, 8,0 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,48–7, 42 (m, 1 H), 7,32 (d, J = 8,3 Hz). , 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 6,6 Hz, 1 H), 4,73 (d, J = 4,0 Hz, 1 H), 3,73-3,65 (m, 1 H), 3,61-3,52 (m, 4 H), 2,96-2,87 (m, 2 H), 2, 42 (s, 4 H), 1,89-1,84 (m, 2 H), 1,57-1,46 (m, 9 H ). A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-(3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2) ).-il)selfish)piperidina I-128 N-ethyl-2-[6-[[2-(2-fluoro-5-isopropyl-phenyl)-5-oxo-6H-1,6-naphthyridin-4-yl]amino]-3-pyridyl]-2 -Methylpropanamide 10cm_Formic_AQ method, m/z = 488 [M + H]+, Ret. Time = 2.9 min. 1RMN-H (400 MHz, DMSO): δ 12,75 (s, 1 H), 11,74 (s, 1 H), 9,05 (d, J = 1,6 Hz, 1 H), 8, 32 (d, J = 2,5 Hz, 1 H). ), 7,81 (dd, J = 2,4, 7,5 Hz, 1 H), 7,68 (dd, J = 2,6, 8,5 Hz, 1 H), 7,48-7, 41 (m, 3 H), 7,30 (dd, J = 8,4, 11,0 Hz, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,66 ( d, J = 7,3 Hz, 1 H), 3,11–2,99 (m, 3 H), 1,48 (s, 6 H), 1,26 (d, J = 6,9 Hz, 6 H), 0,97 (dd, J = 7,2, 7,2 Hz, 3 H). A CA1 2-Fluoro- 5- Isopropyl- Phenyl-Boron I-129 4-[[5-[(3S,5S)-3,5-Dimethylmorpholin-4-yl]-2-Pyridyl]Amino]-2-(2-Fluor-5-Isopropyl-Phenyl)-6H-1,6 -Naphthiridin-5-one 10cm_Formic_AQ method, m/z = 488 [M + H]+, Ret. Time = 3.03 min. 1RMN-H (400 MHz, DMSO): δ 12,65 (s, 1 H), 11,70 (s, 1 H), 8,87 (d, J = 1,6 Hz, 1 H), 8, 10 (d, J = 2,8 Hz, 1 H). ), 7,79 (dd, J = 2,4, 7,5 Hz, 1 H), 7,53 (dd, J = 2,7, 8,7 Hz, 1 H), 7,46–7, 39 (m, 2 H), 7,28 (dd, J = 8,5, 11,0 Hz, 1 H), 7,08 (d, J = 8,7 Hz, 1 H), 6,64 ( d, J = 7,3 Hz, 1 H), 3,84 (dd, J = 3,1, 10,7 Hz, 2 H), 3,53- 3,39 (m, 4 H), 3, 05–2,97 (m, 1 H), 1,26 (d, J = 6,9 Hz, 6 H), 0,87 (d, J = 6,3 Hz, 6 H). A 5-((3S,5S)-3,5-Dimethylmorpholino)-Pyridine-2-Amine-2-Fluoro-5-Isopropyl-Phenyl-Boronsäure I-130 2-[2-Fluoro-4-(2-Methyl-Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on Method 10cm_Formic_AQ, m/z = 557 [M + H]+, Ret. Time = 2.62 min. 1RMN-H (400 MHz, DMSO): γ; 12,51 (s, 1 H), 11,75–11,70 (m, 1 H), 8,78 (s, 1 H), 8,12 (d, J = 2,8 Hz, 1 H) , 8,06 (dd, J = 7,8, 7,8 Hz, 1 H), 7,52 (dd, J = 2,8, 2,8 Hz, 1 H), 7,47 (d, J = 7,3 Hz, 1 H), 7,41–7,34 (m, 2 H ), 7,07 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7 ,3 Hz, 1 H), 4,72 (d, J = 1,3 Hz, 1 H), 3,68 (s, 1 H), 3,61 -3,54 (m, 2 H), 3 ,12-3,08 (m, 2 H), 2,14 (s, 1 H), 1,90-1,84 (m, 2 H), 1,71-1,43 (m, 9 H) , 1,26 (d , J = 6,8 Hz, 3 H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-metil-piperidina (stage 2) I-131 2-[2-Fluoro-4-(4-Methyl-Piperazine-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on 10cm_Formic_AQ method, m/z = 558 [M + H]+, Ret. Time = 2.17 min. 1RMN-H (400 MHz, DMSO): γ; 12,49 (s, 1 H), 11,71 (d, J = 5,3 Hz, 1 H), 8,78 (s, 1 H), 8,20 (s, 2 H), 8,13 –8,03 (m, 2 H), 7,55-7,44 (m, 3 H), 7,44-7,38 (m, 2 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 3,61-3,55 (m, 3 H), 3,43-3,42 (m, 2 H) , 2,96-2,87 (m, 2 H), 2,43 (s, 2 H), 2,38 (s, 2 H), 2,27 (s, 3 H), 1,90-1 ,83 (m, 2H), 1,60-1,48 (m, 2H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-methyl-piperazine (step 2) I-132 1-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]benzoyl ] Piperidin-4-carbonitrile Method 10cm_Formic_AQ, m/z = 568 [M + H]+, Ret. Time = 2.45 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,70 (s, 1 H), 8,78 (s, 1 H), 8,13–8,04 (m , 2 H), 7,55–7,39 (m, 4 H), 7,44 (d, J = 8,1 Hz, 1 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 3,98-3,98 (m, 1 H), 3,72-3,66 (m, 2 H), 3, 62-3,55 (m, 2 H), 3,26-3,19 (m, 2 H), 2,97-2,87 (m, 2 H), 2,04-1,99 (m, 2 H), 1,92–1,86 (m, 5 H), 1,60–1,49 (m, 2 H). F 1-(6-amino-pyridin-3-yl) piperidin-4-ol piperidin-4-carbonitrile (Step 2) I-133 2-[2-Fluoro-4-(3-methoxy-piperidin-1-carbonyl)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 573 [M + H]+, Ret. Time = 2.49 min. 1RMN-H (400 MHz, DMSO): γ; 12,49 (s, 1 H), 11,71 (d, J = 4,3 Hz, 1 H), 8,79 (s, 1 H), 8,14–8,05 (m, 2 H) , 7,55 -7,45 (m, 1 H), 7,43 (s, 6 H), 7,40 (s, 1 H), 7,08 (d, J = 8,8 Hz, 1 H ), 6,63 (d, J = 7,1 Hz, 1 H), 3,71-3,66 (m, 1 H), 3,57 (d, J = 12,6 Hz, 1 H), 3,56-3,20 (m, 4 H), 3,17 (s, 1 H), 2,96-2,87 (m, 2 H), 1,89-1,83 (m, 3 H ), 1,76 (s, 2 H), 1,60-1,49 (m, 4 H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 3-methoxy-piperidina (Schritt 2) I-134 3-Fluoro-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naftiridin-2-yl]benzohexauro 10cm_Formic_AQ method, m/z = 476 [M + H]+, Ret. Time = 2.38 min. 1RMN-H (400 MHz, DMSO): γ; 13,47–13,46 (m, 1 H), 12,50 (s, 1 H), 11,72 (d, J = 5,3 Hz, 1 H), 8,80 (s, 1 H) , 8,17 -8,09 (m, 2 H), 7,95 (dd, J = 1,3, 8,1 Hz, 1 H), 7,86 (s, 1 H), 7,55- 7,44 (m, 2 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,65 (d, J = 6,6 Hz, 1 H), 4,73–4, 67 (m, 1 H), 3,73–3,66 (m, 1 H), 3,61–3,53 (m, 2 H), 2,95– 2,87 (m, 2 H), 1,89 (dd, J = 3,5, 8,8 Hz, 2 H), 1,60–1,49 (m, 2 H). F 1-(6-aminopyridin-3-yl)piperidin-4-ol I-135 2-[2-Fluoro-4-(3-Methyl-Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on 10cm_Formic_AQ method, m/z = 577 [M + H]+, Ret. Time = 2.64 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,73 (d, J = 2,8 Hz, 1 H), 8,79 (s, 1 H), 8, 23 (s, 1 H), 8,12 (d , J = 2,8 Hz, 1 H), 8,07 (dd, J = 8,0, 8,0 Hz, 1 H), 7,55- 7,45 (m, 2 H), 7,43 (s, 1 H), 7,42-7,37 (m, 1 H), 7,07 (d, J = 8,8 Hz, 1 H) , 6,63 (d, J = 7,3 Hz, 1 H), 4,69–4,68 (m, 1 H), 4,35 (s, 1 H), 3,72–3,65 ( m, 1 H), 3,12-3,04 (m, 1 H), 2,95-2,87 (m, 3 H), 2,82-2,80 (m, 1 H), 1, 92-1,86 (m, 2 H), 1,66 F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 3-metil-piperidina (step 2) (s, 1H), 1,60-1,48 (m, 2 H), 1,29-1,19 (m, 2 H), 1.02-0.95 (s, 2H), 0.93- 0.89 (m, 2H), 0.88-0.83 (M, 2H). I-136 2-[2-Fluoro-4-(4-methoxy-piperidin-1-carbonyl)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-uno Method 10cm_Formic_AQ, m/z = 573 [M + H]+, Ret. Time = 2.47 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,72 (s, 1 H), 8,79 (s, 1 H), 8,12 (d, J = 2 ,8 Hz, 1 H), 8,07 (dd , J = 8,0, 8,0 Hz, 1 H), 7,55–7,45 (m, 2 H), 7,45–7,38 (m, 1 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,74–4,74 (m , 1 H), 3,98–3,94 (m, 1 H), 3,71–3,65 (m, 1 H), 3,61–3,51 (m, 4 H), 3,50 – 3,39 (m, 4 H), 3,29 (s, 1 H), 3,22 (s, 1 H), 2,96-2,87 (m, 2 H), 1,88- F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 4-methoxy-piperidina (Schritt 2) 1,87 (m, 4 H), 1,58-1,48 (M, 4H). I-137 2-[4-(3-Azabicyclo[2.2.1]Heptan-3-Carbonyl)2-Fluoro-Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]- 6H-1,6-Nafthyridine-5-on Method 10cm_Bicarb_AQ, m/z = 555 [M + H]+, Ret. Time = 2.9 min. 1RMN-H (400 MHz, DMSO): γ; 12,49 (s, 1 H), 11,71–11,71 (m, 1 H), 8,79 (d, J = 6,8 Hz, 1 H), 8,22 (s, 1 H) , 8,14 -8,07 (m, 2 H), 7,56-7,50 (m, 2 H), 7,50-7,43 (m, 2 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz). , 1 H), 4,59 (s, 1 H), 4,13 (s, 1 H), 3,72-3,65 (m, 1 H), 3,52- 3,41 (m, 1 H), 3,18-3,06 (m, 2 H), 2,95-2,89 (m, 2 H), 2,68 (s, 1 H), 1,89-1,66 (m , 6 H), 1,58-1,42 (m, 4 H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-azabicyclo[2.2.1]heptane (Schritt 2) I-138 2-[4-(6,6-Difluoro-3-azabicyclo[3.1.0]hexane-3-carbonyl)-2-Fluoro-phenyl]-4-[[5-(4-hydroxy-1-piperidyl)- 2-Pyridyl]amino]-6H-1,6-naphthyridin-5-on 10cm_Formic_AQ method, m/z = 577 [M + H]+, Ret. Time = 2.53 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,73 (s, 1 H), 8,79 (s, 1 H), 8,13–8,04 (m , 2 H), 7,56–7,47 (m, 3 H), 7,38-7,33 (m, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,73-4,73 (m, 1 H), 4,18 (dd, J = 12,5, 12,5 Hz, 1 H ), 3,83–3,66 (m, 2 H), 3,61–3,53 (m, 2 H), 3,22 (s, 2 H), 2,96–2,89 (m, 2 H), 2,70–2,59 (m, 2 H), 1,90–1,84 (m, 2 H), 1,60–1,49 (m, 2 H). F 1-(6- Amino-pyridin-3-yl)piperidin- 4-ol 6,6- Difluor-3-azabicyclo[3.1.0]hexane I-139 N-cyclohexyl-3-fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl] benzamide Method 10cm_Formic_AQ, m/z = 557 [M + H]+, Ret. Time = 2.7 min. 1RMN-H (400 MHz, DMSO): γ; 12,48 (s, 1 H), 11,70 (s, 1 H), 8,79 (s, 1 H), 8,43 (d, J = 7,8 Hz, 1 H), 8,12 –8,05 (m, 2 H), 7,90–7,84 (m, 1 H), 7,52 (dd, J = 3,0, 8,8 Hz, 1 H), 7,48– 7,47 (m, 1 H), 7,08 (d, J = 8,8 Hz). , 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,74–4,72 (m, 1 H), 3,88–3,81 (m, 1 H), 3,71–3,66 (m, 1 H), 3,59– 3,52 (m, 2 H), 2,97-2,87 (m, 2 H), 1,94-1,87 ( m, 4 H), 1,82 (s, 2 H), 1,69-1,66 (m, 2 H), F 1-(6-amino-pyridin-3-yl)piperidin-4-ol cyclohexyl-amino (Stage 2) 1.59-1.49 (m, 2H), 1.41- 1,36 (m, 4H), 1,22-1,18 (M, 1H). I-140 2-[2-Fluoro-4-(4-hydroxy-piperidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-on 10cm_Formic_AQ method, m/z = 559 [M + H]+, Ret. Time = 2.34 min. 1RMN-H (400 MHz, DMSO): γ; 12,49 (s, 1 H), 11,72–11,68 (m, 1 H), 8,79 (s, 1 H), 8,12 (d, J = 2,8 Hz, 1 H) , 8,07 (dd, J = 7,8, 7,8 Hz, 1 H), 7,52 (dd, J = 2,9, 8,7 Hz, 1 H), 7,49–7,44 (m, 2 H), 7,43–7,38 (m, 2 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,63 (d, J = 7,3 Hz , 1 H), 4,04–4,02 (m, 1 H), 3,85–3,78 (m, 1 H), 3,72–3,66 (m , 1 H), 3,62 -3,55 (m, 3H), 3,22 (s, 2H), 2,95-2,87 (m, 2H), F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 4-hydroxy-piperidine (Step 2) . 1,89-1,82 (m, 4 H), 1,59- 1.51 (m, 3H), 1.42-1.41 (M, 2H). I-141 2-[2-Fluoro-4-[2-(hydroxymethyl)piperidine-1-carbonyl]phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1 ,6-naphthyridine-5-on Method 10cm_Formic_AQ, m/z = 573 [M + H]+, Ret. Time = 2.45 min. 1RMN-H (400 MHz, DMSO): γ; 12,44 (s, 1 H), 11,67–11,64 (m, 1 H), 8,73 (s, 1 H), 8,07 (d, J = 2,8 Hz, 1 H) , 7,99 (dd, J = 8,0, 8,0 Hz, 1 H), 7,47 (dd, J = 2,8, 8,8 Hz, 1 H), 7,42 (d, J = 5,6 Hz, 1 H), 7,35 (d, J = 7,6 Hz). , 2 H), 7,02 (d, J = 8,8 Hz, 1 H), 6,58 (d, J = 7,3 Hz, 1 H), 4,37-4,37 (m, 1 H), 3,88-3,60 (m, 2 H), 3,52 (dd, J = 5,3, 7,6 Hz, 2 H), 3,17 (s, 2 H), 2, 89–2,82 (m, 2 H), 1,84–1,81 (m, 4 H), 1,61–1,44 (m, 9 STUNDEN). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-piperidin-methanol (Step 2) I-142 2-[2-Fluoro-4-(8-oxa-3-azabicyclo[3.2.1]octan-3-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl ]amino]-6H-naphthyridin-5-on 10cm_Formic_AQ method, m/z = 571 [M + H]+, Ret. Time = 2.43 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,67 (d, J = 4,0 Hz, 1 H), 8,75 (d, J = 1,6 Hz , 1 H), 8,08–8,00 (m, 2 H), 7,51-7,36 (m, 4 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 4,26-4,18 (m, 3 H), 3,68-3,60 (m, 1 H), 3,57-3 ,51 (m, 2 H), 3,49-3,23 (m, 4 H), 3,05-2,98 (m, 1 H), 2,92-2,84 (m, 2 H) , 1,88 -1,82 (m, 4 H), 1,74 (d, J = 2,0 Hz, 1 H), 1,55-1,45 (m, 2 H). F 1-(6-amino-pyridin-3-yl)piperidin-4-ol 8-oxa-3-azabiciclo[3.2.1]octano (Schritt 2) I-143 N-cyclohexyl-3-fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]-amino]-5-oxo-6H-1,6-naphthyridin-2-yl ] -N-methylbenzamide 10cm_Formic_AQ method, m/z = 571 [M + H]+, Ret. Time = 2.68 min. 1RMN-H (400 MHz, DMSO): δ 12,50 (s, 1 H), 11,71 (s, 1 H), 8,80 (s, 1 H), 8,12–8,11 (m , 1 H), 8,09–8,06 (m, 1 H), 7,55-7,45 (m, 1 H), 7,43 (s, 1 H), 7,38 (s, 1 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,73 (d, J = 3,5 Hz, 1 H), 3,69 (dd, J = 3,5, 8,1 Hz, 1 H), 3,61–3,53 (m, 2 H), 3,23 (s, 1 H), 2 ,97–2,85 (m, 4 H), 1,92–1,84 (m, 3 H), 1,76–1,71 (m, 4 H), 1,59–1,50 (m , 5 H), 1,40–1,39 (m, 2 H). ), 1,15–1,09 (m, 2H). F 1-(6-amino-pyridin-3-yl) piperidin-4-ol N-methyl-cyclohexyl-amine (Step 2) . I-144 2-(Azetidin-1-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one Method 10cm_Formic_AQ, m/z = 393 [M + H]+, Ret. Time = 2.32 min. 1RMN-H (400 MHz, DMSO) δ 12,34 (s, 1H), 11,16 (d, J = 4,1 Hz, 1H), 8,29 (s, 1H), 8,01 (d, J = 3,0 Hz, 1H), 7,44 (dd , J = 3,1, 9,0 Hz, 1H), 7,29 (s, 1H), 7,24-7,19 (m, 1H ), 6,90 (d, J = 8,9 Hz, 1H), 6,24 (d, J = 7,3 Hz, 1H), 4,73 (s, 1H), 4,04 (t, J = 7,4 Hz, 4H), 3,67–3,59 (m, 1H), 3,51–3,46 (m, 2H), 2,88–2,80 (m, 2H), 2, 38–2,29 (m, 2H), 1,88–1,80 (m, 2H), 1,56–1,46 (m, 2H). (1 equivalente a Formiatsalz) E 1-(6-amino-pyridin-3-yl)piperidin 4-ol azetidine (stage 2) I-145 2-[2-Fluoro-4-(3-hydroxy-piperidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-on 10cm_Formic_AQ method, m/z = 559 [M + H]+, Ret. Time = 2.38 min. 1RMN-H (400 MHz, DMSO): δ 12,36 (s, 1 H), 11,58 (d, J = 1,3 Hz, 1 H), 8,65 (s, 1 H), 8, 18 (s, 1 H), 7,98 (d , J = 2,8 Hz, 1 H), 7,92 (dd, J = 7,7, 7,7 Hz, 1 H), 7,39 ( dd, J = 2,9, 9,0 Hz, 1 H), 7,32 (s, 1 H), 7,30- 7,27 (m, 1 H), 6,94 (d, J = 9 ,1 Hz, 1 H), 6,49 (d, J = 7,1 Hz, 1 H), 3,57-3,51 (m, 2 H), 3,35 (s, 1 H). ), 3,29–3,24 (m, 4 H), 3,08 (s, 2 H), 2,99–2,98 (m, 1 H), 2,79 (dd, J = 9, 9, F 1-(6-amino-pyridin-3-yl) piperidin-4-ol 3 hydroxy-piperidine (Step 2) . 9.9 Hz, 2H), 1.78-1.73 (m, 4H), 1,44-1,33 (m, 4 h). I-146 2-[2-Fluoro-4-(piperidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-6H-1, 6-naphthyridine-5-on Method 10cm_Bicarb_AQ, m/z = 557 [M+H]+, Ret. Time = 3.17 min. 1RMN-H (400 MHz, DMSO): δ 12,68 (s, 1 H), 11,57-11,57 (m, 1 H), 8,88 (s, 1 H), 8,18 (dd , J = 7,8, 7,8 Hz, 1 H) , 8,12 (d, J = 2,5 Hz, 1 H), 7,52 (dd, J = 2,8, 9,1 Hz, 1 H), 7,45 (s, 1 H), 7,41 (d, J = 6,8 Hz, 1 H), 7,37–7,36 (m, 1 H), 7,07 (d , J = 9,1 Hz, 1 H), 4,74 (s, 1 H), 3,68–3,66 (m, 3 H), 3,58 (d, J = 12,4 Hz, 3 H), 2,96-2,87 (m, 2 H), 2,60-2,30 (m, 3 H), 1,92-1,87 (m, 2 H), 1,67 (s , 3H), 1,59-1,50 (m, 6H). A 2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-8-methyl-1,6-naphthyridin-5(6H)-one (12, 0) (2-Fluoro-4-(piperidine-1-carbonyl)phenyl)borosäure I-147 2-[2-Fluoro-4-[2-(hydroxymethyl)pyrrolidine-1-carbonyl]phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1 ,6-naphthyridine-5-on 10cm_Formic_AQ method, m/z = 559 [M + H]+, Ret. Time = 2.4 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,73–11,71 (m, 1 H), 8,79 (s, 1 H), 8,14–8 ,12 (m, 1 H), 8,06 (dd, J = 7,8, 7,8 Hz, 1 H), 7,53 (dd, J = 2,3, 8,6 Hz, 3 H) , 7,47 (d, J = 7,3 Hz, 1 H), 7,08 (d, J = 9,1 Hz, 1 H). ), 6,64 (d, J = 7,3 Hz, 1 H), 4,89–4,88 (m, 1 H), 4,23–4,17 (m, 1 H), 3,39 (dd, J = 5,4, 15,8 Hz, 4 H), 3,22 (s, 1 H), 3,18 (s, 2 H), 2,97–2,88 (m, 2 H ), 2,13 (s, 1 H), 2,03–1,96 (m, 3 H), 1,91–1,86 (m, 2H), 1,78 (s, 1H), 1, 59-1,49 (m, 2H). A 1-(6-amino-pyridin-3-yl)piperidin-4-ol pyrrolidin-2-yl-methanol (step 2) I-148 2-[2-Fluoro-4-(piperidin-1-carbonyl)phenyl]-4-[[5-(3-oxa-9-azaspiro[5.5]undecan-9-yl)-2-pyridyl]amino]- 6H-Nafthyridin-5-on 10cm_Formic_AQ method, m/z = 597 [M + H]+, Ret. Time = 2.86 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,73–11,67 (m, 1 H), 8,78 (s, 1 H), 8,12 (d , J = 2,8 Hz, 1 H), 8,06 (dd, J = 7,8, 7,8 Hz, 1 H), 7,52 (dd, J = 2,7, 9,0 Hz, 1 H), 7,47 (d, J = 7,3 Hz, 1 H), 7,42–7,36 (m, 2 H), 7,08 (d, J = 9,1 Hz, 1 H ), 6,63 (d, J = 7,1 Hz, 1 H), 3,73–3,60 (m, 7 H), 3,21 (dd, J = 5,3, 5,3 Hz, 4H). ), 1,68 (dd, J = 5,2, 5,2 Hz, 9 H), 1,51 (dd, J = 5,1, 5,1 Hz, 6 H). 5-(3-oxa-9-azaspiro[5.5]undecan-9-yl)pyridine-2-amine (2-fluoro-4-(piperidine-1-carbonyl)phenyl)boronic acid I-149 2-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl]phenyl ]-2-Methyl-propane-nitrile 10cm_Formic_AQ method, m/z = 499 [M + H]+, Ret. Time = 2.6 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,69 (s, 1 H), 8,74 (d, J = 1,6 Hz, 1 H), 8, 07 (d, J = 3,0 Hz, 1 H). ), 8,01 (dd, J = 8,3, 8,3 Hz, 1 H), 7,55-7,46 (m, 3 H), 7,43 (d, J = 7,3 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 4,72 (d, J = 4,1 Hz , 1 H), 3,68–3,58 (m, 1 H), 3,57–3,49 (m, 2 H), 2,91 -2,83 (m, 2 H), 1,87 -1,79 (m, 2 H), 1,76 (s, 6 H), 1,55-1,44 (m, 2 H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(4-(2-cyano-propan-2-yl)-2-fluoro-phenyl)boronic acid I-150 2-[2- Fluor-5-(1-piperidyl-methyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- naphthyridin- 5-ein Method 10cm_Formic_AQ, m/z = 529 [M + H]+, Ret. Time = 2.24 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,68-11,65 (m, 1 H), 8,71 (d, J = 1,5 Hz, 1 H ), 8,06 (d, J = 3,0 Hz, 1 H), 7,85 (dd, J = 2,3, 7,7 Hz, 1 H), 7,48 (dd, J = 3, 1, 9,0 Hz, 1 H), 7,44–7,39 (m, 2 H), 7,28 (dd, J = 8,4, 11,0 Hz, 1 H), 7,03 ( d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 3,9 Hz, 1 H), 3,68 - 3,06 (m, 1 H), 3,55-3,49 (m, 2 H), 3,48 (s, 2 H), 2,91-2,83 A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-(4-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2). ).-El) (M, 2H), 2,39-2,31 (M, 4 selfish) H), 1,88-1,80 (m, 2 H), piperidine 1,54-1,36 (m, 8 H). I-151 N-[3-Fluor-4-[5-oxo-4-[(5-Piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-2-yl]phenyl]ciclohexano -carboxamide Method 10cm_Formic_AQ, m/z = 0 [M + H]+, Ret. Time = 2.42 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,64–11,64 (m, 1 H), 10,23 (s, 1 H), 8,80 (s , 1 H), 8,22 (s, 1 H). ), 8,11 (d, J = 3,0 Hz, 1 H), 8,01 (dd, J = 8,8, 8,8 Hz, 1 H), 7,52 (dd, J = 3, 1, 9,0 Hz, 1 H), 7,47-7,40 (m , 2 H), 7,07 (d, J = 8,9 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 3,29–3,23 (m, 6 H), 2,42–2,33 (m, 1 H), 1,86–1,76 (m, 6 H) , 1,67 (d, J = 11,2 Hz, 1 H), 1,48–1,18 (m, 6 H). A and G tert-butyl-4-(6-aminopyridin-3-yl)-piperazin-1-carboxylate CB19 I-152 N-[3-Fluoro-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ] cyclobutanecarboxamide Method 10cm_Formic_AQ, m/z = 529 [M + H]+, Ret. Time = 2.61 min. 1RMN-H (400 MHz, DMSO): δ 12,38 (s, 1 H), 11,61 (d, J = 4,6 Hz, 1 H), 10,13 (s, 1 H), 8, 73 (s, 1 H), 8,23 (s , 1 H), 8,08 (d, J = 3,0 Hz, 1 H), 8,01 (dd, J = 8,8, 8,8 Hz, 1 H), 7,79 (d, J = 1,9 Hz, 1 H), 7,76 (d, J = 2,0 Hz, 1 H), 7,50–7,38 (m, 2 H), 7,02 (d, J = 8,9 Hz, 1 H), 6,57 (d, J = 7,3 Hz, 1 H), 3,68–3,61 (m , 1 H ), 3,57-3,49 (m, 2 H), 3,42-3,12 (m, 6 H), 2,92-2,83 (m, 2 H), 2,35-2, 10 (m, 2H), 2,02-1,83 Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB24 (m, 1H), 1,57-1,46 (m, 2 h). I-153 N-[3-Fluor-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ]-1-methyl-cyclopropane-carboxamide Method 10cm_Formic_AQ, m/z = 529 [M + H]+, Ret. Time = 2.61 min. 1RMN-H (400 MHz, DMSO): δ 12,39 (s, 1 H), 11,63 (d, J = 4,8 Hz, 1 H), 9,52 (s, 1 H), 8, 72 (s, 1 H), 8,08 (d , J = 3,0 Hz, 1 H), 8,00 (dd, J = 8,9, 8,9 Hz, 1 H), 7,80- 7,76 (m, 1 H), 7,61 (dd, J = 2,1, 8,7 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H ), 7,43-7,38 (m, 1 H), 7,03 (d, J = 9,0 Hz, 1 H), 6,58 (d, J = 7,3 Hz, 1 H) , 3,69-3,60 (m, 1 H), 3,57-3,49 (m, 2 H), 2,92-2,83 (m, 2 H), 1,88-1,80 ( m, 2 H), 1,56-1,46 (m, 3 H), 1,45 (s, 3 H), 1,18-1,13 (m, 2 H), 0,72-0, 68 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB25 I-154 N-[3-Fluoro-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ] cyclopentanocarboxamide 10cm_Formic_AQ method, m/z = 543 [M + H]+, Ret. Time = 2.69 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,62 (s, 1 H), 10,27 (s, 1 H), 8,72 (s, 1 H) , 8,08 (d, J = 3,0 Hz). , 1 H), 8,01 (dd, J = 8,8, 8,8 Hz, 1 H), 7,78 (d, J = 1,9 Hz, 1 H), 7,75 (d, J = 2,0 Hz, 1 H), 7,50–7,44 (m, 2 H), 7,41 (d, J = 7,3 Hz, 1 H), 7,02 (d, J = 8 ,9 Hz, 1 H), 6,57 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz, 1 H), 3,67-3,61 ( m, 1 H), 3,57-3,50 (m, 2 H), 3,18 (d, J = 5,1 Hz, 1 H), 2,92-2,80 (m, 3 H) , 1,92–1,69 (m, 5 H), 1,63–1,46 (m, 5 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB26 I-155 2-[2-Fluoro-4-(piperidine-1-carbonyl)phenyl]-4-[(5-morpholino-2-pyridyl)amino]-6H-1,6-naphthyridin-5-on Method 10cm_Bicarb_AQ, m/z = 529 [M + H]+, Ret. Time = 3.12 min. 1RMN-H (400 MHz, DMSO): δ 12,51 (s, 1 H), 11,69 (s, 1 H), 8,79 (d, J = 1,5 Hz, 1 H), 8, 09 (d, J = 3,0 Hz, 1 H). ), 8,03 (dd, J = 7,8, 7,8 Hz, 1 H), 7,51 (dd, J = 3,1, 9,0 Hz, 1 H), 7,44 (d, J = 7,3 Hz, 1 H), 7,38-7,34 (m , 2 H), 7,08 (d, J = 9,0 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 3,78-3,74 (m, 4 H), 3,70-3,56 (m, 2 H), 3,15 (dd, J = 4,8, 4 ,8 Hz, 4 H), 1,62-1,62 (m, 8 H). A 5-morpholino-pyridine-2-amino-(2-fluoro-4-(piperidine-1-carbonyl)-phenyl)-boronic acid I-156 2-[2- Fluor-5-methoxy-4-(pyrrolidin-1-carbonyl)fenil]-4-[(5-piperazin-1-il-2-pyridyl)amino]-6H-1,6-naphthyridin- 5-ein 10cm_Formic_AQ method, m/z = 544 [M + H]+, Ret. Time = 2.25 min. 1RMN-H (400 MHz, DMSO): δ 12,51 (s, 1 H), 11,88 (s, 1 H), 8,81 (s, 1 H), 8,12-8,09 (m , 1 H), 7,63 (d, J = 5,8 Hz, 1 H), 7,54–7,46 (m, 2 H), 7,30 (d, J = 8,8 Hz, 1 H), 7,09 (d, J = 8,8 Hz, 1 H), 6,66 (d, J = 7,3 Hz, 1 H), 3,92 (s, 3 H), 3,51 (dd, J = 6,1, 6,8 Hz, 2 H), 3,24 (dd, J = 6,3, 6,3 Hz, 2 H), 3,10 (dd, J = 5,1 , 5,1 Hz, 4 H), 2,88 (dd, J = 5,2, 5,2 Hz, 4 H), 1,96–1,84 (m, 5 H). F and G tert-butyl pyrrolidine 4-(6-amino-pyridin-3-yl)piperazine-1-carboxylate (step 2) I-157 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)phenyl]-4-[(5-piperazin-1-yl]-2-pyridyl)amino]-6H-1,6-naphthyridine - 5-ein 10cm_Formic_AQ method, m/z = 558 [M + H]+, Ret. Time = 2.31 min. 1RMN-H (400 MHz, DMSO): δ 12,54 (s, 1 H), 11,77–11,77 (m, 1 H), 8,82 (s, 1 H), 8,33 (s , 1 H), 8,15–8,10 (m, 1 H), 7,61 (d, J = 6,1 Hz, 1 H), 7,56-7,46 (m, 2 H), 7,30 (s, 1 H), 7,10 (d, J = 8,8 Hz, 1 H), 6,66 (d, J = 7,3 Hz, 1 H), 3,91 (s, 3 H), 3,25–3,17 (m, 4 H), 3,01–2,94 (m, 4 H), 2,47–2,47 (m, 2 H), 2,39 - 2,36 (m, 1 H), 1,70-1,60 (m, 4 H), 1,53-1,47 (m, 3 H). F and G tert-butyl 4-(6-amino-pyridin-3-yl) piperazin- 1- carboxy-late piperidine (step 2) . I-158 N-[3-Fluoro-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-8-methyl-5-oxo-6H-1,6-naphthyridine-2 -il]phenyl]cyclohexanocarboxamide AcHSS C18 method, m/z = 571 [M+H]+, Ret. time = 3.61 min. 1RMN-H (400 MHz, DMSO): δ 12,61 (s, 1 H), 11,54–11,50 (m, 1 H), 10,28 (s, 1 H), 8,86 (s , 1 H), 8,19–8,11 (m, 2 H), 7,55-7,48 (m, 2 H), 7,36-7,32 (m, 1 H), 7,05 (d, J = 8,8 Hz, 1 H), 4,76 (d, J = 4,3 Hz, 1 H), 3,71-3,67 (m, 1 H), 3,60-3 ,55 (m, 2 H), 2,94-2,87 (m, 2 H), 2,42 (dd, J = 11,7, 15,0 Hz, 2 H), 2,31 (s, 3 H), 1,92-1,80 (m, 6 H), 1,73 (s, 1 H), 1,57-1,44 (m, 4 H), 1,36-1,26 ( m, 3H). A 2-chloro-4-((5-(4-hydroxypiperidin-1-yl)pyridin-2-yl)amino)-8-methyl-1,6-naphth-thyridin-5(6H)-one (12, 0) CB19 I-159 N-[3-Fluor-4-[4-[(5-morpholino-2-pyridyl)amino]-5-oxo-6H-1,6-naftiridin-2-yl]phenyl]cyclohexancarboxamide BicarbB EHC18 method, m/z = 543 [M + H]+, Ret. Time = 4.57 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,68 (s, 1 H), 10,27 (s, 1 H), 8,82 (s, 1 H) , 8,13 (d, J = 2,5 Hz). , 1 H), 8,05 (dd, J = 8,7, 8,7 Hz, 1 H), 7,56-7,48 (m, 2 H), 7,45 (d, J = 7, 1 Hz, 1 H), 7,10 (d, J = 9,1 Hz, 1 H), 6,62 (d, J = 7,1 Hz, 1 H), 3,84–3,78 (m , 4 H), 3,19 (dd, J = 4,5, 4,5 Hz, 4 H), 2,45–2,38 (m, 1 H), 1,91-1,79 (m, 4 H), 1,74-1,70 (m, 1 H), 1,52-1,23 (m, 6 H). Ein 5-morpholino-pyridine-2-amine CB19 I-160 N-[3-Fluoro-4-[5-oxo-4-[4-(pyrrolidine-1-carbonyl)anilino]-6H-1,6-naphthyridin-2-yl]phenyl]cyclohexanecarboxamide BicarbB EHC18 method, m/z = 554 [M + H]+, Ret. Time = 4.49 min. 1RMN-H (400 MHz, DMSO): δ 11,80 (s, 1 H), 11,69 (d, J = 1,3 Hz, 1 H), 10,26 (s, 1 H), 8, 07 (dd, J = 9,0, 9,0 Hz, 1 H), 7,79-7,77 (m, 1 H), 7,74 (d, J = 1,8 Hz, 1 H), 7,68 (d, J = 8,3 Hz, 2 H), 7,51-7,45 (m, 4 H), 6,62 (d, J = 7,3 Hz, 1 H), 3, 52 (s, 4 H), 1,94–1,93 (m, 1 H), 1,92–1,87 (m, 6 H), 1,86–1,80 (m, 2 H) , 1,72-1,69 (m, 1 H), 1,51-1,41 (m, 2 H), 1,35-1,25 (m, 3 H). A (4-amino-phenyl)(pyrrolidin-1-yl) methanone CB19 I-161 N-[3-Fluor-4-[4-[(5-Methoxy-2-pyridyl)amino]-5-oxo-6H-1,6-naphthiridin-2-yl]fenil]ciclohexancarboxamida AcHSS C18 method, m/z = 488 [M+H]+, Ret. time = 3.81 min. 1RMN-H (400 MHz, DMSO): δ 12,55 (s, 1 H), 11,72 (s, 1 H), 10,29 (s, 1 H), 8,84 (s, 1 H) , 8,17 (d, J = 2,8 Hz). , 1 H), 8,05 (dd, J = 8,8, 8,8 Hz, 1 H), 7,83 (d, J = 1,3 Hz, 1 H), 7,55–7,45 (m, 3 H), 7,17 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 3,89 (s, 3 H) , 2,72 (s, 1 H), 2,44-2,38 (m, 1 H), 1,84 (dd, J = 11,7, 24,6 Hz, 4 H), 1,73- Ein 5-methoxypyridin-2-amine CB19 1,70 (m, 1H), 1,52-1,26 (M, 4H). I-162 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-uno AcHSS C18 method, m/z = 573 [M+H]+, Ret. time = 2.92 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (s, 1 H), 11,67 (s, 1 H), 8,81 (s, 1 H), 8,10 (d, J = 2 ,8 Hz, 1 H), 7,92 (d , J = 8,8 Hz, 1 H), 7,53 (dd, J = 2,9, 9,0 Hz, 1 H), 7,46 ( d, J = 5,6 Hz, 1 H), 7,07 (d, J = 9,1 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,77 (d, J = 4,3 Hz, 1 H), 3,94 (s, 3 H), 3,70–3,53 (m, 6 H), 3,21 (s, 2 H), 2, 96–2,89 (m, 2 H), 1,90–1,84 (m, 2 H), 1,65–1,45 (m, 8 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB27 I-163 2-[2-Fluor-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-8-metil- 6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 587 [M+H]+, Ret. time = 3.32 min. 1RMN-H (400 MHz, DMSO): δ 12,68 (s, 1 H), 11,59 (s, 1 H), 8,89 (s, 1 H), 8,12 (d, J = 2 ,5 Hz, 1 H), 7,78 (d , J = 6,1 Hz, 1 H), 7,53 (dd, J = 2,9, 9,0 Hz, 1 H), 7,37 ( s, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 4,76 (d, J = 4,0 Hz, 1 H), 3,91 (s, 3 H), 3,70–3,53 (m, 6 H), 3,25–3,21 (m, 2 H), 2,94–2,88 (m, 2 H), 2,32 (s , 3 H ), 1,88 (d, J = 9,3 Hz, 2 H), 1,67-1,50 (m, 8 H). A 2-Chloro-4-((5-(4-hidroxipiperidin-1-yl)pyridin-2-yl)amino)-8-methyl-1,6-nafthyridin-5(6H- em (12.0) CB1 I-164 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[(5-morpholino-2-pyridyl)amino]-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 559.2 [M+H]+, Ret. time = 3.39 min. 1RMN-H (400 MHz, DMSO): γ; 12,54 (s, 1 H), 11,73–11,71 (m, 1 H), 8,83 (s, 1 H), 8,13 (d, J = 2,5 Hz, 1 H) , 7,61 (d, J = 6,1 Hz, 1 H), 7,54 (dd, J = 2,8, 8,8 Hz, 1 H), 7,48 (d, J = 7,3 Hz, 1 H), 7,29 (d, J = 9,1 Hz, 1 H), 7,11 (d, J = 9,1 Hz, 1 H), 6,66 (d, J = 7, 3 Hz, 1 H), 3,91 (s, 3 H), 3,80 (dd, J = 4,5, 4,5 Hz, 4 H). ), 3,71-3,59 (m, 2 Std.), 3,23-3,15 (m, 6 Std.), 1,70-1,50 (m, 6 Std.). Ein 5-morpholino-pyridine-2-amine CB1 I-165 2-[2-Fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl]-4-[4-(pyrrolidine-1-carbonyl)amino]-6H-1,6-naphthyridin-5-on BicarbB EHC18 method, m/z = 571 [M + H]+, Ret. Time = 3.98 min. 1RMN-H (400 MHz, DMSO): δ 11,79 (s, 1 H), 11,73 (s, 1 H), 7,64–7,57 (m, 2 H), 7,46–7 ,39 (m, 4 H), 7,19 (d, J = 7,1 Hz, 1 H), 6,60 (d, J = 7,1 Hz, 1 H), 3,84 (s, 3 H), 3,65–3,53 (m, 2 H), 3,46 (dd, J = 10,1, 10,1 Hz). , 4 H), 3,12 (dd, J = 6,1, 6,1 Hz, 2 H), 2,07 (s, 1 H), 1,85-1,84 (m, 4 H), 1,54 (dd, J = 4,4, 21,3 Hz, 4 H), 1,45 (d, J = 5,3 Hz, 2 H). A (4-amino-phenyl)(pyrrolidin-1-yl) methanone CB1 I-166 2-[2-Fluoro-3-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridin-5-one AcHSS C18 Method, m/z 573.3 [M+H]+, Ret. time = 2.95 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,70-11,70 (m, 1 H), 8,71 (d, J = 1,5 Hz, 1 H ), 8,09 (d, J = 2,9 Hz, 1 H), 7,67-7,63 (m, 1 H), 7,49 (dd, J = 3,1, 9,0 Hz, 1 H), 7,44 (d, J = 7,3 Hz, 1 H), 7,17 (d, J = 8,0 Hz, 1 H), 7,04 (d, J = 8,9 Hz , 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 3,90 (s, 3 H), 3,72–3,49 (m, 5 H), 3,21 ( dd, J = 5,5, 5,5 Hz, 3 H), 2,92–2,83 (m, 2 H), 1,88–1,80 (m, 2 H), 1,65–1 ,45 (m, 8 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB28 I-167 2-[2-Fluoro-4-Methyl-5-(Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on AcHSS C18 method, m/z = 557 [M+H]+, Ret. time = 2.95 min. 1RMN-H (400 MHz, DMSO): γ; 12,44 (s, 1 H), 11,67–11,65 (m, 1 H), 8,75 (s, 1 H), 8,06 (d, J = 3,0 Hz, 1 H) , 7,79 (d, J = 8,0 Hz 1 H), 7,49 (dd, J = 3,1, 9,0 Hz, 1 H), 7,42 (d, J = 7,3 Hz , 1 H), 7,04 (d, J = 8,9 Hz, 1 H). ), 6,60 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,3 Hz, 1 H), 3,68-3,49 (m, 6 H), 3,18 (dd, J = 5,3, 5,3 Hz, 2 H), 2,92-2,84 (m, 2 H), 2,30 (s, 3 H), Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB29 1.87-1.81 (m, 2H), 1.64- 1,43 (m, 8 H). I-168 2-(6-Fluor-3-methyl-1H-indazol-5-yl)-4-[[5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 471 [M+H]+, Ret. time = 1.92 min. 1RMN-H (400 MHz, DMSO): δ 12,89–12,80 (m, 1 H), 12,48 (s, 1 H), 11,71–11,59 (m, 1 H), 8 ,76–8,75 (m, 1 H), 8,30– 8,27 (m, 1 H), 8,21 (s, 1 H), 8,10-8,08 (m, 1 H) , 7,51 (dd, J = 3,1, 9,0 Hz, 1 H), 7,45-7,37 (m, 2 H), 7,09-7,06 (m, 1 H), 6,66-6,63 (m, 1 H), 3,85 (s, 3 H), 3,25-3,20 (m, 4 H), 3,06 (t, J = 5,0 Hz , 4 H ). A and G tert-butyl-4-(6-amino-pyridin-3-yl)-piperazin-1-carboxy-lat CB30 I-169 4-[[5-(4,4-difluoro-1-piperidyl)-2-pyridyl]amino]-2-[2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl]-6H- 1,6-Nafthyridine-5-on AcHSS C18 method, m/z = 593 [M+H]+, Ret. time = 3.85 min. 1RMN-H (400 MHz, DMSO): γ; 12,56 (s, 1 H), 11,76–11,71 (m, 1 H), 8,84 (s, 1 H), 8,20 (d, J = 2,8 Hz, 1 H) , 7,64 -7,58 (m, 2 H), 7,47 (d, J = 6,6 Hz, 1 H), 7,27 (s, 1 H), 7,11 (d, J = 8,8 Hz, 1 H), 6,66 (d, J = 7,1 Hz, 1 H), 3,91 (s, 3 H), 3,89 (s, 1 H), 3,68- 3,62 (m, 3 H), 3,21 (s, 3 H), 2,19-2,09 (m, 4 H), 1,64 (dd, J = 4,4, 20,8 Hz , 4 H), 1,52-1,49 (m, 3 H). Ein 5-(4,4-Difluoro-piperidin-1-yl)-pyridin-2-amine CB1 I-170 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)phenyl]-4-[[5-[(1R,5S)-8-oxa-3-azabicyclo[3.2.1]octan- 3-yl]-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 585.3 [M+H]+, Ret. time = 3.45 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,72 (s, 1 H), 8,76 (s, 1 H), 8,04 (d, J = 2 ,5 Hz, 1 H), 7,62 (d , J = 6,1 Hz, 1 H), 7,50-7,41 (m, 2 H), 7,28 (d, J = 8,8 Hz, 1 H), 7,09 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7,1 Hz, 1 H), 4,51–4,45 (m, 2 H), 3,91 (s, 3 H), 3,70–3,62 (m, 2 H), 3,47 (d, J = 10,6 Hz, 2 H), 3,22-3 ,17 (m, 2 H), 2,87 (d, J = 9,6 Hz, 2 H), 1,91 (s, 4 H), 1,67-1,60 (m, 4 H), 1,53-1,47 (m, 2H) . Ein 5-((1R,5S)-8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)pyridin-2-amine CB1 I-171 2-[2- Fluor-3-(morpholino-methyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- naphthyridin- 5- one AcHSS C18 method, m/z = 531 [M+H]+, Ret. time = 1.98 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,66 (d, J = 5,3 Hz, 1 H), 8,63 (d, J = 1,5 Hz , 1 H), 8,22 (s, 1 H). ), 8,06 (d, J = 3,0 Hz, 1 H), 7,81–7,76 (m, 1 H), 7,56–7,46 (m, 2 H), 7,44 –7,40 (m, 1 H), 7,32 (dd, J = 7,6, 7,6 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6 ,58 (d, J = 7,0 Hz, 1 H), 3,51-3,78 (m, 6 H), 3,57-3,48 (m, 2 H), 2,91-2, 83 (m, 2 H), 2,70-2,55 (m, 1 H), 2,44 (d, J = 4,3, A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 4-(2-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2 ).-yl)Benzilo) 4,3 Hz, 4 H), 1,87-1,80 Morpholine (M, 2H), 1.56-1.45 (M, 2 h). I-172 N-[4-[4-[[5-(4,4- Difluor-1-piperidil)-2-piridil]amino]-5-oxo-6H-1,6-naftiridin-2-il]-3- Fluorfenil]ciclohexancarboxamida AcHSS C18 method, m/z = 577 [M+H]+, Ret. time = 4.2 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 10,23 (s, 1 H), 8,78 (s, 1 H), 8,15 (d, J = 2 ,5 Hz, 1 H), 7,99 (dd , J = 8,5, 8,5 Hz, 1 H), 7,75 (d, J = 8,9 Hz, 1 H), 7,56 ( dd, J = 2,9, 8,8 Hz, 1 H), 7,46 (d, J = 8,5 Hz, 1 H), 7,41 (d, J = 7,3 Hz, 1 H) , 7,06 (d, J = 8,9 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 3,34 (dd, J = 5,6, 5,6 Hz, 4 H), 2,40–2,34 (m, 4 H), 2,14–2,04 (m, 4 H), 1,85–1,76 (m, 4 H), 1, 66 (s, 2 H), 1,45–1,39 (m , 2 Std.). Ein 5-(4,4-Difluor-piperidin-1-il)-piridin-2-amina CB19 I-173 N-[3-Fluor-4-[4-[[5-[(1R,5S)-8-oxa-3-azabicyclo[3.2.1]octan-3-yl]-2-pyridyl]amino]-5 -oxo-6H-1,6-nafthyridin-2-yl]-phenyl]-cyclohexane-carboxamide BicarbB EHC18 method, m/z = 569.3 [M+H]+, Ret. tempo = 4.82 min. 1RMN-H (400 MHz, DMSO): γ; 12,36 (s, 1 H), 11,61 (d, J = 4,6 Hz, 1 H), 10,21 (s, 1 H), 8,71 (s, 1 H), 8,03 –7,97 (m, 2 H), 7,75 (dd, J = 2,0, 8,7 Hz, 1 H), 7,46 (dd, J = 2,0, 8,7 Hz, 1 H), 7,39 (dd, J = 3,1, 8,8 Hz, 2 H). ), 7,04 (d, J = 9,0 Hz, 1 H), 6,57 (d, J = 7,3 Hz, 1 H), 4,46–4,43 (m, 2 H), 3,45–3,38 (m, 2 H), 2,85 (dd, J = 2,3, 11,2 Hz, 2 H), 2,42–2,34 (m, 2 H), 1 ,89–1,75 (m, 6 H), 1,69 (d, J = 11,2 Hz, 1 H), 1,48–1,19 (M, 6H). Ein 5-((1R,5S)-8-Oxa-3-azabicyclo[3.2.1]octan-3-yl)pyridin-2-amine CB19 I-174 4-[[5-(3,3- Difluorazetidin-1-yl)-2-pyridyl]amino]-2-[2-fluoro-5-methoxy-4-(piperidin-1-carbonyl)phenyl]- 6H- 1,6-naphthyridin-5-one AcHSS C18 method, m/z = 565 [M+H]+, Ret. time = 3.66 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,68 (d, J = 1,5 Hz, 1 H), 8,77 (s, 1 H), 7, 79 (d, J = 2,0 Hz, 1 H). ), 7,58 (d, J = 5,8 Hz, 1 H), 7,42 (d, J = 7,1 Hz, 1 H), 7,25 (dd, J = 2,9, 8, 5 Hz, 1 H), 7,14 (dd, J = 2,9 , 8,5 Hz, 1 H), 7,08 (d, J = 8,6 Hz, 1 H), 6,61 (d , J = 7,3 Hz, 1 H), 4,33 (dd, J = 12,3, 12,3 Hz, 4 H), 3,85 (s, 3 H), 3,64-3,54 (m, 2 H), 3,15 (s, 2 H), 1,62-1,54 (m, 4 H), 1,48-1,43 (m, 2 H). A 5-(3,3-difluoroazetidin-1-yl)pyridine-2-amine CB1 I-175 2-[4-(Azetidina-1-carbonyl)-2-fluoro-5-methoxy-phenyl]-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthyridine - 5-ein AcHSS C18 method, m/z = 530 [M+H]+, Ret. time = 1.99 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 8,74 (s, 1 H), 8,05 (d, J = 2,8 Hz, 1 H), 7, 58 (d, J = 6,1 Hz, 1 H). ), 7,48-7,40 (m, 2 H), 7,29 (d, J = 8,8 Hz, 1 H), 7,03 (d, J = 8,8 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 4,05-3,92 (m, 4 H), 3,88 (s, 3 H), 3,19 (s, 1 H) , 3,07 (dd, J = 4,5, 4,5 Hz, 4 H), 2,86 (dd, J = 4,9, 4,9 Hz, 4 H), 2,33-2,20 (m, 3H). F and G tert-butyl azetidine 4-(6-amino-pyridin-3-yl)piperazine-1-carboxylate (Step 2) I-176 5-Fluor-2- methoxy- N,N- dimethyl-4- [5-oxo-4- [(5- piperazin-1-yl-2- pyridyl) amino]-6H- 1,6- naphthiridina-2- il]benzamide AcHSS C18 method, m/z = 518 [M+H]+, Ret. time = 1.97 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (s, 1 H), 8,75 (s, 1 H), 8,05 (d, J = 2,8 Hz, 1 H), 7, 58 (d, J = 5,8 Hz, 1 H). ), 7,48-7,40 (m, 2 H), 7,24 (d, J = 8,8 Hz, 1 H), 7,03 (d, J = 8,8 Hz, 1 H), 6,61 (d, J = 7,1 Hz, 1 H), 3,86 (s, 3 H), 3,23-3,13 (m, 2 H), 3,07 (dd, J = 4 ,4, 4,4 Hz, 4 H), 2,99 (s, 3 H), 2,86 (dd, J = 4,4, 4,4 Hz, 4 H), 2,82 (s, 3 h). F and G tert-butyl dimethylamine 4-(6-amino-pyridin-3-yl)piperazine-1-carboxylate (step 2) I-177 2-[2-Fluoro-5-(pyrrolidin-1-ylmethyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine- on the 5th AcHSS C18 method, m/z = 515 [M+H]+, Ret. time = 2.06 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 8,71 (d, J = 1,6 Hz, 1 H), 8,06 (d, J = 3,0 Hz , 1 H), 7,87 (dd, J = 2,3, 7,7 Hz, 1 H), 7,50–7,41 (m, 3 H), 7,28 (dd, J = 8, 4, 11,2 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6,61 (d , J = 7,2 Hz, 1 H), 4,71-4 ,70 (m, 1 H), 3,64 (s, 3 H), 3,57-3,48 (m, 2 H), 2,91-2,83 (m, 2 H), 2,49 - 2,41 (m, 5 H), 1,88-1,80 (m, 2 H), 1,73-1,68 (m, 4 H), 1,56-1,45 (m, 2 h). A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-(4-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2) ).-il) Selfish) Pyrrolidine I-178 2-(6-Fluor-2-methyl-1,3-benzoxazol-5-yl)-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthyridin-5 -eins AcHSS C18 method, m/z = 472 [M+H]+, Ret. time = 2.02 min. 1RMN-H (400 MHz, DMSO): δ 12,50 (s, 1 H), 8,77 (d, J = 2,0 Hz, 1 H), 8,22-8,14 (m, 2 H ), 8,08 (d, J = 3,0 Hz, 1 H), 7,83 (d, J = 10,4 Hz, 1 H), 7,52-7,42 (m, 2 H), 7,09-7,05 (m, 1 H), 6,64-6,62 (m, 1 H), 3,32 (br , 1 Std.), 3,19–3,14 (m, 4 estándar), 3,01–2,96 (m, 4 estándar), 2,67–2,66 (m, 3 estándar). A and G tert-butyl-4-(6-amino-pyridin-3-yl)piperazin-1-carboxylate CB31 I-179 N-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridin-2-yl]phenyl ] Norbornan-2-carboxamide AcHSS C18 method, m/z = 569.3 [M+H]+, Ret. time = 3.46 min. 1RMN-H (400 MHz, DMSO): δ 12,36 (s, 1 H), 11,59 (d, J = 5,3 Hz, 1 H), 10,16 (s, 1 H), 8, 71 (s, 1 H), 8,17 (s , 1 H), 8,06 (d, J = 2,5 Hz, 1 H), 8,02-7,94 (m, 1 H), 7 ,77-7,71 (m, 1 H), 7,49-7,43 (m, 2 H), 7,38 (dd, J = 6,6, 6,6 Hz, 1 H), 7, 01 (d, J = 9,1 Hz, 1 H), 6,56 (d, J = 7,3 Hz, 1 H), 4,68–4,68 (m, 1 H), 3,66 - 3,60 (m, 1 H), 2,87 (dd, J = 9,7, 9,7 Hz, 3 H), 2,43 (d, J = 13,6 Hz, 1 H), 2, 28-2,22 (metro, 2H), 1,87-1,81 (metro, 2H), 1,69-1,65 Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB32 (m, 2H), 1.58-1.22 (m, 8 h). I-180 2-[2-Fluor-4-[(4-methyl-piperazin-1-yl)methyl]phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H - 1,6-Naphthiridina- AcHSS C18 method, m/z = 544.3 [M+H]+, Ret. time = 2.22 min. 1RMN-H (400 MHz, DMSO): γ; 12,43 (s, 1 H), 11,67 (s, 1 H), 8,46 (s, 1 H), 8,11 (d, J = 2,3 Hz, 1 H), 7,60 –7,54 (m, 1 H), 7,50 (dd, J = 2,8, 8,8 Hz, 1 H), 7,45 (d, J = 7,1 Hz, 1 H), 7 ,32–7,25 (m, 2 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,58 (d, J = 7,3 Hz, 1 H), 4,73 (d, J = 3,8 Hz, 1 H), 3,71-3,64 (m, 1 H), 3,37 (s, 3 H ), 2,89 (dd, J = 10,0, 10,0 Hz, 2H), 2,34–2,28 Ein 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-1-(3-fluoro-4-(4,4,5,5-tetrametil-1,3,2-dioxa-borolano) ) ) -2).- 5 how (m, 4H), 2,15-2,14 (m, 4 il)bencilo)- H), 2,07 (s, 3 H), 1,88- 4- 1.86 (metro, 3H), 1.59-1.47 Methyl- (M, 2H). piperazine I-181 2-[2- Fluor-4-(morpholino-methyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- naphthyridin- 5- one BicarbB EHC18 method, m/z = 531 [M + H]+, Ret. Time = 3.41 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,67 (s, 1 H), 8,75 (s, 1 H), 8,11 (d, J = 2 ,5 Hz, 1 H), 7,97 (dd , J = 8,1, 8,1 Hz, 1 H), 7,52 (dd, J = 2,8, 8,8 Hz, 1 H), 7,45 (d, J = 7,1 Hz, 1 H), 7,34 (d, J = 7,3 Hz, 1 H), 7,06 (d, J = 9,1 Hz, 1 H) , 6,62 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 3,5 Hz, 1 H), 3,63 (s, 4 H), 3,66–3 ,53 (m, 6 H), 2,91 (dd, J = 9,9, 9,9 Hz, 2 H), 2,45 (s, 4 H), 1,88 (d, J = Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 4-(3-Fluor-4-(4,4,5,5-Tetrametil-1,3,2-Dioxa-Borolan-2 ).-yl)Benzilo) 10,1 Hz, 2 H), 1,60–1,49 Morpholine (M, 2H). I-182 2-(6-Fluor-2-methyl-1,3-benzoxazol-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6 - Naphthyridin-5-one AcHSS C18 method, m/z = 487.2 [M+H]+, Ret. time = 2.66 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,72-11,66 (m, 1 H), 8,73 (d, J = 2,0 Hz, 1 H ), 8,14 (d, J = 7,0 Hz, 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,83 (d, J = 10,4 Hz, 1 H), 7,50–7,42 (m, 2 H), 7,05–7,02 (m, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4, 69 (d, J = 3,1 Hz, 1 H), 3,67–3,63 (m, 1 H), 3,57–3,49 (m, 2 H), 2,91–2,83 ( m, 2 h), 2,67 (s, 3 h), 1,86–1,81 (m, 2 h), 1,55–1,45 (m, 2 h). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4ol CB31 I-183 2-[2- Fluor-3-(1-piperidyl-methyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2- pyridyl]amino]-6H- 1,6- naphthyridin- 5-ein AcHSS C18 method, m/z = 529 [M+H]+, Ret. time = 2.03 min. 1RMN-H (400 MHz, DMSO): γ; 12,42 (s, 1 H), 11,68–11,64 (m, 1 H), 8,63 (d, J = 1,5 Hz, 1 H), 8,22 (s, 1 H) , 8,06 (d, J = 3,0 Hz, 1 H), 7,79-7,74 (m, 1 H), 7,54-7,39 (m, 3 H), 7,31 ( t, J = 7,6 Hz, 1 H), 7,05-7,01 (m , 1 H), 6,58 (d, J = 7,3 Hz, 1 H), 3,50 (d, J = 9,3 Hz, 2 H), 2,91-2,83 (m, 2 H), 2,44-2,37 (m, 6 H), 1,87-1,79 (m, 2 H), 1,55-1,38 (m, 9 H). A 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 1-(2-Fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2). ). ).-- hand) selfish) piperidine I-184 N-[3-Fluor-4-[4-[[5-(4-Methyl-piperazin-1-yl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl ] fenil] ciclohexancarboxamide AcHSS C18 method, m/z = 556.3 [M+H]+, Ret. time = 2.59 min. 1RMN-H (400 MHz, DMSO): δ 12,40 (s, 1 H), 11,62 (d, J = 5,8 Hz, 1 H), 10,22 (s, 1 H), 8, 75 (s, 1 H), 8,08 (d , J = 3,0 Hz, 1 H), 8,01 (dd, J = 8,8, 8,8 Hz, 1 H), 7,77 ( d, J = 1,9 Hz, 1 H), 7,51-7,44 (m, 1 H), 7,43- 7,38 (m, 1 H), 7,04 (d, J = 8 ,9 Hz, 1 H), 6,58 (d, J = 6,9 Hz, 1 H), 3,17 (dd, J = 5,0, 5,0 Hz, 4 H), 2,50 - 2,45 (m, 3 H), 2,42-2,34 (m, 2 H), 2,24 (s, 3 H), 1,86-1,77 (m, 4 H), 1, 68 (d, J = 11,0 Hz, 1 H), 1,49-1,18 (m, 6H). Ein 5-(4-Methyl-piperazin-1-yl)-pyridin-2-amine CB19 I-185 2-(2-Fluoro-6-methoxy-fenil)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 462 [M+H]+, Ret. time = 2.75 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,66 (d, J = 5,6 Hz, 1 H), 8,31 (s, 1 H), 8, 18 (s, 1 H), 8,04 (d , J = 3,0 Hz, 1 H), 7,50–7,38 (m, 2 H), 7,02–6,90 (m, 2 H), 6,53 (d, J = 7,0 Hz, 1 H), 4,69–4,68 (m, 1 H), 3,75 (s, 3 H), 3,66-3, 56 (m, 2 H), 3,53-3,46 (m, 3 H), 3,18 (s, 1 H), 2,88-2,80 (m, 2 H), 1,85 - 1,78 (m, 2 H), 1,53-1,42 (m, 2 H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(2-fluoro-6-methoxyphenyl)boronic acid I-186 2-[2-Fluoro-5-isopropoxy-4-(piperidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-on AcHSS C18 method, m/z = 601.3 [M+H]+, Ret. time = 3.25 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (s, 1 H), 11,67 (d, J = 4,1 Hz, 1 H), 8,74 (d, J = 1,4 Hz , 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,56 (d, J = 6,1 Hz, 1 H), 7,51-7,40 (m, 2 H), 7,21 (d, J = 8,9 Hz, 1 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 4,70–4,62 (m, 1 H), 3,67–3,49 (m, 6 H), 3,23–3,16 (m, 2 H), 2,92 - 2,83 (m, 2 H), 1,88-1,80 (m, 2 H), 1,62-1,45 (m, 8 H), 1,29 (dd, J = 5,9, 17,1 Hz, 6 H). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB33 I-187 2-[3-Methoxy-4-(piperidin-1-carbonyl)phenyl]-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-5-one BicarbB EHC18 method, m/z = 540 [M + H]+, Ret. Time = 3.4 min. 1RMN-H (400 MHz, DMSO): γ; 12,47 (s, 1 H), 8,90 (s, 1 H), 8,13 (d, J = 3,0 Hz, 1 H), 7,75 (s, 1 H), 7,65 –7,62 (m, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,43 (d, J = 7,3 Hz, 1 H), 7 ,31 (d, J = 7,8 Hz, 1 H), 7,07 (d, J = 8,9 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 3,92 (s, 3 H), 3,67 (d, J = 5,5 Hz, 1 H), 3,59-3,58 (m , 1 H), 3,30 (d, J = 3 ,9 Hz, 2 H), 3,18-3,05 (m, 6 H), 2,86 (dd, J = 4,8, 4,8 Hz, 4 H), 1,63-1,57 (m, 4 H), 1,43–1,42 (m, 2 H). A and G tert-butyl-4-(6-amino-pyridin-3-yl)piperazin-1-carboxylate CB34 I-188 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[[5-[3-(2-methoxy-ethoxy)azetidin-1-yl]-2-pyridyl] amino]-6H-1,6- AcHSS C18 method, m/z = 603.3 [M+H]+, Ret. time = 3.39 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,70 (d, J = 5,1 Hz, 1 H), 8,72 (s, 1 H), 7, 71 (d, J = 2,0 Hz, 1 H). ), 7,63 (d, J = 5,8 Hz, 1 H), 7,47 (dd, J = 6,7, 6,7 Hz, 1 H), 7,29 (d, J = 7, 1 Hz, 1 H), 7,08–7,05 (m, 1 H), 6,65 (d, J = 7,1 Hz, 1 H), 4,52–4,47 (m, 1 H ), 4,16 (dd, J = 6,9, 6,9 Hz, 4 H), 3,91 (s, 3 H), 3,61 -3,47 (m, 4 H), 3,34 (d, J = 20,5 Hz, 4 H), 3,21 A CA3 CB1 naphthyridin- (dd, J = 4,8, 4,8 Hz, 4 H), 5 how 1,67-1,60 (m, 4 H), 1,52- 1.50 (m, 2H). I-189 N-[3-Fluoro-4-[4-[[5-[3-(2-Methoxy-ethoxy)azetidin-1-yl]-2-pyridyl]amino]-5-oxo-6H-1,6- Naftiridin-2-yl]fenil]ciclohexancarboxamide AcHSS C18 method, m/z = 587.3 [M+H]+, Ret. time = 3.8 min. 1RMN-H (400 MHz, DMSO): δ 12..31 (s, 1 H), 11,57 (d, J = 5,8 Hz, 1 H), 10,19 (s, 1 H), 8 ,64 (s, 1 H), 7,99 (dd, J = 9,0, 9,0 Hz, 1 H), 7,75 (d, J = 1,5 Hz, 1 H), 7,66 (d, J = 1,5 Hz, 1 H), 7,45 (d, J = 9,1 Hz, 1 H), 7,38 (dd, J = 6,6, 6,6 Hz, 1 H ), 7,01 (s, 1 H), 6,55 (d, J = 7,6 Hz, 1 H), 4,48–4,42 (m, 1 H), 4,11 (dd, J = 7,2, 7,2 Hz, 2 H), 3,64 (dd, J = 4,5, 7,8 Hz, 2 UNCA3CB19 H), 3,55-3,44 (m, 4 H), 3,27-3,19 (m, 4 H), 2,40- 2,32 (m, 1H), 1,85-1,75 (m, 4 Std.), 1,68 (s, 1 Std.), 1.47-1.37 (m, 2H), 1.33- 1,21 (M, 4H). I-190 4-[[5-[(3R, 5R)-3,5-Dimethyl-morpholin-4-yl]-2-pyridyl]amino]-2-[2-fluoro-5-methoxy-4-(piperidin- 1 -Carbonyl)phenyl]-6H-1,6-naphthyridin-5-on BicarbB EHC18 method, m/z = 587.3 [M + H]+, Ret. Time = 4.48 min. 1RMN-H (400 MHz, DMSO): δ 12,64 (s, 1 H), 11,72-11,72 (m, 1 H), 8,92 (s, 1 H), 8,10 (d , J = 2,0 Hz, 1 H), 7,57 (d, J = 5,8 Hz, 1 H), 7,51 (dd, J = 2,1, 8,7 Hz, 1 H), 7,45–7,43 (m, 1 H), 7,24 (d, J = 8,8 Hz, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6, 62 (d, J = 7,3 Hz, 1 H), 3,86 (s, 3 H), 3,81 (dd, J = 2,5, 10,9 Hz, 2 H), 3,64 - 3,57 (m, 2 H), 3,52-3,37 (m, 4 H), 3,16 (dd, J = 5,3, 5,3 Hz, 2 H), 1,61-1 ,54 (m, 4 H), 1,47-1,42 (m, 2 H), 0,85 (d, J = 6,1 Hz, 6 H). Ein 5-((3R,5R)-3,5-Dimetilmorpholino)piridina-2-amina CB1 I-191 N-[4-[4-[[5-[(3R,5R)-3,5-Dimethylmorpholin-4-yl]-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridine - 2 -il]-3-fluor-phenyl]cyclo-hexane-carboxamide BicarbB EHC18 method, m/z = 571.3 [M + H]+, Ret. Time = 4.98 min. 1RMN-H (400 MHz, DMSO): δ 12,58 (s, 1 H), 11,65-11,63 (m, 1 H), 10,21 (s, 1 H), 8,91 (s , 1 H), 8,10 (d, J = 1,8 Hz, 1 H), 8,00 (dd, J = 9,2, 9,2 Hz, 1H), 7,77 (dd, J = 2,4, 9,0 Hz, 1 H), 7,51 (dd, J = 2,4, 9,0 Hz, 1 H), 7,47–7,39 (m, 2 H), 7, 06 (d, J = 8,3 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 3,83 (dd, J = 2,1, 10,2 Hz, 2 h). ), 3,52–3,49 (m, 2 H), 3,41 (dd, J = 5,8, 10,6 Hz, 2 H), 2,36–2,32 (m, 2 H) , 1,85–1,75 (m, 4 H), 1,44–1,38 (m, 2 Ein 5-((3R,5R)-3,5-Dimetilmorpholino)piridina-2-amina CB19 H), 1,30-1,24 (m, 3 H), 0,86 (d, J = 6,1 Hz, 6 H). I-192 2-(6-Fluoro-3-hydroxyindan-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 488 [M+H]+, Ret. time = 2.63 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (s, 1 H), 11,68 (d, J = 3,5 Hz, 1 H), 8,73 (s, 1 H), 8, 10 (d, J = 2,8 Hz, 1 H). ), 7,97 (d, J = 7,3 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,45 (dd, J = 5, 6, 5,6 Hz, 1 H), 7,25 (d, J = 9,1 Hz, 1 H), 7,07 (d, J = 9,1 Hz, 1 H), 6,64 (d , J = 7,3 Hz, 1 H), 5,45-5,38 (m, 1 H), 5,14 (dd, J = 5,8, 5,8 Hz, 1 H), 4,78 –4,69 (m, 1 H), 3,71–3,65 (m, 1 H), 3,41–3,35 (m, 2 H), 3,08–2,99 (m, 1 H) h), Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB35 2.94-2.81 (m, 3H), 2.49- 2,40 (m, 1 H), 1,94-1,83 (m, 3H), 1.60-1.49 (m, 2 h). I-193 2-[2-Fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl]-4-[[5-(4-methyl-piperazin-1-yl)-2-pyridyl]amino]-6H- Naphthyridine-5-eins BicarbB ECH18 method, m/z = 572[M+H]+, Ret. time = 4.19 min. 1RMN-H (400 MHz, DMSO): δ 12,51 (s, 1H), 11,72 (s, 1H), 8,80 (s, 1H), 8,12 (d, J = 3,0 Hz , 1H), 7,62 (d, J = 6,1 Hz, 1H), 7,53 (dd, J = 2,8, 9,1 Hz, 1H), 7,50–7,45 (m, 1H), 7,28 (d, J = 10,4 Hz, 1H), 7,09 (d, J = 9,1 Hz, 1H). ), 6,66 (d, J = 7,3 Hz, 1H), 3,91 (s, 3H), 3,70–3,60 (m, 2H), 3,24–3,18 (m, 6H), 2,60–2,40 (m, 4H), 2,28 (s, 3H), 1,69–1,60 (m, 4H), 1,52–1,47 (m, 2H) . Ein 5-(4-Methyl-piperazin-1-yl)pyridin-2-amine CB1 I-194 2-[2-Fluoro-5-methyl-4-(1-piperidyl-methyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridin-5-one BicarbB EHC18 method, m/z = 543 [M + H]+, Ret. Time = 4.85 min. 1RMN-H (400 MHz, DMSO): δ 12,41 (s, 1 H), 11,64 (d, J = 5,4 Hz, 1 H), 8,70 (d, J = 1,3 Hz , 1 H), 8,07 (d, J = 3,0 Hz, 1 H), 7,74 (d, J = 7,8 Hz, 1 H), 7,47 (dd, J = 3,1 , 9,0 Hz, 1 H), 7,43–7,39 (m, 1 H), 7,21 (d, J = 8,9 Hz, 1 H), 7,02 (d, J = 8 ,9 Hz, 1 H), 6,59 (d, J = 7,2 Hz, 1 H), 3,68-3,60 (m, 1 H), 3,57-3,48 (m, 2 H), 2,91-2,83 (m, 2 H), 2,43-2,36 (m, 6 H), 2,35 (s, 3 H), 1,87-1,79 (m , 2H), 1,56-1,38 (m, 9H). D 1-(6-amino-pyridin-3-yl) piperidin- 4-ol 4-bromo- 5-fluoro- 2- methyl-benzaldehyde Piperidine (Schritt 6) . I-195 4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-2-[3-methoxy-4-(piperidin-1-carbonyl)fenil]-6H-1,6-naphthiridina- 5-ein BicarbB EHC18 method, m/z = 555 [M + H]+, Ret. Time = 3.64 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (s, 1 H), 11,62 (d, J = 5,3 Hz, 1 H), 8,87 (s, 1 H), 8, 14 (d, J = 1,3 Hz, 1 H). ), 7,73 (s, 1 H), 7,61 (d, J = 7,6 Hz, 1 H), 7,48 (dd, J = 2,0, 9,1 Hz, 1 H), 7,41 (dd, J = 6,6, 6,6 Hz, 1 H), 7,30 (d, J = 7,8 Hz, 1 H), 7,04 (d, J = 8,3 Hz , 1 H), 6,61 (d, J = 7,1 Hz, 1 H), 4,69 (d, J = 3,5 Hz). , 1 H), 3,91 (s, 3 H), 3,67-3,64 (m, 2 H), 3,55- 3,50 (m, 3 H), 3,13 (s, 2 H), 2,91-2,82 (m, 2 H), 1,86-1,82 (m, 2 H), 1,61- A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB34 1,53 (m, 6 H), 1,49-1,47 (M, 2H). I-196 2-[5-Ethoxy-2-Fluoro-4-(Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-1, 6-naphthyridine-5-on AcHSS C18 method, m/z = 587.3 [M+H]+, Ret. time = 3.1 min. 1RMN-H (400 MHz, DMSO): δ 12,29 (s, 1 H), 11,52 (s, 1 H), 8,59 (s, 1 H), 7,92 (d, J = 2 ,3 Hz, 1 H), 7,41 (d , J = 5,8 Hz, 1 H), 7,33 (dd, J = 2,4, 8,7 Hz, 1 H), 7,27 ( d, J = 6,1 Hz, 1 H), 6,88 (d, J = 8,8 Hz, 1 H), 6,45 (d, J = 7,1 Hz, 1 H), 4,54 (s, 1 H), 3,99 (q, J = 6,7 Hz, 2 H), 3,43-3,37 (m, 3 H), 3,18 (s, 3 H), 3, 02 (s, 2 H), 2,75-2,68 (m, 2H), 1,69 (d, J = 9,6 Hz, 2 H), 1,47 (s, 2 H), 1, 42-1,30 (m, 6 H), 1,19 (dd, J = 6,9, 6,9 Hz, 3 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB9 I-197 N-[5-Fluor-2-metil-4-[5-oxo-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-2-yl]phenyl ] ciclohexancarboxamide AcHSS C18 method, m/z = 556.2 [M+H]+, Ret. time = 2.61 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,69–11,65 (m, 1 H), 9,32 (s, 1 H), 8,78 (s , 1 H), 8,27 (s, 1 H). ), 8,12 (d, J = 2,3 Hz, 1 H), 7,87 (d, J = 8,6 Hz, 1 H), 7,67 (d, J = 7,6 Hz, 1 H), 7,56–7,49 (m, 1 H), 7,46 (d, J = 7,6 Hz, 1 H), 7,08 (d, J = 8,8 Hz, 1 H) , 6,64 (d, J = 7,3 Hz, 1 H), 3,17 (s, 4 H), 2,98- 2,95 (m, 4 H), 2,33 (s, 3 H ), 1,86 (dd, J = 12,4, 32,1 Hz, 4 H), 1,74-1,69 (m, 2 H), 1,54-1,44 (m, 2 H) , 1,37–1,27 (m, 3 H). A and G tert-butyl-4-(6-amino-pyridin-3-yl)-piperazin-1-carboxy-lat CB36 I-198 4-[(5-Cyclopropyl-2-Pyridyl)-Amino]-2-[2-Fluoro-5-Methoxy-4-(Piperidin-1-Carbonyl)Phenyl]-6H-1,6-Naphthyridine-5-On BicarbB EHC18 method, m/z = 514 [M + H]+, Ret. Time = 4.85 min. 1RMN-H (400 MHz, DMSO): γ; 12,71 (s, 1 H), 11,80–11,79 (m, 1 H), 9,02 (s, 1 H), 8,28 (d, J = 1,5 Hz, 1 H) , 7,63 (d, J = 5,8 Hz, 1 H), 7,54–7,47 (m, 1 H), 7,31 (d, J = 8,3 Hz, 1 H), 7 ,07 (d, J = 8,3 Hz, 1 H), 6,69 (d, J = 7,3 Hz, 1 H), 3,91 (s, 3 H), 3,70–3,63 (m, 2 H), 3,21 (dd, J = 5,7, 5,7 Hz, 2 H), 2,03–1,95 (m, 2 H), 1,67-1,61 ( m, 4 H), 1,53 (d, J = 5,3 Hz, 2 H), 1,03-0,98 (m, 2 H), 0,77 (dd, J = 4,9, 4 ,9Hz, 2H). Ein 5-cyclopropyl-pyridin-2-amine CB1 I-199 2-[2- Fluor-5-methoxy-4-(piperidin-1-carbonyl)phenyl]-4- [(1- methyl-pyrazol-4-yl)amino]- 6H-1,6- naphthyridina- 5- one BicarbB EHC18 method, m/z = 477 [M + H]+, Ret. Time = 3.56 min. 1RMN-H (400 MHz, DMSO): δ 11,42 (d, J = 4,0 Hz, 1 H), 10,86 (s, 1 H), 7,75 (s, 1 H), 7, 38 (d, J = 3,8 Hz, 2 H). ), 7,22 (dd, J = 6,2, 6,2 Hz, 1 H), 7,02 (d, J = 7,3 Hz, 1 H), 6,87 (s, 1 H), 6,38 (d, J = 7,3 Hz, 1 H), 3,68 (d, J = 6,3 Hz, 6 H), 3,13–3,11 (m, 1 H), 2, 96 (dd, J = 5,6, 5,6 Hz, 3 H), 1,42 (dd, J = 4,7, 21,6 Hz, 4 H), 1,28 (s, 2 H). Ein 1-Methyl-1H-pyrazol-4-amine CB1 I-200 2-[6-Fluoro-3-(methyl-amino)indan- 5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6- Naphthyridine-5-one AcHSS C18 method, m/z = 501 [M+H]+, Ret. time = 2.08 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (s, 1 H), 8,72 (s, 1 H), 8,10 (d, J = 2,5 Hz, 1 H), 7, 99 (d, J = 7,3 Hz, 1 H). ), 7,52 (dd, J = 2,7, 9,0 Hz, 1 H), 7,45 (d, J = 7,3 Hz, 1 H), 7,06 (d, J = 9, 1 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,74-4,74 (m, 1 H), 4,26 (dd, J = 4,8, 6,8 Hz, 1 H), 3,68 (d, J = 1,8 Hz, 1 H), 3,56 (d, J = 12,4 Hz, 2 H), 3,11-3,02 (m, 1 H), 2,94-2,85 (m, 4 H), 2,44 (s, 4 H), 1,98- Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB37 1,88 (m, 5H), 1,61-1,50 (M, 2H). I-201 N-[4-[4-[[5-(3,3- Difluorazetidin-1-yl)-2-pyridyl]amino]-5-oxo-6H-1,6-naftiridin-2-yl]-3- Fluorphenyl]cyclohexancarboxamide BicarbB EHC18 method, m/z = 549 [M + H]+, Ret. Time = 5.01 min. 1RMN-H (400 MHz, DMSO): γ; 12,45 (s, 1 H), 11,63 (s, 1 H), 10,22 (s, 1 H), 8,76 (d, J = 1,0 Hz, 1 H), 8,05 –7,99 (m, 1 H), 7,82-7,73 (m, 2 H), 7,48-7,39 (m, 2 H), 7,17-7,07 (m, 2 H), 6,58 (d, J = 7,3 Hz, 1 H), 4,35 (t, J = 12,2 Hz, 4 H), 2,42–2,33 (m, 1 H) , 2,09 (s, 1 H), 1,86–1,77 (m, 4 H), 1,68 (d, J = 10,9 Hz, 1 Std.), 1,49–1,19 (m, 4 Est.). A 5-(3,3-difluoroazetidin-1-yl)pyridin-2-amine CB19 I-202 2-[2-Fluor-4-methoxy-5-(1-piperidil-metil)fenil]-4-[[5-(4-hidroxi-1-piperidil)-1-2-pyridyl]amino]-6H- 1,6-naphthyridin-5-one AcHSS C18 method, m/z = 559.3 [M+H]+, Ret. time = 2.2 min. 1RMN-H (400 MHz, DMSO): δ 12,37 (s, 1 H), 11,61 (d, J = 5,5 Hz, 1 H), 8,71 (s, 1 H), 8, 21 (s, 1 H), 8,05 (d , J = 3,0 Hz, 1 H), 7,99 (d, J = 9,4 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,42-7,38 (m, 1 H), 7,04- 7,01 (m, 2 H), 6,58 (d, J = 7 ,3 Hz, 1 H), 3,87 (s, 4 H), 3,69–3,61 (m, 3 H), 3,32 (s, 1 H), 2,92–2,83 ( m, 2 H), 2,41 (s, 4 H), 1,88-1,80 (m, 2 H), 1,56-1,36 (m, 8 H). D 1-(6-amino-pyridin-3-yl)piperidin-4-ol 5-Bromo-4-fluoro-2-methoxybenzaldehyde Piperidine (Schritt 6) . I-203 2-(4-Fluoro-1-hydroxyindan-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 488 [M+H]+, Ret. time = 2.59 min. 1RMN-H (400 MHz, DMSO): δ 12,27 (s, 1 H), 11,50 (d, J = 5,1 Hz, 1 H), 8,53 (s, 1 H), 7, 94 (d, J = 2,8 Hz, 1 H). ), 7,68 (dd, J = 7,3, 7,3 Hz, 1 H), 7,34 (dd, J = 28,9, 9,1 Hz, 1 H), 7,28 (dd, J = 6,3, 6,3 Hz, 1 H), 7,17 (d , J = 7,8 Hz, 1 H), 6,89 (d, J = 9,1 Hz, 1 H), 6 ,45 (d, J = 7,1 Hz, 1 H), 5,01 (dd, J = 6,6, 6,6 Hz, 1 H), 3,54-3,48 (m, 1 H) , 2,91 (ddd, J = 3,7, 8,8, 15,9 Hz, 1 H), 2,79-2,63 (m, 3 H), 2,38-2,33 (m, 5 H), 1,81-1,69 (m, 3 H), 1,43-1,31 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB38 I-204 N-[3-Fluoro-4-[4-[[5-(4-Methoxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]fenil ] ciclohexancarboxamide AcHSS C18 method, m/z = 571 [M+H]+, Ret. time = 3.96 min. 1RMN-H (400 MHz, DMSO): δ 12,39 (s, 1 H), 11,61 (d, J = 5,0 Hz, 1 H), 10,22 (s, 1 H), 8, 74 (s, 1 H), 8,09 (d , J = 3,0 Hz, 1 H), 8,00 (dd, J = 8,8, 8,8 Hz, 1 H), 7,76 ( d, J = 7,4 Hz, 1 H), 7,51–7,44 (m, 2 H), 7,40 (dd, J = 6,0, 7,2 Hz, 1 H), 7, 03 (d, J = 8,9 Hz, 1 H), 6,57 (d, J = 7,4 Hz, 1 H), 3,53–3,46 (m, 2 H), 3,42 - 3,24 (m, 3 H), 2,97-2,89 (m, 2 H), 2,42-2,34 (m, 2 H), 1,99-1,92 (m, 2 H) ), 1,86-1,76 (m, 4 Ein 5-(4-Methoxypiperidin-1-yl)pyridin-2-amine CB19 H), 1,69 (s, 1 H), 1,61- 1,51 (m, 2 H), 1,35-1,16 (M, 3H). I-205 N-[3-Fluor-4-[4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]fenil ]-3-methyl-oxetano-3-carboxamide AcHSS C18 method, m/z = 545 [M+H]+, Ret. time = 2.64 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,65 (s, 1 H), 10,18 (s, 1 H), 8,78 (s, 1 H) , 8,14–8,05 (m, 2H). ), 7,57-7,53 (m, 1 H), 7,83 (d, J = 7,3 Hz, 1 H), 7,50 (s, 1 H), 7,45 (d, J = 7,3 Hz, 1 H), 7,07 (d, J = 9,1 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4,91 (d, J = 6,1 Hz, 2 H), 4,74 (d, J = 4,0 Hz, 1 H), 4,43 (d, J = 6,1 Hz, 2 H), 3,73-3 ,65 (m, 1 H), 3,57 (d, J = 12,6 Hz, 2 H), 2,97-2,87 (m, 2 H), 1,88 (d, J = 9, 3 Hz, 2 H), 1,69 (s, 3 H), Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB39 1.59-1.49 (m, 2H). I-206 2-[2-Fluoro-5-methoxy-4-(pyrrolidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-naphthyridine- 5-eins AcHSS C18 method, m/z = 559 [M+H]+, Ret. time = 2.7 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,67 (s, 1 H), 8,73 (s, 1 H), 8,06 (d, J = 2 ,8 Hz, 1 H), 7,58 (d , J = 6,1 Hz, 1 H), 7,49-7,39 (m, 2 H), 7,24 (d, J = 8,8 Hz, 1 H), 7,02 (d, J = 8,8 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 4,73–4,63 (m, 1 H), 3,86 (s, 3 H), 3,67–3,60 (m, 1 H), 3,55–3,42 (m, 4 H), 3,19 (dd , J = 6,3, 6,3 Hz, 2 H), 2,91–2,82 (m, 2 H), 1,90–1,77 (m, 6 H), 1,54–1,43 (m , 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB40 I-207 N-[4-[4- [[5- [(3aR,6aS)-1,3,3a,4,6,6a-hexahydrofuro[3,4-c]pyrrol-5-yl]-2-pyridyl] amino]-5-oxo-6H-1,6-naphthyridin-2-yl]-3-fluoro-phenyl]cyclo-hexane-carboxamide AcHSS C18 method, m/z = 569 [M+H]+, Ret. time = 3.8 min. 1RMN-H (400 MHz, DMSO): δ 12,27 (s, 1 H), 11,58-11,56 (m, 1 H), 10,21 (s, 1 H), 8,60 (s , 1 H), 7,99 (dd, J = 8,7, 8,7 Hz, 1 H), 7,82 (d, J = 2,5 Hz, 1 H), 7,74 (dd, J = 8,7, 8,7 Hz, 1 H), 7,44 (dd, J = 1,6, 8,7 Hz, 1 H). ), 7,38 (d, J = 7,1 Hz, 1 H), 7,18 (dd, J = 2,9, 9,0 Hz, 1 H), 7,03 (d, J = 8, 8 Hz, 1 H), 6,55 (d, J = 7,3 Hz, 1 H), 3,90–3,83 (m, 2 H), 3,54 (dd, J = 3,0, 8,6 Hz, 2 H), 3,25–3,17 (m, 2 H), 3,00 (s, 2 H), 2,39 -2,32 (m, 2 Std.), Ein 5-((3aR,6aS)-Tetrahidro-1H-furo[3,4-c]pyrrol-5(3H)-il)piridina-2-amina CB19 1,81 (dd, J = 12,4, 24,0 Hz, 4 H), 1,66 (d, J = 10,6 Hz, 1 H), 1,47-1,36 (m, 2 H), 1,33–1,18 (m, 4 H). I-208 N-[5-Fluoro-2-methoxy-4-[5-oxo-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-2-yl]fenilo ] ciclohexancarboxamide AcHSS C18 method, m/z = 572 [M+H]+, Ret. time = 2.77 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 9,29 (s, 1 H), 8,80 (s, 1 H), 8,23 (s, 1 H) , 8,14 (d, J = 7,2 Hz). , 1 H), 8,08 (d, J = 3,0 Hz, 1 H), 7,66 (d, J = 7,2 Hz, 1 H), 7,49 (dd, J = 3,0 , 9,0 Hz, 1 H), 7,42 (d, J = 7,3 Hz, 1 H), 7,05 (d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 3,94 (s, 3 H), 3,15 (dd, J = 5,0, 5,0 Hz, 4 H), 2,95 (dd, J = 4,9, 4,9 Hz, 4 H), 2,71-2,62 (m, 1 H), 1,84-1,73 (m, 4 H), 1,67 (d, J = 11 ,2 Hz, 1 H), 1,46-1,17 (m, 6 H). A and G tert-butyl-4-(6-amino-pyridin-3-yl)piperazin-1-carboxylate CB66 I-209 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[[5-(4-methoxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridin-5-one BicarbB ECH18 method, m/z = 587.3 [M+H]+, Ret. time = 4.4 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,73-11,68 (m, 1 H), 8,75 (d, J = 1,4 Hz, 1 H ), 8,09 (d, J = 3,0 Hz, 1 H), 7,58 (d, J = 6,0 Hz, 1 H), 7,49 (dd, J = 3,1, 9, 0 Hz, 1 H), 7,44 (d, J = 6,9 Hz, 1 H), 7,24 (d, J = 8,9 Hz, 1 H), 7,04 (d, J = 8 ,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 3,87 (s, 3 H), 3,66-3,56 (m, 2 H), 3 ,53–3,45 (m, 2 H), 3,43–3,30 (m, 3 H), 3,17 (dd, J = 5,6, 5,6 Hz, 2 H), 2, 96–2,88 (m, 2 H), 1,99–1,91 (m, 2 Std.), 1,65-1,44 (m, 9 Std.). Ein 5-(4-methoxy-piperidin-1-yl)-pyridin-2-amine CB1 I-210 N-[3-Fluor-4-[4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]fenil ]-N,3-Dimethyl-oxetano-3-carboxamide AcHSS C18 method, m/z = 559.2 [M+H]+, Ret. time = 2.57 min. 1RMN-H (400 MHz, DMSO): δ 12,50 (s, 1 H), 11,70–11,70 (m, 1 H), 8,79 (s, 1 H), 8,14–8 ,04 (m, 2 H), 7,57–7,49 ( m, 2 H), 7,47 (d, J = 7,3 Hz, 1 H), 7,40 (d, J = 8, 1 Hz, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,89–4,81 (m , 2 H), 4,77–4,74 (m, 1 H), 4,01–4,01 (m, 2 H), 3,69 (s, 1 H), 3,57 (d, J = 12,1 Hz, 2 H), 3,21 (s, 3 H), 2,92 (dd, J = 10,0, 10,0 Hz, 2 H), 1,88 (d, J = 9 ,3 Hz, 2 H), 1,71 (s, 3 H), 1,60-1,48 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB41 I-211 N-[3-Fluor-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ]-1-methyl-cyclobutane-carboxamide AcHSS C18 method, m/z = 543 [M+H]+, Ret. time = 3.15 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,68–11,65 (m, 1 H), 9,78 (s, 1 H), 8,77 (s , 1 H), 8,12 (d, J = 2,5 Hz, 1 H), 8,05 (dd, J = 8,8, 8,8 Hz, 1 H), 7,86 (d, J = 8,6 Hz, 1 H), 7,61 (d, J = 8,6 Hz, 1 H), 7,52 (dd, J = 2,7, 9,0 Hz, 1 H), 7, 44 (d, J = 7,1 Hz, 1 H), 7,07 (d, J = 9,1 Hz, 1 H), 6,62 (d, J = 7,1 Hz, 1 H). ), 4,74 (d, J = 3,8 Hz, 1 H), 3,69 (dd, J = 4,0, 7,8 Hz, 1 H), 3,62–3,52 (m, 2 H), 2,92 (dd, J = 10,1, 10,1 Hz). , 2H), 2,53- A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB42 2,46 (m, 2 H), 2,03-1,84 (m, 5 H), 1,82-1,71 (m, 1 H), 1,53 (s, 5 H). I-212 N-[4-[4-[(5-Cyclopropyl-2-pyridyl)amino]-5-oxo-6H-1,6-nafthyridin-2-yl]-3-fluor-phenyl]cyclo-hexane-carboxamida AcHSS C18 method, m/z = 498 [M+H]+, Ret. time = 4.25 min. 1RMN-H (400 MHz, DMSO): δ 12,64 (s, 1 H), 12,57 (s, 1 H), 11,93–11,89 (m, 1 H), 8,54 (s , 1 H), 8,43 (s, 1 H). ), 8,18 (d, J = 2,5 Hz, 1 H), 8,13-8,08 (m, 1 H), 7,55-7,49 (m, 1 H), 7,12 -7,09 (m, 1 H), 7,05 (d, J = 9,1 Hz, 1 H), 6,53 (d, J = 7,1 Hz, 1 H), 4,75 (s , 1 H), 4,08 (d, J = 7,3 Hz, 1 H), 3,70-3,68 (m, 2 H), 3,61–3,56 (m, 2 H), 3,27 (s, 2 H), 2,93 (dd, J = 9,7, 9,7 Hz, 2 H), 2,04 Ein 5-Ciclopropil-Piridin-2-Amin CB19 (s, 1H), 1,90-1,85 (m, 2 H), 1,59-1,48 (m, 2 H), 1,35-1,21 (m, 2 H). I-213 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(1-methyl-2-oxo-3H-pyrrolo[2,3-b]pyridin-5-yl) -6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 484 [M+H]+, Ret. time = 2.4 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,68 (s, 1 H), 8,90 (s, 2 H), 8,31 (s, 1 H) , 8,21 (d, J = 2,5 Hz). , 1 H), 7,53 (dd, J = 2,9, 9,0 Hz, 1 H), 7,45 (d, J = 7,3 Hz, 1 H), 7,08 (d, J = 9,1 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,76-4,76 (m, 1 H), 3,77 (s, 2 H) , 3,74-3,67 (m, 1 H), 3,60 (d, J = 12,4 Hz, 2 H), 3,25 (s, 3 H), 2,98-2,91 ( m, 2 H), 1,91-1,88 (m, 2 H), 1,62-1,53 (m, 2 H). A 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl) -1,3- Dihydro-2H-pyrrolo[2,3- b]pyridine- 2-like I-214 N-[3-Fluor-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-nafthyridin-2-yl]phenyl ]-N,1-Dimethyl-cyclopropane-carboxamide AcHSS C18 method, m/z = 543.2 [M+H]+, Ret. time = 2.88 min. 1RMN-H (400 MHz, DMSO): δ 13,18–13,13 (m, 1 H), 12,52–12,51 (m, 1 H), 8,67 (s, 1 H), 8 ,21 (d, J = 2,3 Hz, 1 H) , 7,94 (dd, J = 8,5, 8,5 Hz, 1 H), 7,84–7,79 (m, 1 H) , 7,68 (d, J = 8,8 Hz, 1 H), 7,61–7,50 (m, 1 H), 7,26 (d, J = 8,8 Hz, 1 H), 6 ,79 (d, J = 7,1 Hz, 1 H), 3,74–3,69 (m, 1 H), 3,66–3,60 (m, 2 H), 3,00 (dd, J = 10,4, 10,4 Hz, 2 H), 1,89–1,83 (m, 2 H), 1,58–1,47 (m, 2 H), 1,20–1,03 (m, 8 H), 0,60 (dd, J = 5,4, 5,4 Hz). , 4ª.). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB43 I-215 2-[4-[(Dimetilamino)metil]-2-fluor-5-methoxy-fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridine-5-uno AcHSS C18 method, m/z = 519.2 [M+H]+, Ret. time = 2.05 min. 1RMN-H (400 MHz, DMSO): δ 12,41 (s, 1 H), 11,66 (d, J = 5,5 Hz, 1 H), 8,73 (d, J = 1,3 Hz , 1 H), 8,23 (s, 1 H). ), 8,08 (d, J = 3,0 Hz, 1 H), 7,53 (d, J = 6,4 Hz, 1 H), 7,48 (dd, J = 3,1, 9, 0 Hz, 1 H), 7,44–7,40 (m, 1 H), 7,03 (d, J = 8,8 Hz, 1 H), 6,61 (d, J = 7,3 Hz , 1 H), 3,86 (s, 4 H), 3,68–3,60 (m, 2 H), 3,57–3,49 (m, 3 H), 2,92-2,83 (m, 2 H), 2,23 (s, 6 H), 1,88-1,80 (m, 2 H), 1,56-1,45 (m, 2 H). D 1-(6-Amino-pyridin-3-yl)piperidin-4-ol I-216 2-[2-Fluor-5-methoxy-4-(1-piperidyl-metil)fenil]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1, 6-naphthyridin-5-one AcHSS C18 method, m/z = 559.3 [M+H]+, Ret. time = 2.18 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,69 (d, J = 4,5 Hz, 1 H), 8,78 (s, 1 H), 8, 21 (s, 1 H), 8,12 (d , J = 2,3 Hz, 1 H), 7,57 (d, J = 6,3 Hz, 1 H), 7,52 (dd, J = 2,8, 2,8 Hz, 1 H), 7,49–7,43 (m, 1 H), 7,31 (d, J = 8,8 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 3,89 (s, 3 H), 3,73-3,64 (m , 2 H ), 2,91 (dd, J = 10,1, 10,1 Hz, 2 H), 2,53-2,33 (m, 6 H), 1,88 (d, J = 10,4 Hz, 2 H), 1,64-1,42 (m, 9 H). D 1-(6-amino-pyridin-3-yl)piperidin-4-ol piperidine (Stage 6) I-217 2-[2- Fluor-5-(methyl-amino-methyl)phenyl]-4-[[5-(4- hydroxy-1-piperidyl)-2-pyridyl]amino]-6H- 1,6- naphthyridina- 5-ein AcHSS C18 method, m/z = 238 [M+H]+, Ret. time = 2 min. 1RMN-H (400 MHz, DMSO): δ 12,47 (s, 1 H), 8,75 (s, 1 H), 8,34 (s, 1 H), 8,11 (d, J = 2 ,5 Hz, 1 H), 7,98 (d , J = 7,3 Hz, 1 H), 7,55-7,51 (m, 2 H), 7,47 (d, J = 7,3 Hz, 1 H), 7,39-7,33 (m, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 3,84 (s, 3 H), 3,71–3,65 (m, 2 H), 3,22–3,21 (m, 1 H), 2,96–2,88 ( m, 2 H), 2,38 (s, 3 H), 2,14 (s, 1 H), 1,88 (d, J = 8,3 Hz, 2 H), 1,58–1,50 (m, 2H). A and G 1-(6-aminopyridin-3-yl)piperidin-4-ol CB68 I-218 2-[2-Fluor-4-(methyl-amino-methyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-naphthiridin-5-one AcHSS C18 method, m/z = 475 [M+H]+, Ret. time = 1.94 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 8,74 (s, 1 H), 8,11 (d, J = 2,4 Hz, 1 H), 7, 95 (dd, J = 7,9, 7,9 Hz, 1 H), 7,52 (dd, J = 2,6, 8,7 Hz, 1 H), 7,45 (d, J = 7, 3 Hz, 1 H), 7,36–7,29 (m, 2 H), 7,06 (d, J = 8,9 Hz). , 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4,76-4,75 (m, 1 H), 3,76 (s, 2 H), 3,72- 3,67 (m, 1 H), 3,57 (d, J = 12,6 Hz, 4 H), 2,91 (dd, J = 9,9, 9,9 Hz, 2 H), 2, 34 (s, 3 H), 1,90–1,86 A and G 1-(6-aminopyridin-3-yl)piperidin-4-ol CB44 (m, 2H), 1.60-1.50 (m, 2 h). I-219 N-[3-Fluor-4-[5-Oxo-4-[(5-Piperazin-1-yl-2-Pyridyl)Amino]-6H-1,6-Naphtiridin-2-yl]Phenyl]-1- Methyl-cyclobutano-carboxamide AcHSS C18 method, m/z = 528 [M+H]+, Ret. time = 2.35 min. 1RMN-H (400 MHz, DMSO): δ 12,40 (s, 1 H), 9,75 (s, 1 H), 8,75 (s, 1 H), 8,07–7,99 (m , 2 H), 7,80 (d, J = 7,3 Hz, 1 H), 7,57 (dd, J = 2,0, 8,7 Hz, 1 H), 7,47 (dd, J = 3,1, 9,0 Hz, 1 H), 7,41 (d, J = 7,3 Hz, 1 H), 7,04 (d, J = 8,9 Hz, 1 H), 6, 58 (d, J = 7,3 Hz, 1 H), 3,10–3,05 (m, 4 H), 2,88–2,83 (m, 4 H), 2,49–2,41 (m, 4 H), 2,01-1,83 (m, 3 H), 1,78-1,69 (m, 1 H), 1,48 (s, 3 H). A and G tert-butyl-4-(6-aminopyridin-3-yl)-piperazin-1-carboxylate CB42 I-220 2-(6-Fluoro-1,2,3,4-Tetrahydroisoquinolin-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]-amino]-6H- 1,6-naphthyridin-5-one BicarbB EHC18 method, m/z = 487 [M + H]+, Ret. Time = 2.79 min. 1RMN-H (400 MHz, DMSO): γ; 12,52 (s, 1 H), 8,33 (s, 1 H), 8,04 (d, J = 3,0 Hz, 1 H), 7,48–7,40 (m, 2 H) , 7,18 -7,05 (m, 2 H), 7,00-6,97 (m, 1 H), 6,53 (d, J = 7,3 Hz, 1 H), 4,69- 4,66 (m, 1 H), 3,90-3,87 (m, 2 H), 3,65-3,62 (m, 2 H), 2,89-2,80 (m, 6 H ), 2,68 (dd, J = 1,7, 3,7 Hz, 1 H), 2,18-2,12 (m, 2H), 1,84-1,78 (m, 2 H), 1,53–1,43 (m, 2 H). A and G 1-(6-aminopyridin-3-yl)piperidin-4-ol CB45 I-221 2-[2-Fluoro-5-[1-(methylamino)cyclopropyl]phenyl]-4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6 - Naphthyridine -5-one AcHSS C18 method, m/z = 501 [M+H]+, Ret. time = 2.12 min. 1RMN-H (400 MHz, DMSO): δ 9,90 (s, 1 H), 8,79 (s, 1 H), 8,58 (d, J = 8,5 Hz, 1 H), 8, 39 (d, J = 6,9 Hz, 1 H). ), 8,05 (d, J = 2,9 Hz, 1 H), 7,85 (s, 1 H), 7,70 (dd, J = 8,8, 16,9 Hz, 2 H), 7,49 (dd, J = 3,0, 9,0 Hz, 1 H), 7,34–7,30 (m, 1 H), 7,01 (d, J = 8,9 Hz, 1 H ), 6,95 (dd, J = 6,3, 6,3 Hz, 1 H), 4,40 (s, 2 H), 3,43 -3,21 (m, 6 H), 3,18 (s, 2 H), 2,94 (s, 4 H), 2,57 (s, 3 H). A and G 1-(6-aminopyridin-3-yl)piperidin-4-ol CB69 I-222 1-(Difluoromethyl)-N-[3-fluoro-[4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1, 6- Naftiridin-2-yl]phenyl]cyclopropanecarboxamida AcHSS C18 Method, m/z 565 [M+H]+, Ret. time = 3.08 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 8,77 (s, 1 H), 8,12 (d, J = 2,5 Hz, 1 H), 8, 06 (dd, J = 8,8, 8,8 Hz, 1 H), 7,79 (dd, J = 7,2, 7,2 Hz, 1 H), 7,60 (d, J = 8, 3 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,45 (d , J = 7,3 Hz, 1 H), 7,07 (d , J = 8,8 Hz, 1 H), 6,66–6,60 (m, 2 H), 4,74 (d, J = 4,0 Hz, 1 H), 3,70–3,66 (m, 2 H), 3,60–3,53 (m, 2 H), 2,95–2,87 (m, 2 H), 2,73 (s, 1 H), 1,88 (d , J = 8,6 Hz, 2 H), 1,55 (q , J = 9,0 Hz, 2H), 1,44 (s, 2H), 1,24–1,18 (m, 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB47 I-223 1-ethyl-N-[3-fluoro-4-[4-[[5-(4-hidroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthyridine-2 -yl]phenyl]cyclopropanecarboxamide AcHSS C18 method, m/z = 543 [M+H]+, Ret. time = 3.13 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,65 (d, J = 4,5 Hz, 1 H), 9,65 (s, 1 H), 8, 76 (s, 1 H), 8,12 (d , J = 2,8 Hz, 1 H), 8,03 (dd, J = 9,0, 9,0 Hz, 1 H), 7,82 ( d, J = 8,8 Hz, 1 H), 7,63 (dd, J = 1,5, 8,6 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz , 1 H), 7,44 (dd, J = 6,4, 6,4 Hz, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4,74–4,74 (m, 1 H), 3,72–3,66 (m, 1 H), 3,61–3,53 (m, 2 H ), Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB48 2.97-2.87 (m, 2H), 1.92- 1,87 (m, 2 H), 1,78 (q, J = 7,2 Hz, 2 H), 1,60-1,49 (M, 2H), 1.18-1.14 (M, 2 H), 1,01 (dd, J = 7,3, 7,3 Hz, 3 H), 0,76–0,71 (m, 2 h). I-224 2-[6-Fluoro-1-(methyl-amino)tetralin-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6- Naphthyridine-5-one AcHSS C18 method, m/z = 515.2 [M+H]+, Ret. time = 2.04 min. 1HRMN (400MHz, DMSO) δ1RMN-H (400 MHz, DMSO): δ 12,52 (s, 1 H), 8,31 (s, 1 H), 8,05–8,03 (m, 1 H), 7,53–7 ,39 (m, 2 H), 7,18–7,05 (m, 2 h), 7,00–6,96 (m, 1 h), 6,54–6,52 (m, 1 h) , 4,69–4,66 (m, 1 h), 3,64–3,60 (m, 2 h), 2,88–2,80 (m, 2 H), 2,68 (dd, J = 1,7, 3,7 Hz, 1 H), 2,35–2,34 (m, 6 H), 2,18–2,12 (m, 1 H), 1,83–1,72 ( m, 3 H), 1,57–1,42 (m, 4 H), 1,27–1,23 (m, 2 H). A and G 1-(6-aminopyridin-3-yl)piperidin-4-ol CB49 I-225 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-4-[(5-THF-3-yl-1H-pyrazol-3-yl)amino]-6H-1, 6-naphthyridin-5-one AcHSS C18 method, m/z = 533 [M+H]+, Ret. time = 2.99 min. 1HRMN (400MHz, DMSO) δ1RMN-H (400 MHz, DMSO): δ 12,46–12,41 (m, 1 H), 12,03–12,00 (m, 1 H), 11,65 (d, J = 5,3 Hz, 1 H), 8,30–8,28 (m, 1 H), 7,61 (d, J = 6,0 Hz, 1 H), 7,42 (dd, J = 5,9, 7 ,0 Hz, 1 H), 7,26–7,23 (m, 1 H), 6,60 (d, J = 7,2 Hz, 1 H), 6,05 (s, 1 H), 4 ,01 (t, J = 7,7 Hz, 1 H), 3,87-3,86 (m, 4 H), 3,66-3,56 (m, 2 H), 3,49-3, 42 (m, 3 H), 3,18–3,13 (m, 2 H), 2,35–1,96 (m, 2 H), 2,05–1,96 (m, 2 H), 1,64–1,56 (m, 2 H), 1,50–1,43 (m , 2 Std.). Ein 5-(THF-3-yl)-1H-pyrazol-3-amine CB1 I-226 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(2-oxo-1,3-di-hydro-pyrrolo[2,3-b]pyridin- 5- il) -6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 470 [M+H]+, Ret. time = 2.23 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,62 (d, J = 4,4 Hz, 1 H), 11,24 (s, 1 H), 8, 82 (s, 1 H), 8,79 (d , J = 2,0 Hz, 1 H), 8,23 (d, J = 1,6 Hz, 1 H), 8,17 (d, J = 2,9 Hz, 1 H), 7,50 (dd, J = 3,1, 9,0 Hz, 1 H), 7,41 (dd, J = 5,2, 6,8 Hz, 1 H) , 7,05 (d, J = 8,9 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 4,72 (d, J = 1,6 Hz, 1 H ), 3,68 (s, 2 H), 3,66 (s, 1 H), 3,57-3,51 (m, 2 H), 2,94-2,85 (m, 2 A 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-5-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl)-1 ,3 - Dihydro- H), 1,92-1,83 (m, 2 H), 2H- 1.58-1.47 (m, 2H). Pyrrole[2,3- b]pyridine- 2-like I-227 4-[[5-(4-Methyl-piperazin-1-yl)-2-pyridyl]amino]-2-(2-oxo-1,3-dihydro-pyrrolo[2,3-b]pyridin- 5- il)-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 469 [M+H]+, Ret. time = 1.76 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,6 (s, 1 H), 11,3 (s, 1 H), 8,84 (d, J = 5 ,3 Hz, 1 H), 8,79 (d , J = 2,1 Hz, 1 H), 8,23 (d, J = 2,1 Hz, 1 H), 8,16 (d, J = 3,0 Hz, 1 H), 7,50 (dd, J = 3,0, 8,9 Hz, 1 H), 7,42 (d, J = 7,3 Hz, 1 H), 7,07 (d, J = 8,9 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 3,42–3,36 (m, 2 H), 3,18 (dd , J = 4,9, 4,9 Hz, 4 H), 2,48 (d, J = 5,0 Hz, 4 H), 2,25 (s, 3 H). Ein 5-(4-Methyl-piperazin-1-yl)pyridin-2-amine 5-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl)-1,3 - Di-hydro-2H-pyrrolo[2,3-b]pyridin-2-on I-228 4-[(1-Ethyl-2-oxopyrimidin-4- yl)amino]-2-[2-Fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl]-6H-1,6- Naphthyridine- 5 em BicarbB EHC18 method, m/z = 519 [M + H]+, Ret. Time = 3.49 min. 1RMN-H (400 MHz, DMSO): δ 12,98 (s, 1H), 11,94 (s, 1H), 9,35 (s, 1H), 8,01 (d, J = 7,1 Hz , 1H), 7,58–7,49 (m, 2H). ), 7,27 (d, J = 10,1 Hz, 1H), 6,73 (d, J = 7,3 Hz, 1H), 6,13 (d, J = 7,1 Hz, 1H), 3,86 (s, 3H), 3,81 (q, J = 7,0 Hz, 2H), 3,66–3,54 (m, 2H), 3,19–3,13 (m, 2H) , 1,61–1,56 (m, 4H), 1,48–1,44 (m, 2H), 1,22 (t, J = 7,1). hercios, 3H). C 4-Amino- 1-ethyl-pyrimidine- 2(1H)-enenos CB1 I-229 4-[[5-[(3aR,6aS)-1,3,3a,4,6,6a-Hexahydrofuro[3,4-c]pyrrol-5-yl]-2-pyridyl]amino]-2-[ 2-Fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl]-6H-1,6-naphthyridin-5-on AcHSS C18 method, m/z = 585 [M+H]+, Ret. time = 3.38 min. 1RMN-H (400 MHz, DMSO): δ 12,35-12,32 (m, 1 H), 11,64 (d, J = 1,5 Hz, 1 H), 8,62 (s, 1 H ), 7,82 (d, J = 2,5 Hz, 1 H), 7,56 (d, J = 6,1 Hz, 1 H), 7,41 (d, J = 6,8 Hz, 1 H), 7,22-7,15 (m, 2 H), 7,04 (d, J = 8,8 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 2 H) , 3,85 (s, 3 H), 3,62–3,51 (m, 5 H), 3,24–3,14 (m, 6 H), 3,00 (d, J = 1,5 Hz, 2 H), 1,65-1,54 (m, 4 H), 1,45 (d, J = 5,6 Hz, 2 H). Ein 5-((3aR,6aS)-Tetrahidro-1H-furo[3,4-c]pyrrol-5(3H)-il)piridina-2-amina CB1 I-230 5-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6-naphthiridin-2-yl]-2-methoxy - N,N-Dimethylbenzamide AcHSS C18 method, m/z = 533 [M+H]+, Ret. time = 2.59 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,67 (s, 1 H), 8,73 (s, 1 H), 8,06 (d, J = 2 ,8 Hz, 1 H), 7,58 (d , J = 5,8 Hz, 1 H), 7,49-7,39 (m, 2 H), 7,22 (d, J = 9,1 Hz, 1 H), 7,02 (d, J = 9,1 Hz, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 3,86 (s, 3 H), 3,66–3,60 (m, 1 H), 3,56–3,49 (m, 2 H), 2,99 (s, 3 H), 2,87 (d, J = 10,1 Hz , 2 H), 2,82 (s, 4 H), 1,82 (d, J = 9,6 Hz, 2 H), 1,54–1,43 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB50 I-231 2-[2,5- Difluoro-4-(piperidine-1-carbonyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H- 1,6- Naphthyridine-5-on AcHSS C18 method, m/z = 561 [M + H]+, Ret. Time = 3 min. 1RMN-H (400 MHz, DMSO): δ 12,49 (s, 1 H), 11,76-11,64 (m, 1 H), 8,80 (d, J = 1,3 Hz, 1 H ), 8,08 (d, J = 3,0 Hz, 1 H), 7,88 (dd, J = 6,0, 9,7 Hz, 1H), 7,52-7,43 (m, 3 H), 7,06-7,03 (m, 1 H), 6,60 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz, 1 H) , 3,66–3,58 (m, 3 H), 3,56–3,50 (m, 2 H), 2,92–2,84 (m, 2 H), 1,88–1,81 (m, 2 Std.), 1,68–1,46 (m, 10 Std.). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB51 I-232 2-[2- Fluor-5-methoxy-4-(piperidin- 1-carbonyl)fenil]-4-[[5-(1-piperidyl)-2-pyridyl]amino]-6H- 1,6- naphthiridina- 5-ein AcHSS C18 method, m/z = 557 [M+H]+, Ret. time = 3.98 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 8,74 (s, 1 H), 8,07 (d, J = 2,9 Hz, 1 H), 7, 57 (d, J = 6,0 Hz, 1 H). ), 7,50-7,42 (m, 2 H), 7,24 (d, J = 8,9 Hz, 1 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 3,87 (s, 3 H), 3,66-3,56 (m, 2 H), 3,19-3,12 (m , 6 H), 1,67-1,45 (m, 12 H). Ein 5-(Piperidin-1-yl)pyridin-2-amine CB1 I-233 2-[2-(cyclo-propyl-carbonyl)--2,8-diazaspiro[4.5]decan-8-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino ] -6H-1,6-Nafthyridine-5-on AcHSS C18 method, m/z = 544.3 [M+H]+, Ret. time = 2.57 min. 1RMN-H (400 MHz, DMSO) δ 12,26 (d, J = 2,6 Hz, 1H), 11,15 (s, 1H), 8,04 (dd, J = 2,4, 2,4 Hz, 1H), 7,97 (d, J = 4,9 Hz). , 1H), 7,44 (dd, J = 3,0, 8,9 Hz, 1H), 7,25–7,19 (m, 1H), 6,90 (d, J = 9,0 Hz, 1H), 6,24 (dd, J = 1,6, 7,2 Hz, 1H), 4,71-4,70 (m, 1H), 3,78-3,41 (m, 10H), 3 ,26 (s, 1H), 2,89-2,80 (m, 2H), 1,92-1,75 (m, 5H), 1,63-1,46 (m, 6H), 0,76 -0,70 (m, 4H). P 1-(6-amino-pyridin-3-yl)piperidin-4-ol tert-butyl-2,8-diaza-spiro[4,5]decane-2-carboxylate I-234 2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)phenyl]-4- [(6-piperazin-1-yl-2-pyridyl)amino]-6H- 1,6- naphthiridina- 5-ein AcHSS C18 method, m/z = 558 [M+H]+, Ret. time = 2.43 min. 1RMN-H (400 MHz, MeOD) δ 9,00 (d, J = 2,3 Hz, 1 H), 7,44–7,38 (m, 2 H), 7,28 (d, J = 7 ,3 Hz, 1 H), 7,11–7,05 (m , 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 6,32 (d, J = 8,3 Hz, 1 H), 6,22 (d, J = 7,6 Hz, 1 H), 3,83 (s, 3 H), 3,63 (d, J = 4,8 Hz, 2 H), 3,44–3,37 (m, 4 H), 2,79 (dd, J = 4,9, 4,9 Hz, 4 H), 2,61–2,57 (m, 1 H), 1 ,62-1,55 (m, 6 H), 1,53-1,44 (m, 4 H). A and G tert-butyl-4-(6-aminopyridin-2-yl)piperazin-1-carboxylate CB1 I-235 2-(2,8-Diazaspiro[4.5]decan-8-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-naphthyridin-5-one AcHSS C18 method, m/z = 476 [M+H]+, Ret. time = 1.85 min. 112.31 (s, 1H), 11.21 (s, 1H), 8.43 (s, 1H), 8.08 (d, J=2.5Hz, 1H), 7.99 (s, 1H ), 7.48 (dd, J=2.5, 8.8Hz), 1H), 7.26 (d, J=7.3Hz, 1H), 6.93 (d, J=8.8 Hz, 1H), 6.28 (d, J=7.3 Hz, 1H), 4.75 (s, 1H), 3.75-3.49 (m, 6H), 3.23 (s, 1H ), 3.19 (t, J = 7.0 Hz, 2H), 2.97 (s, 2H), 2.90 (t, J = 11.2 Hz, 2H), 1.90–1.77 (m, 4H), 1.66–1.51 (m, 6H). (1 Äq. Formiatsalz, 1 austauschbares Proton nicht beobachtet) P 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-tert-butyl-2,8-diazaspiro[4.5]decan-2-carboxylate I-236 2-(1,8-Diazaspiro[4.5]decan-8-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5 -eins AcHSS C18 method, m/z = 476 [M+H]+, Ret. time = 1.89 min. 1RMN-H (400 MHz, DMSO) δ 12,29 (s, 1H), 11,16 (s, 1H), 8,08 (d, J = 2,3 Hz, 1H), 7,98 (s, 1H), 7,48 (dd, J = 2,8, 8,8 Hz, 1H), 7,25 (d, J = 7,3 Hz, 1H), 6,93 (d, J = 8,8 Hz, 1H), 6,27 (d, J = 7,1 Hz, 1H), 4,73 (d, J = 4,0 Hz). , 1H), 3,76-3,65 (m, 5H), 3,52 (d, J = 12,1 Hz, 2H), 2,93-2,86 (m, 4H), 1,92- 1,85 (m, 2H), 1,79-1,72 (m, 2H), 1,61-1,55 (m, 9H). P 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-tert-butyl-1,8-diaza-spiro[4.5]decane-1-carboxylate I-237 2-[1-(Dimethylamino)-4-fluoro-indan-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6 - Naphthyridine -5-en AcHSS C18 method, m/z = 515 [M+H]+, Ret. time = 2.07 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,66 (d, J = 1,3 Hz, 1 H), 8,71 (s, 1 H), 8, 11 (d, J = 2,5 Hz, 1 H). ), 7,62 (d, J = 7,1 Hz, 1 H), 7,42 (dd, J = 2,9, 9,0 Hz, 1 H), 7,35 (d, J = 7, 3 Hz, 1 H), 7,18 (d, J = 8,8 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7 ,3 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 4,30 (dd, J = 7,1, 7,1 Hz, 1 H), 3,71- 3,66 (m, 1 H), 3,61-3,52 (m, 2 H), 3,01-2,84 (m, 4 H), 2,24 (s, 6 H), 2, 11 ( q, J = 7,3 Hz, 2 H), 1,88 (d, J = 9,1 Hz, 2 H), 1,60–1,49 (m, 2 H). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB52 I-238 2-[1-(Dimethylamino)-6-fluoro-indan-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6 - Naphthyridine -5-en AcHSS C18 method, m/z = 515 [M+H]+, Ret. time = 2.03 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 11,66 (d, J = 1,3 Hz, 1 H), 8,71 (s, 1 H), 8, 11 (d, J = 2,5 Hz, 1 H). ), 7,82 (d, J = 7,1 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,45 (d, J = 7, 3 Hz, 1 H), 7,18 (d, J = 8,8 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7 ,3 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 4,39 (dd, J = 7,1, 7,1 Hz, 1 H), 3,71- 3,66 (m, 1 H), 3,61-3,52 (m, 2 H), 3,01-2,84 (m, 4 H), 2,24 (s, 6 H), 2, 11 ( q, J = 7,3 Hz, 2 H), 1,88 (d, J = 9,1 Hz, 2 H), 1,60–1,49 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB53 I-239 2-(2,9-Diazaspiro[5.5]undecan-9-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridin-5 -eins AcHSS C18 method, m/z = 490 [M+H]+, Ret. time = 1.97 min. 1H-NMR (400 MHz, DMSO) 12,25 (1H, s), 11,11 (1H, s), 8,04 (1H, d, J = 3,0 Hz), 7,92 (1H, s ), 7,44 (1H, dd, J = 3,1, 9,0 Hz), 7,21 (1H, d, J = 7,3 Hz), 6,89 (1H, d, J = 8, 9 Hz), 6,23 (1H, d, J = 7,3 Hz), 4,70 (1H, d, J = 4,3 Hz), 3,70–3,46 (8H, m), 2 ,89–2,80 (2H, m), 2,69–2,66 (2H, m), 2,59–2,56 (2H, m), 1,88–1,81 (2H, m) , 1,56– 1,43 (10H, m). P 1-(6-Amino-pyridin-3-yl)piperidin-4-ol-tert-butyl-2,9-diazaspiro[5.5]undecan-2-carboxylate I-240 1-Ethyl-N-[3-fluoro-4-[5-oxo-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-nafthyridin-2-yl]phenyl ] cyclobutanecarboxamide AcHSS C18 method, m/z = 542 [M+H]+, Ret. time = 2.5 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 9,76 (s, 1 H), 8,78 (s, 1 H), 8,10 (d, J = 2 ,5 Hz, 1 H), 8,04 (dd , J = 8,8, 8,8 Hz, 1 H), 7,86 (d, J = 7,3 Hz, 1 H), 7,64- 7,58 (m, 1 H), 7,51 (dd, J = 2,9, 9,0 Hz, 1 H), 7,45 (d, J = 7,3 Hz, 1 H), 7, 08 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 3,11 (dd, J = 4,7, 4,7 Hz, 4 H), 2,89 (dd, J = 4,7, 4,7 Hz, 4 H), 2,49–2,38 (m, 3 H), 1,98–1,90 (m, 6 H ), 1,83–1,73 (m, 1 H), 0,83 (dd, J = 7,3, 7,3 Hz, 3 H). A and G tert-butyl-4-(6-aminopyridin-3-yl)-piperazin-1-carboxylate CB54 I-241 2-[2,3- Difluor-5-methoxy-4-(piperidin- 1-carbonyl)fenil]-4-[[5-(4-hidroxi-1-piperidyl)-2-pyridyl]amino]-6H- 1,6-naphthyridin-5-one AcHSS C18 method, m/z = 591 [M+H]+, Ret. time = 3.1 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,71 (s, 1 H), 8,77 (d, J = 1,4 Hz, 1 H), 8, 10 (d, J = 3,0 Hz, 1 H). ), 7,51–7,43 (m, 2 H), 7,40 (dd, J = 1,6, 5,0 Hz, 1 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,71 (d, J = 4,1 Hz, 1 H), 3,89 (s, 3 H), 3, 69–3,59 (m, 2 H), 3,58–3,50 (m, 3 H), 3,24–3,22 (m, 2 H), 2,92-2,48 (m, 2 H), 1,88-1,80 (m, 2 H), 1,67-1,54 (m, 4 H), 1,52-1,44 (m, 4 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB10 I-242 6-[[2-[2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)fenil]-5-oxo-6H-1,6-naphthiridin-4-yl]amino]pyridin-3-carbonitrile BicarbB EHC18 method, m/z = 499 [M + H]+, Ret. Time = 4.34 min. 1RMN-H (400 MHz, DMSO): δ 13,29 (s, 1 H), 11,97 (s, 1 H), 9,15 (s, 1 H), 8,89 (s, 1 H) , 8,24–8,20 (m, 1 H). ), 7,64 (d, J = 5,8 Hz, 1 H), 7,56 (d, J = 7,3 Hz, 1 H), 7,32–7,25 (m, 2 H), 6,76 (d, J = 7,3 Hz, 1 H), 3,92 (s, 3H), 3,71–3,61 (m, 2H), 3,22 (dd, J = 5,4 , 5,4 Hz, 2H), 1,75–1,44 (m, 6H). Ein 6-aminonicotinonitrilo CB1 I-243 2-(2-Fluoro-5-pyrrolidin-2-yl-phenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine-5 -eins AcHSS C18 method, m/z = 501 [M+H]+, Ret. time = 2.09 min. 1RMN-H (400 MHz, DMSO): δ 12,4 (s, 1 H), 11,97 (s, 1 H), 8,72 (d, J = 1,5 Hz, 1 H), 8, 23 (s, 1 H), 8,06 (d , J = 2,9 Hz, 1 H), 8,01 (dd, J = 2,1, 7,4 Hz, 1 H), 7,60– 7,57 (m, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,45–7,38 (m, 2 H), 7,03 (d , J = 8,8 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 3,67–3,60 (m, 1 H), 3,55–3,49 (m, 2 h), 3,23–3,12 (m, 2 h), 2,91–2,83 (m, 2 h), 2,35–2,29 (m, 2 h), 1 ,94–1,79 (m, 6 h), 1,54–1,45 (M, 2H). A and G 1-(6-aminopyridin-3-yl)piperidin-4-ol CB70 I-244 2-[3-(Dimethylamino)-6-fluoro-indan-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6 - Naphthyridine -5-en AcHSS C18 method, m/z = 515.2 [M+H]+, Ret. time = 2.12 min. 1RMN-H (400 MHz, DMSO) δ 12,43 (s, 1H), 11,69–11,66 (m, 1H), 8,75 (s, 1H), 8,10 (d, J = 2 ,5 Hz, 1H), 7,90 (d, J = 7,3 Hz, 1H), 7,52 (dd, J = 2,9, 9,0 Hz, 1H), 7,46 (d, J = 3,0 Hz, 1H), 7,24 (d, J = 8,8 Hz, 1H), 7,06 (d, J = 8,8 Hz, 1H), 6,64 (d, J = 7 ,3 Hz, 1H), 4,77–4,74 (m, 1H), 4,35 (dd, J = 6,4, 6,4 Hz, 1H), 3,72–3,68 (m, 1H), 3,57 (d, J = 12,1 Hz, 2H), 3,01-2,88 (m, 4H), 2,21 (s, 6H), 2,16- A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB4 2.04 (metro, 2H), 1.90-1.86 (m, 2H), 1.60-1.49 (m, 2H). I-245 2-[2- Fluor-5-methoxy-4-(piperidin- 1-carbonyl)fenil]-4-[[5-(4-piperidyl)-2-pyridyl]amino]-6H- 1,6- naphthiridina- 5-ein AcHSS C18 method, m/z = 557 [M+H]+, Ret. time = 2.41 min. 1RMN-H (400 MHz, DMSO): δ 12,75 (s, 1 H), 9,06 (s, 1 H), 8,32 (s, 1 H), 7,72 (dd, J = 2 ,1, 8,5 Hz, 1 H), 7,64 (d, J = 5,8 Hz, 1 H), 7,51 (d, J = 7,3 Hz, 1 H), 7,12 ( d, J = 8,6 Hz, 1 H), 6,69 (d, J = 7,3 Hz, 1 H), 3,92 (s, 3 H), 3,69-3,60 (m, 2 H), 3,22 (s, 2 H), 3,11 (d, J = 12,1 Hz, 2 H), 2,67 (dd, J = 10,2, 12,0 Hz). , 2 H), 1,78 (dd, J = 11,6, 11,6 Hz, 2 H), 1,70–1,51 (m, 9 H), 1,29 (s, 2 H), 0,93–0,82 (m, 1 H). A and G tert-butyl-4-(6-amino-pyridin-3-yl)piperidin-1-carboxylate CB1 I-246 4-[[5-(4,7-Diazaspiro[2.5]octan-7-yl)-2-pyridyl]amino]-2-[2-fluoro-5-methoxy-4-(piperidine-1-carbonyl)phenyl ]-6H-1,6-Nafthyridine-5-on AcHSS C18 method, m/z = 584 [M+H]+, Ret. time = 2.39 min. 1RMN-H (400 MHz, DMSO): γ; 12,75 (s, 1 H), 9,07 (s, 1 H), 8,33–8,30 (m, 1 H), 7,73 (d, J = 8,3 Hz, 1 H) , 7,64 (d, J = 6,1 Hz, 1 H), 7,50 (d, J = 7,3 Hz, 1 H), 7,30 (d, J = 8,3 Hz, 1 H ), 7,12 (d, J = 8,3 Hz, 1 H), 6,69 (d, J = 7,3 Hz, 1 H), 3,92 (s, 3 H), 3,69- 3,62 (m, 2 H), 3,21 (d, J = 4,5 Hz, 2 H), 3,12 (d, J = 11,4 Hz, 2 H), 2,73-2, 65 (m, 2 H), 1,77 (d, J = 12,6 Hz, 2 H), 1,67-1,57 (m, 6 H), 1,51 (d, J = 5,1 Hz, 3 H), 1,32-1,27 (m, 2 H), 0,90 (dd, J = 4,3, 12,4 Hz, 1 H). A and G tert-butyl-7-(6-amino-pyridin-3-yl)-4,7-diazaspiro[2,5]octan-4-carboxylate CB1 I-247 1-ethyl-N-[3-fluoro-4-[5-oxo-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1,6-naphthiridin-2-yl]phenyl ]-N-methyl-ciclobutano-carboxamide AcHSS C18 method, m/z = 556.3 [M+H]+, Ret. time = 2.47 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 8,78 (s, 1 H), 8,26 (d, J = 2,0 Hz, 1 H), 8, 09 (d, J = 3,1 Hz, 1 H). ), 7,98 (dd, J = 8,4, 8,4 Hz, 1 H), 7,51 (dd, J = 2,1, 9,0 Hz, 1 H), 7,43 (d, J = 7,3 Hz, 1 H), 7,39 (d, J = 8,3 Hz, 1 H), 7,30 (d, J = 8,3 Hz, 1 H), 7,07 (d , J = 8,9 Hz, 1 H), 6,62 (d, J = 7,2 Hz, 1 H), 3,49–3,39 (m, 4 H), 3,20 (s, 2 H), 3,03 (s, 4 H), 2,69 (dd, J = 1,8, 1,8 Hz, 1 H), 2,62 (s, 1 H), 2,46 (d, J = 10,3 Hz, 2 H), 2,34 (dd, J = 1,8, 1,8 Hz, 2 H), 1,84 (s, 2 H), 1,63 (s, 2 H ), 0,85 (dd, J = 7,3, 7,3 Hz). , 3ª.). A and G tert-butyl-4-(6-amino-pyridin-3-yl)-piperazin-1-carboxylate CB55 I-248 2-[5-(1-Acetylpyrrolidin-2-yl)-2-fluorophenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine -5-one AcHSS C18 method, m/z = 543.2 [M+H]+, Ret. time = 2.7 min. 1H-RMN (400 MHz, DMSO): δ 12,37 (d, J = 4,0 Hz, 1 H), 8,69 (d, J = 2,9 Hz, 1 H), 8,05 (d , J = 2,9 Hz, 1 H), 7,74 (d, J = 7,0 Hz, 1 H), 7,66 (d, J = 6,8 Hz, 1 H), 7,47 ( dd, J = 2,9, 9,0 Hz, 1 H), 7,41 (d, J = 7,3 Hz, 1 H), 7,36–7,22 (m, 2 H), 7, 03 (d, J = 8,9 Hz, 1 H), 6,63 (dd, J = 3,6, 7,3 Hz, 1 H), 5,16 (d, J = 8,0 Hz, 1 H), 5,06 (dd, J = 2,2, 7,8 Hz, 1 H), 3,79–3,74 (m, 1 H), 3,53–3,47 (m, 3 H ), 2,89–2,82 (m, 2 H), 2,41 -2,22 (m, 2 H), 2,04 (s, 2 H), 1,93-1,89 (m, 1 H), 1,82 (dd, J = 3,5, 6,8 Hz, 3 H), 1,75 (s, 2 H), 1,54-1,44 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB11 I-249 2-[6-fluoro-3-(1-piperidyl)indan-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-naphthyridina- 5- .ein AcHSS C18 method, m/z = 555.3 [M+H]+, Ret. time = 2.23 min. 1RMN-H (400 MHz, DMSO): δ 12,40 (s, 1 H), 11,63 (s, 1 H), 8,70 (d, J = 1,5 Hz, 1 H), 8, 05 (d, J = 3,0 Hz, 1 H). ), 7,80 (d, J = 7,4 Hz, 1 H), 7,48 (dd, J = 3,0, 9,0 Hz, 1 H), 7,41 (d, J = 7, 3 Hz, 1 H), 7,20 (d, J = 8,9 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6,60 (d, J = 7 ,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz, 1 H), 4,31 (dd, J = 6,9, 6,9 Hz, 1 H), 3,68- 3,61 (m, 1 H), 3,55-3,50 (m, 2 H), 2,95-2,83 (m, 4 H), 2,40-2,32 (m, 3 H ), 2,15-2,00 (m, 3 H), 1,87-1,81 (m, 2 H), 1,56-1,38 (m, 8 H). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB56 I-250 2-[2-Fluoro-5-(2-Oxa-6-Azaspiro[3,3-Heptan-6-ylmetil)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidilo)-2-Pyridyl ]amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 543 [M+H]+, Ret. time = 2.04 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,66 (s, 1 H), 8,71 (d, J = 1,6 Hz, 1 H), 8, 07 (d, J = 3,0 Hz, 1 H). ), 7,81 (dd, J = 2,3, 7,7 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,42 (d, J = 7,3 Hz, 1 H), 7,40-7,35 (m , 1 H), 7,31-7,25 (m, 1 H), 7,03 (d, J = 8,8 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,3 Hz, 1 H), 4,62 (s, 4 H), 3,67-3,60 (m, 1 H), 3,57-3,50 (m, 4 H), 3,32 (s, 4 H), 2,91-2,83 (m, 2 H ), 1,88-1,80 (m, 2H), 1,55-1,45 (m, 2H). Ein 1-(6-amino-pyridin-3-yl) piperidin-4-ol CB12 I-251 2-[4-(6,6-Difluoro-3-azabicyclo[3.1.0]hexan-3-carbonyl)-2-fluoro-5-methoxyphenyl]-4-[[5-(4-hidroxy-1-piperidyl ) )-2-pyridyl]amino]-6H-1,6- BicarbB EHC18 method, m/z = 607 [M + H]+, Ret. Time = 3.81 min. 1RMN-H (400 MHz, DMSO): δ 12,38 (s, 1 H), 8,70 (d, J = 5,6 Hz, 1 H), 8,19 (s, 1 H), 8, 04 (d, J = 2,5 Hz, 1 H). ), 7,56 (d, J = 6,1 Hz, 1 H), 7,48-7,39 (m, 2 H), 7,18 (d, J = 8,8 Hz, 1 H), 7,01 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7,1 Hz, 1 H), 3,96 (d, J = 12,6 Hz, 1 H) , 3,95–3,80 (m, 6 H), 3,75–3,57 (m, 3 H), 3,49–3,41 (m, 3 H), 2,89-2,81 (m, 2 h), 2,65-2,53 (m, 2 h), 1,86-1,79 (m, 2 Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB67 naphthyridin- H), 1.53–1.41 (m, 2H). 5 how I-252 (1R,2S,4S)-N-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 - Naphthyridin-2-yl]phenyl]norbornan-2-carboxamide Racemisch BicarbB EHC18 method, m/z = 569 [M + H]+, Ret. Time = 4.28 min. 1RMN-H (400 MHz, DMSO): δ 12,36 (s, 1 H), 11,59 (d, J = 5,6 Hz, 1 H), 10,17 (s, 1 H), 8, 71 (s, 1 H), 8,06 (d , J = 2,5 Hz, 1 H), 7,98 (dd, J = 8,8, 8,8 Hz, 1 H), 7,46 ( dd, J = 3,3, 8,6 Hz, 1 H), 7,38 (dd, J = 6,4, 6,4 Hz). , 1 H), 7,01 (d, J = 9,1 Hz, 1 H), 6,56 (d, J = 7,1 Hz, 1 H), 3,67-3,60 (m, 1 H), 2,86 (dd, J = 9,7, 9,7 Hz, 4 H), 1,83 (d, J = 9,9 Hz, 4 H), 1,71–1,66 (m , 1 H), 1,60–1,41 (m, 6 H), 1,38–1,23 (m, 6 H). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB57 I-253 2-(6-Fluoro-3-morpholino-indan-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6 naphthyridin- 5- . .eins AcHSS C18 method, m/z = 557 [M+H]+, Ret. time = 2.14 min. 1RMN-H (400 MHz, DMSO): δ 12,37 (s, 1 H), 8,69 (s, 1 H), 8,29 (d, J = 8,4 Hz, 1 H), 8, 03 (d, J = 1,0 Hz, 1 H). ), 7,84–7,81 (m, 1 H), 7,48 (d, J = 9,2 Hz, 1 H), 7,42–7,39 (m, 1 H), 7,21 (d, J = 8,9 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4, 30 (d, J = 5,0 Hz, 1 H), 3,43 (s, 6 H), 2,99-2,81 (m , 6 H), 2,38 (s, 3 H), 2 ,15–2,07 (m, 2 H), 1,83 (dd, J = 2,9, 12,6 Hz, 2 H), 1,53–1,45 (m, 3 H). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB58 I-254 2-[5-(2-Azaspiro[3.3]heptano-2-ylmethyl)-2-fluorophenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H - 1 ,6-naphthyridin-5-one AcHSS C18 method, m/z = 541 [M+H]+, Ret. time = 2.22 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,66 (s, 1 H), 8,70 (d, J = 1,5 Hz, 1 H), 8, 07 (d, J = 3,0 Hz, 1 H). ), 7,81 (dd, J = 2,2, 7,6 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H), 7,44–7, 35 (m, 2 H), 7,30–7,24 (m, 1 H ), 7,03 (d, J = 9,0 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,1 Hz, 1 H), 3,67–3,61 (m, 2 H), 3,55 (s, 3 H), 3,51 (dd, J = 4,3, 4,3 Hz, 2 H), 3,12 (s, 2 H), 2,91–2,83 (m, 2 H), 2,05 (dd, J = 7,6, 7,6 Hz, 4 H), 1,87–1,73 (m, 4 H), 1,55–1,45 (m, 2 H). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB59 I-255 2-[2-Fluoro-5-(5-oxa-2-azaspiro[3.4]octan-2-ylmethyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino ]-6H-1,6-naphthyridin-5-one BicarbB EHC18 method, m/z = 557.2 [M+H]+, Ret. tempo = 3.82 min. 1RMN-H (400 MHz, DMSO): δ 12,46–12,42 (m, 1 H), 8,71 (d, J = 1,5 Hz, 1 H), 8,06 (d, J = 3,0 Hz, 1 H), 7,83 (dd, J = 2,2, 7,7 Hz, 1 H), 7,48 (dd, J = 3,1, 9,0 Hz, 1 H) , 7,44–7,38 (m, 2 H), 7,28 (dd, J = 8,5, 11,1 Hz, 1 H), 7,03 (d, J = 8,9 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,70 (d, J = 4,3 Hz, 1 H), 3,69–3,63 (m, 6 H) , 3,55 -3,50 (m, 2 H), 3,32-3,26 (m, 2 H), 3,05 (d, J = 8,2 Hz, 2 H), 2,91- 2,83 (m, 2 H), 2,02 (dd, J = 7,2, 7,2 Hz, 2H), 1,86-1,79 (m, 4H), 1,55-1,46 (m, 2H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB60 I-256 2-[2-Fluoro-5-(6-oxa-2-azaspiro[3.4]octan-2-ylmethyl)phenyl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino ]-6H-1,6-naphthyridin-5-one BicarbB EHC18 method, m/z = 557 [M + H]+, Ret. Time = 3.7 min. 1RMN-H (400 MHz, DMSO): δ 12,46 (s, 1 H), 11,70 (d, J = 5,1 Hz, 1 H), 8,74 (s, 1 H), 8, 23 (s, 1 H), 8,10 (d , J = 2,8 Hz, 1 H), 7,88 (dd, J = 1,6, 7,2 Hz, 1 H), 7,54– 7,43 (m, 3 H), 7,36–7,29 (m, 1 H), 7,07 (d, J = 8,8 Hz, 1 H), 6,65 (d, J = 7 ,3 Hz, 1 H), 3,71 3,64 (m, 6 H), 3,60-3,52 (m, 2 H), 3,27-3,21 (m, 4 H), 2 ,94-2,86 (m, 2 H), 2,14-2,03 (m, 3 H), 1,92-1,86 (m, 2 H), 1,60-1,48 (m , 2H). Ein 1-(6-Amino-Piridin-il)-Piperidin-4-ol CB61 I-257 2-[2-Fluoro-4-(4-pyridyl-methoxy)fenil]-4-[(3-THF-3-yl-1H-pyrazol-5-yl)amino]-6H-1,6-naphthyridin- 5-ein AcHSS C18 method, m/z = 499 [M+H]+, Ret. time = 2.25 min. 1RMN-H (400 MHz, DMSO): δ 12,44 (d, J = 1,1 Hz, 1 H), 11,96 (s, 1 H), 11,59 (d, J = 4,3 Hz , 1 H), 8,62 (d, J = 5,6 Hz, 2 H), 8,25 (s, 1 H), 8,01 (dd, J = 8,9, 8,9 Hz, 1 H), 7,48 (d, J = 6,0 Hz, 2 H), 7,42–7,37 (m, 1 H), 7,09 (d, J = 7,3 Hz, 1 H) , 7,08–7,02 (m, 1 H), 6,56 (d, J = 7,3 Hz, 1 H), 6,04 (d, J = 2,0 Hz, 1 H). ), 5,31 (s, 2 H), 4,02 (dd, J = 7,8, 7,8 Hz, 1 H), 3,92–3,77 (m, 2 H), 3,63 (dd, J = 7,7, 7,7 Hz, 1 H), 3,49-3,40 (m, 1 H), 2,35-2,55 (m, 1 H), 2,01 Ein 5-(THF-3-il)-1H-pirazol-3-amina CB5 (ddd, J = 7,2, 12,2, 15,0 Hopping, 1H). I-258 2-[3-(2-Azaspiro[3.3]heptan-2-yl)-6-fluoro-indan-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl] amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 567.3 [M+H]+, Ret. time = 2.39 min. 1RMN-H (400 MHz, DMSO): δ 12,40 (s, 1 H), 11,67 (d, J = 5,1 Hz, 1 H), 8,68 (d, J = 1,4 Hz , 1 H), 8,37 (s, 1 H). ), 8,06 (d, J = 3,0 Hz, 1 H), 7,77 (d, J = 7,4 Hz, 1 H), 7,48 (dd, J = 3,1, 9, 0 Hz, 1 H), 7,44–7,39 (m, 1 H), 7,03 (d, J = 9,0 Hz, 1 H), 6,60 (d, J = 7,2 Hz , 1 H), 3,76 (dd, J = 2,9, 6,6 Hz, 2 H), 3,67–3,60 (m, 4 H), 3,25 (d, J = 6, 3 Hz, 2 H), 3,12 (d, J = 6,1 Hz, 3 H), 3,03-2,77 (m, 4 H), 2,56 (s, 1 H), Ein 1-(6-Amino-Piridin-3-il) Piperidin-4-ol CB62 2,11-2,06 (m, 1H), 2,04 (dd, J = 7,6, 7,6 Hz, 2 H), 1,89-1,73 (m, 4 H), 1,56- 1.45 (metro, 2H). I-259 2-(6-fluoro-1-morphoindan-5-y)-4-[[5-(4-hydroxy-1-piperidin)-2-pyridi]amino]-6H-1,6-naphthiridina-5-einas AcHSS C18 method, m/z = 557 [M+H]+, Ret. time = 2.07 min. 1H-NMR (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,70 (brs, 1 H), 8,71 (s, 1 H), 8,11 (d, J = 2 ,8 Hz, 1 H), 7,82 (d , J = 7,3 Hz, 1 H), 7,52 (dd, J = 2,9, 9,0 Hz, 1 H), 7,45 ( d, J = 7,3 Hz, 1 H), 7,21 (d, J = 8,8 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,62 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 3,8 Hz, 1 H), 4,39 (dd, J = 7,1, 7,1 Hz). , 1 H), 3,68-3,62 (m, 6 H), 3,60-3,54 (m, 3 H), 2,99-2,85 (m, 4 H), 2,47 -2,42 (m, 2 H), 2,21-2,12 (m , 2 Std.), 1,90–1,84 (m, 2 Std.), 1,59–1,49 (m , 2ª.). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-o CB63 I-260 2-[6-Fluoro-3-(6-oxa-2-azaspiro[3.4]octan-2-yl)indan-5-yl]-4-[[5-(4-hydroxy-1-piperidyl)-2 -Pyridyl]amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 583 [M+H]+, Ret. time = 2.13 min. 1RMN-H (400 MHz, DMSO): δ 12,45 (s, 1 H), 8,72 (s, 1 H), 11,79 (S, 1 H), 8,08 (d, J = 2 ,8 Hz, 1 H), 7,83 (d , J = 7,3 Hz, 1 H), 7,52 (dd, J = 3,0, 8,8 Hz, 1 H), 7,45 ( d, J = 7,1 Hz, 1 H), 7,22 (d, J = 8,8 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,63 (d, J = 7,3 Hz, 1 H), 4,73 (d, J = 3,5 Hz, 1 H), 3,89 (dd, J = 2,5, 6,3 Hz). , 1 H), 3,75-3,64 (m, 6 H), 3,60-3,53 (m, 2 H), 3,22 (d, J = 7,6 Hz, 2 H), 3,20 (dd, J = 6,3, 6,3 Hz, 2 H), 3,10-3,00 (m, 1 A 1-(6-amino-pyridin-3-y)piperidin-4-ol CB64 H), 2,95-2,85 (m, 3 H), 2.15-2.00 (m, 3H), 1.97- 1.83 (metro, 3H), 1.60-1.49 (M, 2H). I-261 (1R,2S,4S)-N-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 - Naphthyridin-2-yl]phenyl]norbornan-2-carboxamide Isomer 1 during SFC obtaining AcHSS C18 method, m/z = 569 [M+H]+, Ret. time = 3.5 min. 1RMN-H (400 MHz, DMSO): γ; 11,16 (s, 1 H), 9,67 (s, 1 H), 8,43–8,40 (m, 1 H), 7,97 (d, J = 2,9 Hz, 1 H) , 7,43 (dd, J = 3,0, 9,0 Hz, 1 H), 7,39–7,35 (m, 2 H), 7,15 (d, J = 7,9 Hz, 1 H), 7,07 (dd, J = 7,2, 7,2 Hz, 1 H). ), 6,98-6,94 (m, 2 H), 4,14-4,14 ​​(m, 2 H), 3,63 (s, 2 H), 3,10 (dd, J = 4,9, 4,9 Hz, 4 H), 2,69-2,64 (m , 2 H), 2,48 (dd, J = 4,8, 4,8 Hz, 4 H), 2, 23 (s, 3 H), 2,09 (s, 2 H), 1,22 (dd, J = 7,6, 7,6 Hz, 3 H). Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB57 I-262 2-[4-(6,6-difluoro-3-azabicyclo[3.1.0]hexano-3-carbonyl)-2-fluoro-phenyl]-4-[(5-piperazin-1-yl-2-pyridyl) ; amino]-6H-1,6-nafthyridin-5-ona AcHSS C18 method, m/z = 562 [M+H]+, Ret. time = 2.16 min. 1RMN-H (400 MHz, DMSO): δ 12,52 (s, 1 H), 8,84–8,80 (m, 1 H), 8,14 (d, J = 2,5 Hz, 1 H ), 8,03 (dd, J = 7,8, 7,8). Hz, 1 H), 7,57 (dd, J = 2,8, 9,1 Hz, 1 H), 7,48-7,44 (m, 3 H), 7,13 (d, J = 8 ,8 Hz, 1 H), 6,68 (d, J = 7,3 Hz, 1 H), 5,72 (s, 1 H), 4,17 (d, J = 12,9 Hz, 1 H ), 4,02-3,94 (m, 2 H), 3,67 (d, J = 11,4 Hz, 2 H), 3,40–3,36 (m, 4H), 3,24 ( dd, J = 5,3, 5,3 Hz, 4H), 2,66–2,59 (m, 2H). F and G tert-butyl 6,6-difluoro-3-azabicyclo[3.1.0]hexane 4-(6-amino-pyridin-3-yl)piperazine-1-carboxylate (step 2) I-263 2-[2-Fluoro-5-[2,2,2-Trifluoro-1-Methyl-1-(Methyl-Amino)Ethyl]Phenyl]-4-[[5-(4-Hydroxy-1-Piperidilo)- 2-pyridyl]amino]-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 557 [M+H]+, Ret. time = 2.73 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,7 (brs, 1 H), 8,77 (s, 1 H), 8,24 (d, J = 5 ,6 Hz, 1 H), 8,11 (d , J = 2,5 Hz, 1 H), 7,77-7,74 (m, 1 H), 7,53 (dd, J = 2,9 , 9,0 Hz, 1 H), 7,50-7,40 (m, 2 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,85 (s, 1 H) , 6,66 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 3,71-3,65 (m, 1 H ), 3 ,57 (d, J = 12,4 Hz, 2 H), 2,97-2,89 (m, 2 H), 2,15 (s, 3 H), 1,89 (d, J = 9, 1 Hz, 2 H), 1,80 (s, 3 Std.), 1,61–1,50 (m, 2 Std.). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB13 I-264 2-[4-(3-Azabicyclo[2.2.2]octan-3-carbonyl)-2-fluoro-phenyl]-4-[(5-piperazin-1-yl-2-pyridyl)amino]-6H-1 ,6-naphthyridin-5-uno AcHSS C18 method, m/z = 554 [M+H]+, Ret. time = 2.34 min. 1RMN-H (400 MHz, DMSO): δ 12,51 (s, 1 H), 11,80 (brs, 1 H), 8,80 (s, 1 H), 8,12–8,04 (m , 2 H), 7,54–7,45 (m, 3 H), 7,39 (d, J = 7,8 Hz, 1 H), 7,09 (d, J = 9,1 Hz, 1 H), 6,66–6,61 (m, 1 H), 3,62–3,57 (m, 1 H), 3,52 (s, 2 H), 3,45-3,41 (m , 1 H), 3,16-3,10 (m, 4 H), 2,92 (dd, J = 4,5, 4,5 Hz, 4 H), 2,06 (s, 1 H) , 1,91–1,86 (m, 2 H), 1,75–1,63 (m, 6 H). F and G tert-butyl 4-(6-amino-pyridin-3-yl)piperazin-1-carboxy-lato 2-azabicyclo[2.2.2]octane (stage 2); I-265 2-(6-Fluoro-1,2,3,4-tetrahydro-isoquinolin-5-yl)-4-[(3-THF-3-yl-1H-pyrazol-5-yl)amino]-6H-1 ,6-naphthyridin-5-one AcHSS C18 method, m/z = 447 [M+H]+, Ret. time = 2 min. 1RMN-H (400 MHz, DMSO): δ 12,32 (s, 1 H), 12,03 (s, 1 H), 7,80 (s, 1 H), 7,40 (d, J = 7 ,3 Hz, 1 H), 7,14 (dd , J = 5,8, 8,5 Hz, 1 H), 7,07 (dd, J = 8,9, 8,9 Hz, 1 H), 6,51 (d, J = 7,3 Hz, 1 H), 6,02–6,00 (m, 1 H), 3,99 (dd, J = 7,8, 7,8 Hz, 1 H ), 3,88-3,77 (m, 4 H), 3,60 (dd, J = 7,6, 7,6 Hz, 1 H), 3,47-3,38 (m, 1 H) , 2,84 (dd, J = 6,0, 6,0 Hz, 2 H), 2,70–2,67 (m, 1 H), 2,45 (dd, J = 5,5, 5, 5 Hz, 2 H), 2,36–2,23 (m, 2 H), 2,02–1,93 (M, 1 H). A e G 5-(THF-3-yl)-1H-pyrazol-3-amine CB45 I-266 (1R,2R,4S)-N-[3-Fluor-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 - Naftiridin-2-yl]phenyl]norbornano-2-carboxamide Racemisch Method AcHSS C18, m/z = 570 [M+H]+, Ret. rhythm = 3.49 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,64 (d, J = 5,3 Hz, 1 H), 10,30 (s, 1 H), 8, 76 (s, 1 H), 8,12 (d , J = 2,8 Hz, 1 H), 8,04 (dd, J = 8,8, 8,8 Hz, 1 H), 7,78- 7,75 (m, 1 H), 7,55-7,47 (m, 2 H), 7,44 (dd, J = 6,6, 6,6 Hz, 1 H), 7,07 (d , J = 8,8 Hz, 1 H), 6,61 (d, J = 6,6 Hz, 1 H), 4,75-4,71 (m, 1 H), 3,71-3,66 (m, 1 H), 3,59-3,55 (m, 2 H), 2,95-2,89 (m, 2 H), 2,49 (d, J = 13,1 Hz, 1 H ), 2,39-2,33 (m, 1 Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB65 H), 1,95-1,85 (m, 3 H), 1,60-1,25 (m, 8 H), 1,17- 1,14 (m, 1H), 0,96–0,92 (M, 1H). I-267 Isomer N-[3-Fluoro-4-[5-oxo-4-[(5-THF-3-yl-2-pyridyl)amino]-6H-1,6-naphthyridin-2-yl]phenyl]cyclohexanecarboxamide 2 separated by SFC BicarbB EHC18 method, m/z = 528 [M + H]+, Ret. Time = 4.79 min. 1RMN-H (400 MHz, DMSO): δ 12,84 (s, 1 H), 11,73 (s, 1 H), 10,28 (s, 1 H), 9,07 (s, 1 H) , 8,35 (d, J = 1,8 Hz). , 1 H), 8,06 (dd, J = 8,8, 8,8 Hz, 1 H), 7,82-7,74 (m, 2 H), 7,54-7,46 (m, 2 H), 7,11 (d, J = 8,3 Hz, 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,11-3,98 (m, 2 H ), 3,86 (q, J = 7,8 Hz, 1 H), 3,61 (dd, J = 7,8, 7,8 Hz, 1 Std.), 3,50–3,41 (m , 1 estándar), 2,46–2,33 (m, 1 estándar), 2,04–1,79 (m, 5 estándar), 1,73 Ein 5-(THF-3-il)-Piridin-2-Amin CB19 (d, J = 11,1 Hz, 1 H), 1,53- 1,23 (M, 6H). I-268 2-(6-Fluoro-2-methyl-3,4-di-hydro-1H-isoquinolin-5-yl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino] -6H - 1,6-naphthyridin-5-one AcHSS C18 method, m/z = 501 [M+H]+, Ret. time = 2.06 min. 1RMN-H (400 MHz, DMSO): γ; 12,56 (s, 1 H), 11,72 (s, 1 H), 8,37 (s, 1 H), 8,08 (d, J = 2,8 Hz, 1 H), 7,52 –7,43 (m, 2 H), 7,23 (dd, J = 5,8, 8,6 Hz, 1 H), 7,15 (dd, J = 9,0, 9,0 Hz, 1 H), 7,02 (d, J = 8,8 Hz, 1 H), 6,58 (d, J = 7,3 Hz, 1 H), 4,71 (d, J = 4,0 Hz, 1 H), 3,70–3,63 (m, 1 H), 3,54–3,42 (m, 2 H), 3,36 (s, 2 H), 2,93-2,83 ( m, 2 H), 2,67-2,59 (m, 2 H), 2,36 (s, 3 H), 1,85 (d, J = 9,9 Hz, 2 H), 1,57 -1,46 (m , 4 Est.). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB46 I-269 (1R,2R,4S)-N-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 - Isomer 1 of naphthyridin-2-yl]phenyl]norbornan-2-carboxamide during the preparation of SFC BicarbB EHC18 method, m/z = 569 [M + H]+, Ret. Time = 4.35 min. 1RMN-H (400 MHz, DMSO): γ; 12,48–12,48 (m, 1 H), 10,33 (s, 1 H), 8,75 (s, 1 H), 8,13 (d, J = 2,8 Hz, 1 H) , 8,04 -7,99 (m, 1 H), 7,82 (s, 1 H), 7,55-7,48 (m, 3 H), 7,09 (d, J = 8,6 Hz, 1 H), 6,64 (d, J = 7,3 Hz, 1 H), 4,78–4,71 (m, 1 H), 3,69 (dd, J = 4,3, 4 ,3 Hz, 1 H), 3,58 (d, J = 12,9 Hz, 2 H), 2,93 (dd, J = 9,9, 9,9 Hz, 2 H), 2,51– 2,45 (m, 2 H), 2,39–2,31 (m, 2 H), 1,96–1,84 (m, 3 H), 1,60–1,51 (m, 4 H ), 1,51–1,23 ( m, 4 H), 1,16 (d, J = 9,1 Hz, 1 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB65 I-270 2-[5-Cyclopropyl-2-Fluoro-4-(Piperidin-1-Carbonyl)Phenyl]-4-[[5-(4-Hydroxy-1-Piperidyl)-2-Pyridyl]Amino]-6H-Nafthyridine- 5-eins BicarbB EHC18 method, m/z = 583 [M + H]+, Ret. Time = 4.23 min. 1RMN-H (400 MHz, DMSO): γ; 12,47 (s, 1 H), 11,70 (s, 1 H), 8,72 (s, 1 H), 8,11 (d, J = 2,8 Hz, 1 H), 7,54 –7,45 (m, 3 H), 7,23 (d, J = 8,8 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,64 (d , J = 7,1 Hz, 1 H), 4,74–4,73 (m, 1 H), 3,74 - 3,66 (m, 3 H), 3,57 (d, J = 12, 6 Hz, 2 H), 3,25 (dd, J = 4,9, 4,9 Hz, 2 H), 2,96 - 2,86 (m, 2 H), 1,94–1,86 ( m, 3 H), 1,65 (dd, J = 4,5, 5,3 Hz, 4 H), 1,58–1,50 (m, 4 H), 1,01 (d, J = 8 ,3Hz, 2H). ), 0,89-0,83 (m, 1 H), 0,69-0,65 (m, 1 H). Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB14 I-271 4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-2-[4-(1-methyl-imidazol-2-yl)piperazin-1-yl]-6H-1, 6-naphthyridin-5-one AcHSS C18 method, m/z = 502 [M+H]+, Ret. time = 1.9 min. 1RMN-H (400 MHz, DMSO) δ 12,34 (s, 1H), 11,24 (d, J = 4,8 Hz, 1H), 8,11–8,08 (m, 1H), 8, 01 (s, 1H), 7,49 (dd, J = 2,8, 9,1 Hz, 1H), 7,29 (t, J = 6,6 Hz, 1H), 6,98–6,94 (m, 2H), 6,67 (d, J = 1,0 Hz, 1H), 6,31 (d, J = 6,8 Hz, 1H), 4,74-4,73 (m, 1H) , 3,84-3,77 (m, 4H), 3,71-3,64 (m, 1H), 3,57-3,52 (m, 5H), 3,14-3,08 (m, 4H), 2,92-2,85 (m, 2H), 1,90-1,84 (m, 2H), 1,61-1,50 (m, 2H). E 1-(6-Amino-pyridin-3-yl)piperidin-4-ol 1-(1-Methyl-1H-imidazol-2-yl)piperazine I-272 N-[3-Fluoro-4-[5-oxo-4-[(5-THF-3-yl-2-pyridyl)amino]-6H-1,6-naphthyridin-2-yl]phenyl]cyclohexanecarboxamide isomer 1 separated by SFC BicarbB EHC18 method, m/z = 528 [M + H]+, Ret. Time = 4.79 min. 1RMN-H (400 MHz, DMSO): δ 12,76 (s, 1 H), 11,73 (s, 1 H), 10,26 (s, 1 H), 9,08 (s, 1 H) , 8,36 (d, J = 2,0 Hz). , 1 H), 8,07 (dd, J = 8,8, 8,8 Hz, 1 H), 7,82-7,74 (m, 2 H), 7,54-7,45 (m, 2 H), 7,11 (d, J = 8,3 Hz, 1 H), 6,65 (d, J = 7,1 Hz, 1 H), 4,12–3,98 (m, 2 H ), 3,87 (q, J = 7,8 Hz, 1 H), 3,61 (dd, J = 7,8, 7,8 Hz, 1 H), 3,50-3,41 (m, 1 H), 2,46-2,33 (m, 2 H), 1,99 (ddd, J = 8,1, 12,3 16,0 Hz, 1 H), 1,93-1,79 ( m, 3 H), 1,73 (d, J = 11,1 Hz, 1 H), 1,53-1,22 (m, 6 H). Ein 5-(THF-3-il)-Piridin-2-Amin CB19 I-273 2-(2-Fluoro-5-(1,1,1-Tifluoro-2-hydroxy-propan-2-yl)phenyl)-4-((5-(4-hydroxy-piperidin-1-yl)pyridin- 2-yl)amino)-1,6-naphthyridine-5(6H)-in isomer 2 during SFC production AcHSS C18 method, m/z = 544 [M+H]+, Ret. time = 2.95 min. 1RMN-H (400 MHz, DMSO): δ 12,50 (s, 1 H), 11,46–11,44 (m, 1 H), 8,77 (s, 1 H), 8,27–8 ,22 (m, 1 H), 8,11 (d, J = 2,5 Hz, 1 H), 7,76-7,72 (m, 1 H), 7,53 (dd, J = 2, 8, 8,8 Hz, 1 H), 7,48 (d, J = 7,3 Hz, 1 H), 7,45-7,40 (m, 1 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,85-6,82 (m, 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,73 (d, J = 2, 5 Hz, 1 H), 3,69 (d, J = 2,5 Hz, 1 H), 3,57 (d, J = 12,4 Hz, 2 H), 2,97–2,89 (m , 2 H), 1,93–1,84 (m, 2 H ), 1,80 (s, 3 H), 1,61–1,49 (m, 2 H). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB8 I-274 2-(2-Fluoro-5-(1,1,1-hydroxy-propan-2-yl)phenyl)-4-((5-(4-hydroxy-piperidin-1-y)pyridin-2-yl) amino)-1,6-naphthyridin-5(6H)-one Isomer 1 separated by SFC BicarbB EHC18 method, m/z = 544 [M + H]+, Ret. Time = 3.72 min. 1RMN-H (400 MHz, DMSO): δ 12,48 (s, 1 H), 11,7 (brs, 1 H), 8,77 (s, 1 H), 8,24 (d, J = 5 ,6 Hz, 1 H), 8,11 (d , J = 2,5 Hz, 1 H), 7,77-7,74 (m, 1 H), 7,53 (dd, J = 2,9 , 9,0 Hz, 1 H), 7,50-7,40 (m, 2 H), 7,08 (d, J = 8,8 Hz, 1 H), 6,85 (s, 1 H) , 6,66 (d, J = 7,3 Hz, 1 H), 4,74 (d, J = 4,0 Hz, 1 H), 3,71-3,65 (m, 1 H ), 3 ,57 (d, J = 12,4 Hz, 2 H), 2,97–2,89 (m, 2 H), 1,89 (d, J = 9,1 Hz, 2 H), 1,80 (s, 3H), 1,61–1,50 (M, 2H). A 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB8 I-275 (1R,2S,4S)-N-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 - Isomer 2 of naphthyridin-2-yl]phenyl]norbornan-2-carboxamide AcHSS C18 method, m/z = 569.3 [M+H]+, Ret. time = 3.54 min. 1RMN-H (400 MHz, DMSO): δ 12,42 (s, 1 H), 11,64 (d, J = 5,3 Hz, 1 H), 10,22 (s, 1 H), 8, 76 (s, 1 H), 8,27 (s , 1 H), 8,12 (d, J = 2,8 Hz, 1 H), 8,03 (dd, J = 8,8, 8,8 Hz, 1 H), 7,82 (d, J = 1,5 Hz, 1 H), 7,55–7,49 ( m, 2 H), 7,44 (dd, J = 6,3, 6 ,3 Hz, 1 H), 7,06 (d, J = 8,8 Hz, 1 H), 6,61 (d, J = 7,1 Hz, 1 H), 4,73–4,73 ( m, 1 H), 3,72-3,66 (m, 2 H), 3,60-3,55 (m, 2 H), 3,22 (s, 1 H), 2,96-2, 89 (m, 3 H), 2,67-2,49 Ein 1-(6-Amino-piridin-3-il)piperidin-4-ol CB57 SFC (M, 2H), 1.91-1.87 (M, 2 H), 1,77-1,71 (m, 1 H), 1,65-1,51 (m, 2 H), 1,43- 1.32 (m, 2H), 1.30 (s, 2nd norm). I-276 2-(2-fluoro-4-THF-2-yl-phenyl)-4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-6H-1,6-naphthyridine-5 -eins AcHSS C18 method, m/z = 502 [M+H]+, Ret. time = 3.05 min. 1RMN-H (400 MHz, DMSO): δ 12,84 (s, 1 H), 11,73 (s, 1 H), 10,28 (s, 1 H), 9,07 (s, 1 H) , 8,35 (d, J = 1,8 Hz). , 1 H), 8,06 (dd, J = 8,8, 8,8 Hz, 1 H), 7,82-7,74 (m, 2 H), 7,54-7,46 (m, 2 H), 7,11 (d, J = 8,3 Hz, 1 H), 6,65 (d, J = 7,3 Hz, 1 H), 4,11-3,98 (m, 2 H ), 3,86 (q, J = 7,8 Hz, 1 H), 3,61 (dd, J = 7,8, 7,8 Hz, 1 H), 3,50–3,41 (m, 1 H), 2,46–2,33 (m, 1 H), 2,04–1,79 (m, 3 H), 1,73 (d, J = 11,1 Hz, 1 H), 1 ,53–1,23 (M, 6H). A 1-(6-amino-pyridin-4-ol 2-[2-fluoro-4-(tetra-hydro- 2-furanyl)phenyl]-4,4,5,5-tetramethyl- 1,3,2- Dioxa- Borolan I-277 (1R,2R,4S)-N-[3-Fluoro-4-[4-[[5-(4-hydroxy-1-piperidyl)-2-pyridyl]amino]-5-oxo-6H-1,6 - Isomer 2 of naphthyridin-2-yl]phenyl]norbornan-2-carboxamide is obtained by AcHSS C18 method, m/z = 569 [M+H]+, Ret. time = 3.53 min. 1RMN H (400 MHz, DMSO): δ 12,47 (s, 1 H), 11,70 (s, 1 H), 10,33–10,30 (m, 1 H), 8,76 (s , 1 H), 8,12 (d, J = 3,0 Hz, 1 H), 8,06–8,01 (m, 1 H), 7,81–7,76 (m, 1 H), 7,55–7,49 (m, 2 H), 7,45 (d, J = 7,3 Hz, 1 H), 7,06 (d, J = 9,1 Hz, 1 H), 6, 61 (d, J = 7,3 Hz, 1 H), 4,74–4,72 (m, 1 H), 3,68–3,66 (m, 1 H), 3,59–3,54 (m, 2 H), 2,95-2,89 (m, 1 H), 2,39-2,33 (m, 2 H), 1,94-1,84 (m, 3 H), 1 ,59-1,51 (m, 4 Ein 1-(6-Amino-pyridin-3-yl)piperidin-4-ol CB65 SFC H), 1,47-1,44 (m, 2 H), 1,38-1,25 (m, 3 H), 1,16 (d, J=9.1Hz, 2H). I-278 2-[4-(3-Azabicyclo[2.2.2]octan-3-carbonyl)-2-fluoro-phenyl]-4-[[5-(4-methyl-piperazin-1-yl)-2-pyridyl] Amino]-6H-1,6-Nafthyridin-5-on AcHSS C18 method, m/z = 568 [M+H]+, Ret. time = 2.39 min. 1H-NMR (400 MHz, DMSO) &dr; 12,48 (s, 1H), 11,71 (d, J = 5,4 Hz, 1H), 8,77 (d, J = 1,4 Hz, 1H), 8,26 (s, 1H), 8,09–8,00 (m, 2H), 7,52–7,33 (m, 2H), 7,07–7,04 (m, 1H), 6,62–6,58 (m, 1H ), 3,48 (d, J = 2,1 Hz, 2H), 3,38 (d, J = 2,0 Hz, 1H), 3,19–3,13 (m, 4H), 2,50 –2,44 (m, 4H), 2,25–2,23 (m, 3H), 2,04–2,02 (m, 1H), 1,90–1,84 (m, 2H). ), 1,69-1,63 (m, 6H). F 1-methylpiperazine 2-azabicyclo[2.2.2]octane (Step 2) I-279 2-[4-(6,6-Difluor-3-azabiciclo[3.1.0]hexan-3-carbonyl)-2-fluor-fenil]-4-[[5-(4-metil-piperazin-1-ilo ) ) )-2-pyridyl]amino]-6H- 1,6- AcHSS C18 method, m/z = 576 [M+H]+, Ret. time = 2.23 min. 1RMN-H (400 MHz, DMSO) 12,49–12,47 (m, 1H), 11,71–11,68 (m, 1H), 8,78 (d, J = 1,5 Hz, 1H) , 8,08–8,01 (m, 2H), 7,52–7,42 (m, 4H), 7,07-7,04 (m, 1H), 6,60 (d, J = 7, 3 Hz, 1H), 4,15 (d, J = 12,8 Hz, 1H), 4,02-3,95 (m, 1H), 3,79- 3,64 (m, 2H), 3, 20–3,14 (m, 4H), 2,69–2,55 (m, 2H), 2,48 (t, J = 5,0 Hz, 4H), 2,24 (s, 3H). F 1-methylpiperazine 6,6-difluoro-3-azabicyclo[3.1.0]hexane (step 2) naphthyridin- 5 how I-280 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(3-methyl-4-pyridyl)-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 429 [M+H]+, Ret. time = 2.18 min. 1RMN-H (400 MHz, DMSO): γ; 12,54 (s, 1 H), 11,73 (d, J = 4,3 Hz, 1 H), 8,60 (s, 1 H), 8,59–8,53 (m, 2 H) , 8,10 (d, J = 2,8 Hz, 1 H), 7,54–7,44 (m, 3 H), 7,06 (d, J = 8,8 Hz, 1 H), 6 ,61 (d, J = 7,1 Hz, 1 H), 4,73 (d, J = 4,0 Hz, 1 H), 3,71-3,64 (m, 1 H), 3,59 -3,51 (m, 2 H), 2,94-2,85 (m, 2 H), 2,42 (s, 3 H), 1,91 -1,82 (m, 2 H), 1 ,58-1,48 (m, 2H). A 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol(3-methyl-pyridin-3-yl)-ácido boronic I-281 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(4-methyl-3-pyridyl)-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 429 [M+H]+, Ret. time = 2.2 min. 1RMN-H (400 MHz, DMSO): δ 12,55 (s, 1 H), 11,71–11,65 (m, 1 H), 8,62 (s, 1 H), 8,57–8 ,52 (m, 2 H), 8,11 (d, J = 2,8 Hz, 1 H), 7,54-7,45 (m, 2 H), 7,42 (d, J = 5, 1 Hz, 1 H), 7,06 (d, J = 9,1 Hz, 1 H), 6,61 (d, J = 7,3 Hz, 1 H), 4,73-4,72 (m , 1 H), 3,67 (s, 1 H), 3,57-3,50 (m, 2 H), 2,94-2,86 (m, 2 H), 2,46 (s, 3 H), 1,86 (dd, J = 2,9, 12,0 Hz, 2 H), 1,59-1,48 (m, 2 H). 1-(6-amino-pyridin-3-yl)piperidin-4-ol(4-methyl-pyridin-3-yl)boronic acid I-282 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(1H-pyrrolo[2,3-b]pyridin-4-yl)-6H-1,6-naphthyridine - 5-ein AcHSS C18 method, m/z = 454 [M+H]+, Ret. time = 3.44 min. 1RMN-H (400 MHz, DMSO): δ 12,28 (s, 1 H), 11,54–11,53 (m, 1 H), 9,70 (s, 1 H), 8,43–8 ,40 (m, 2 H), 8,08–8,03 ( m, 2 H), 7,48 (dd, J = 3,2, 9,0 Hz, 1 H), 7,35 (d, J = 7,1 Hz, 1 H), 7,24-7,15 (m, 2 H), 6,71 (d, J = 3,8 Hz, 1 H), 6,47 (d, J = 7,3 Hz, 1 H), 4,64 (d, J = 4,3 Hz, 1 H), 3,66-3,35 (m, 3 H), 2,88-2,79 (m, 2 H), 1,82–1,75 (m, 2 H), 1,51–1,41 (m, 2 H). Ein 1-(6-Amino-pyridin-3-yl)-piperidin-4-ol-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl)-1H -pyrrolo[2,3-b]pyridine I-283 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(4-metil-1H-pyrazol-3-yl)-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 418 [M+H]+, Ret. time = 2.49 min. 1RMN-H (400 MHz, DMSO): δ 12,75 (s, 1 H), 12,23–12,23 (m, 1 H), 11,46–11,45 (m, 1 H), 8 ,67 (s, 1 H), 8,01 (d, J = 2,8 Hz, 1 H), 7,54-7,52 (m, 1 H), 7,40 (dd, J = 3, 0, 9,1 Hz, 1 H), 7,29 (dd, J = 6,1, 6,1 Hz, 1 H), 6,94 (d, J = 8,8 Hz, 1 H), 6 ,46 (d, J = 7,3 Hz, 1 H), 4,62 (d, J = 4,0 Hz, 1 H), 3,60–3,53 (m, 1 H), 3,24 (s, 2 H), 2,84-2,76 (m, 2 H), 2,33 (s, 3 H), 1,80-1,73 (m, 2 H), 1,49-1 ,38 (m, 2H). Ein 1-(6-Amino-pyridin-3-yl)-Piperidin-4-ol-1-BOC-4-Methyl-pyrazol-3-borosäurepinakolester I-284 4-[[5-(4-Hydroxy-1-piperidin)-2-pyridyl]amino]-2-imidazo[1,2-a]pyridin-3-yl-6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 454 [M+H]+, Ret. time = 2.37 min. 1RMN-H (400 MHz, DMSO): δ 12,43 (s, 1 H), 11,65 (d, J = 5,0 Hz, 1 H), 10,21 (d, J = 7,0 Hz , 1 H), 8,92 (s, 1 H). ), 8,35 (s, 1 H), 8,21 (d, J = 3,0 Hz, 1 H), 7,77 (d, J = 9,0 Hz, 1 H), 7,51– 7,42 (m, 3 H), 7,18–7,15 ( m, 1 H), 7,04 (d, J = 8,9 Hz, 1 H), 6,71 (d, J = 7 ,2 Hz, 1 H), 4,72 (d, J = 4,3 Hz, 1 H), 3,70–3,63 (m , 1 Std.), 3,59–3,51 (m, 2 Std.), 2,94–2,86 (m, 2 Std.), 1,91–1,82 (m, 2 Std.), 1,59–1,48 (m, 2 Std.). Um 1-(6-amino-pyridin-3-yl)piperidin-4-ol 3-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl)imidazo [1, 2-a] pyridine I-285 2-(6-Fluoro-1,2,3,4-tetrahydro-isoquinolin-5-yl)-4-[(5-tetrahydro-pyran-4-yl-1H-pyrazol-3-yl)amino]-6H -1,6-naphthyrdidin-5-one AcHSS C18 method, m/z = 461 [M+H]+, Ret. time = 2.07 min. 1RMN-H (400 MHz, DMSO): δ 12,02 (s, 1 H), 8,29 (s, 1 H), 7,82 (s, 1 H), 7,45 (d, J = 7 ,3 Hz, 1 H), 7,39 (dd , J = 5,7, 8,7 Hz, 1 H), 7,29 (dd, J = 8,8, 8,8 Hz, 1 H), 6,59 (d, J = 7,3 Hz, 1 H), 5,99 (s, 1 H), 4,32 (s, 2 H), 3,45 (dd, J = 9,9, 11 ,6 Hz, 2 H), 2,95–2,88 (m, 1 H), 2,81 (dd, J = 5,7, 5,7 Hz, 4 H), 1,83 (dd, J = 1,8, 12,9 Hz, 4 H), 1,70-1,58 (m, 4 H). A and G 5-(Tetrahydro-2H-pyran-4-yl)-1H-pyrazol-3-amine CB45 I-286 2-(6-fluoro-1,2,3,4-tetrahydro-isoquinolin-5-yl)-4-[(1-methyl-5-tetrahydro-pyran-4-yl-pyrazol-3-yl)amino] - 6H-1,6-naphthyridin-5-one AcHSS C18 method, m/z = 475 [M+H]+, Ret. time = 2.17 min. 1RMN-H (400 MHz, DMSO): δ 8,36 (s, 1 H), 7,73 (d, J = 3,5 Hz, 1 H), 7,45 (d, J = 7,3 Hz , 1 H), 7,35 (dd, J = 5,8, 8,6 Hz, 1 H), 7,26 (dd, J = 9,1, 9,1 Hz, 1 H), 6,58 (d, J = 7,3 Hz, 1 H), 6,00 (s, 1 H), 4,22 (s, 2 H), 3,93 (dd, J = 3,5, 11,1 Hz , 2 H), 3,72 (s, 3 H), 3,49 (dd, J = 10,1, 11,9 Hz, 2 H), 3,20 (dd, J = 6,2, 6, 2 Hz, 2 H), 3,02–2,93 (m, 1 H), 2,74 (dd, J = 5,2, 5,2 Hz, 2 H), 1,79 (dd, J = 2,1, 13,0 Hz, 2 H), 1,67–1,55 (M, 4H). A and G 1-Methyl-5-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-3-amine CB45 I-287 4-[[5-(4-Hydroxy-1-piperidyl)-2-pyridyl]amino]-2-(2-methyl-3-pyridyl)-6H-1,6-naphthiridin-5-one AcHSS C18 method, m/z = 429 [M+H]+, Ret. time = 2.14 min. 1RMN-H (400 MHz, DMSO) δ 12,53 (s, 1H), 11,71 (s, 1H), 8,58 (dd, J = 1,8, 4,8 Hz, 1H), 8, 53 (s, 1H), 8,11 (d, J = 3,0 Hz, 1H), 7,87 (dd, J = 1,8, 7,6 Hz, 1H), 7,51 (dd, J = 3,1, 9,1 Hz, 1H), 7,46 (d, J = 7,5 Hz, 1H), 7,40 (dd, J = 4,8, 7,8 Hz, 1H), 7 ,06 (d, J = 8,8 Hz, 1H), 6,61 (d, J = 7,1 Hz, 1H), 4,73 (d, J = 3,8 Hz, 1H), 3,71 –3,64 (m, 1H), 3,58–3,50 (m, 2H), 2,94–2,85 (m, 2H), 1-(6-amino-pyridin-3-yl)-piperidin-4ol(2-methyl-piperidin-3-yl)boronic acid 2.61 (s, 3H), 1.90-1.83 (m, 2H), 1.58–1.47 (m, 2H). I-288 6-(2-Fluoro-5-isopropyl-phenyl)-8-((5-(4-hidroxy-piperidin-1-yl)pyridin-2-yl)amino)isoquinoline-1(2H)-ona Method: C3, m/z = 473.87 [M+H]+, Ret. tempo = 1.7 min. 1H-RMN (400 MHz, DMSO-d6) δ 1,26 (d, J = 7,0 Hz, 6H), 1,58 (s, 2H), 1,88 (s, 2H), 2,95– 3,03 (m, 2H), 3,52 (s, 2H), 3,69 (s, 1H), 6,63 (d, J = 6,9 Hz, 1H), 7,08 (s, 1H ), 7,17-7,32 (m, 3H), 7,35 (s, 1H), 7,43 (d, J = 7,3 Hz, 1H), 7,67 (s, 1H), 8 ,10 (s, 1H), 8,61 (s, 1H), 11,48 (s, 1H), 12,47 (s, 1H). AP 1-(6-amino-pyridin-3-yl)piperidin-4-ol (2-fluoro-5-isopropyl-phenyl) borosaurium I-289 N-(3-Fluoro-4-(8-((5-(4-hydroxy-piperidin-1-yl)pyridin-2-y)amino)-1-oxo-1,2-dihydro-isoquinoline-6- 2. yl)phenyl)cyclohexanecarboxamide Method: C3, m/z = 556.96 [M+H]+, Ret. tempo = 1.59 min. 1H-RMN (400 MHz, DMSO-d6) δ 1,27–1,32 (dt, J = 24,0, 11,9 Hz, 4H), 1,43 (q, J = 11,7 Hz, 2H ), 1,57 (s, 2H), 1,67 (d, J = 10,7 Hz, 1H), 1,76 (s, 3H), 1,84 (s, 3H), 2,36 (s , 1H), 3,00 (s, 2H), 3,51 (d, J = 11,7 Hz, 2H), 3,68 (s, 2H), 6,61 (d, J = 7,1 Hz , 1H), 7,06 (d, J = 8,8 Hz, 1H), 7,20 (d, J = 4,3 Hz, 2H), 7,45 (d, J = 8,8 Hz, 1H) ), 7,54 (t, J = 8,6 Hz, 1H), 7,65 (s, 1H), 7,76 (d, J = 13,4 Hz, 1H), 8,10 (s, 1H ), AP 1-(6-Amino-pyridin-3-yl)piperidin-4-ol PB1 8,59 (s, 1H), 10,19 (s, 1H), 11,45 (d, J = 5,6 Hz, 1H), 12,43 (s, 1H). I-290 6-(2-Fluoro-5-methoxy-4-(piperidin-1-carbonyl)phenyl)-8-((5-(4-hydroxy-piperidin-1-yl)pyridin-2-yl)amino)isoquinoline- 1(2H)-eins Method: C3, m/z = 572.58 [M+H]+, Ret. tempo = 1.49 min. 1H-NMR (400 MHz, DMSO-d6) δ 1,48 (s, 2H), 1,56 (s, 4H), 1,63 (s, 2H), 1,85 (s, 2H), 2, 56 (s, 3H), 2,93 (s, 2H), 3,18 (t, J = 5,5 Hz, 2H), 3,51 (s, 2H), 3,66 (s, 4H), 3,87 (s, 3H), 6,62 (d, J = 7,0 Hz, 1H), 7,00 (d, J = 11,7 Hz, 1H), 7,18-7,28 (m , 4H), 7,57 (s, 1H), 8,07 (s, 1H), 8,70 (s, 1H), 11,49 (s, 1H), 12,42 (s, 1H) AP 1-(6-Aminopyridin-3-yl)piperidin-4-ol CB1 I-291 N-(3- Fluor-4-(5-oxo-4- ((5-(tetrahidro- 2H-pyran-3- yl)pyridin- 2-yl)amino)- 5,6-dihidro- 1,6- Naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 542.50 [M+H]+, Ret. tempo = 1.74 min. RMN 1H (400 MHz, DMSO-d6) δ 1,16–1,37 (m, 2H), 1,43 (q, J = 12,1, 11,3 Hz, 2H), 1,66 (s, 3H), 1,63–1,71 (m, 4H), 1,74–1,89 (m, 2H), 2,32–2,4 (m, 2H), 2,86 (t, J = 10,8 Hz, 1H), 3,35–3,43 (m, 2H), 3,86 (s, 2H), 6,78 (d, J = 7,4 Hz, 1H), 7,29 ( d, J = 8,4 Hz, 1H), 7,53-7,66 (m, 1H), 7,79-7,93 (m, 4H), 8,42 (d, J = 2,3 Hz , 1H), 8,92 (s, 1H), 10,44 (s, 1H), 12,54 (s, 1H), 13,31 (s, 1H). PA AP1 PB1 I-292 N-(3-Fluor-5-(5-oxo-4-((5-(1,1,1-trifluoro-2-hidroxy-propan-2-yl)pyridin-2-yl)amino)- 5, 6-Dihidro-1,6-naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 570.68 [M+H]+, Ret. tempo = 1.66 min. 1H-NMR (400 MHz, DMSO-d6) δ 1,35 (ddq, J = 61,7, 23,8, 12,0 Hz, 4H), 1,48 (q, 2H), 1,68 (d , J = 11,3 Hz, 1H), 1,71 (s, 3H), 1,76 (s, 2H), 1,86 (d, J = 12,6 Hz, 2H), 6,75 (d , J = 7,3 Hz, 1H), 6,87 (s, 1H), 7,28 (d, J = 8,5 Hz, 1H), 7,55 (d, J = 9,2 Hz, 1H ), 7,70 (s, 1H), 7,82–7,98 (m, 1H), 8,06 (d, J = 8,6 Hz, 1H), 8,64 (s, 1H), 9 ,08 (s, 1H), 10,38 (s, 1H), 12,29 (s, 1H), 13,23 (s, 1H). BP 2-(4-aminofenil)-1,1,1-trifluoropropan-2-ol PB1 I-293 N-(3- Fluor-4- (4-((5-(2- Methyl- tetra-hidro- 2H-pyran-2- yl)pyridin- 2-yl)amino)- 5-oxo-5,6- di-hidro-1,6-naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 556.82 [M+H]+, Ret. tempo = 1.76 min. 1H-NMR (400 MHz, DMSO-d6) δ 0.87 (q, J = 7.1 Hz, 2H), 1.26 (d, J = 13.5 Hz, 5H), 1.40 (d, J = 34.7Hz, 3H), 1.52 (s, 4H), 1.67 (s, 2H), 1.81 (dd, J = 25.3, 12.1Hz, 4H), 2, 24 (d, J=12.7Hz, 1H), 3.69 (d, J=11.5Hz, 1H), 6.61 (d, J=7.2Hz, 1H), 7.09 ( d, J=8.5Hz, 1H), 7.45 (dd, J=18.2, 8.0Hz, 2H), 7.73-7.82 (m, 2H), 8.02 (t , J = 8.8Hz, 1H), 8.38 (d, J = 2.4Hz, 1H), 9.11 (s, 1H), 10.25 (s, 1H), 11.72 PA PA2 PB1 (d, J = 5,8 Hz, 1H), 12,75 (s, 1H). I-294 N-(3- Fluor-4- (4-((5-(4- hidroxy-4- methyl-piperidin-1- yl)pyridin- 2-yl)amino)-5-oxo-5,6-dihidro- 1,6-nafthyridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 571.55 [M+H]+, Ret. tempo = 1.64 min. RMN de 1H (400 MHz, DMSO-d6) δ 1,18 (s, 3H), 1,22–1,34 (m, 3H), 1,37–1,51 (m, 2H), 1,59 (s, 4H), 1,59 (d, J = 11,4). Hz, 1H), 1,68 (d, J = 10,6 Hz, 1H), 1,79 (d, J = 11,7 Hz, 2H), 1,85 (d, J = 12,7 Hz, 1H), 2,40–2,35 (m, 2H), 3,14 (s, 2H), 3,08-3,19 (m, 1H), 3,32 (d, J = 11,3 Hz , 2H), 4,35 (s, 1H), 6,58 (d, J = 7,3 Hz, 1H), 7,03 (d , J = 8,9 Hz, 1H), 7,37–7 ,53 (m, 3H), 7,27–7,81 (m, 1H), 8,01 (t, J = 8,8 Hz, 1H), 8,09 (d, J = 3,1 CP 1-(6-amino-pyridin-3-yl)-4-methyl-piperidin-4-ol PB1 Hz, 1H), 8,74 (s, 1H), 10,25 (s, 1H), 11,65 (s, 1H), 12,39 (s, 1H). I-295 N-(4-(4-((5-(2-oxa-7-azaspiro[3.5]nonan-7-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1, 6-nafthyridin-2-yl)-3-fluor-phenyl)cyclo-hexane-carboxamide Method: C3, m/z = 583.98 [M+H]+, Ret. tempo = 1.37 min. RMN 1H (400 MHz, DMSO-d6) δ 1,24 (s, 1H), 1,25–1,36 (m, 3H), 1,43 (d, J = 12,5 Hz, 2H), 1 ,77 (s, 1H), 1,80–1,92 (m, 3H), 3,17 (s, 3H), 3,71 (s, 2H), 3,86 (s, 2H), 3, 98 (s, 2H), 4,39 (s, 2H), 5,23 (s, 1H), 6,67 (d , J = 7,2 Hz, 1H), 7,35 (d, J = 8 ,9 Hz, 1H), 7,42–7,53 (m, 2H), 7,83 (d, J = 14,1 Hz, 1H), 8,06 (t, J = 8,9 Hz). , 1H), 8,21–8,28 (m, 1H), 8,85 (d, J = 2,9 Hz, 1H), 9,18 (s, 1H), 10,29 (s, 1H) , 11,85 (s, 1H), 13,11 9s, 1H ). BP 5-(2-Oxa-7-Azaspiro[3.5]Nonan-7-yl)pyridin-2-amine PB1 I-296 N-(3-fluor-4-(5-oxa-4-((5-(tetrahidro- 2H-pyran- 4-yl)pyridin-2-yl)amino)-5,6-dihidro- 1,6- Naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 542.88 [M+H]+, Ret. tempo = 1.58 min. RMN 1H (400 MHz, DMSO-d6) δ 1,24 (s, 2H), 1,25–1,37 (m, 2H), 1,43 (q, J = 12,1, 11,5 Hz, 2H), 1,65–1,88 (m, 6H), 2,37 (dd, J = 13,5, 10,2 Hz, 1H), 2,78 (s, 1H), 3,39–3 ,50 (m, 2H), 3,92–4,00 (m, 2H), 6,63 (d, J = 7,3 Hz, 1H), 7,09 (d, J = 8,5 Hz, 1H), 7,48 (d, J = 8,7 Hz, 2H), 7,69–7,83 (m, 2H), 7,99 (t, J = 8,8 Hz, 1H), 8, 31 (d, J = 2,4 Hz, 1H), 9,01 (s, 1H), 10,27 (s, 1H), 11,80 (s, 1H), 12,75 (s, 1H). BP 5-(Tetrahydro-2H-pyran-4-yl)pyridin-2-amine PB1 I-297 (R)-N-(4- (4-((5-(3-amino-piperidin-1- yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hidro-1, 6- Naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: J, m/z = 556.3 [M + H]+, Ret. Tempo = 4.35 min. RMN 1H (400 MHz, DMSO-d6) δ 1,23–1,33 (m, 8H), 1,37–1,48 (m, 1H), 1,68 (d, J = 13,4 Hz, 1H), 1,80 (s, 4H), 1,85 (d, J = 15,0 Hz, 2H), 2,85 (d, J = 9,4 Hz, 1H), 3,08 (s, 1H), 3,49–3,60 (m, 2H), 6,59 (d, J = 7,3 Hz, 1H), 7,07 (d, J = 9,2 Hz, 1H), 7, 38–7,51 (m, 2H), 7,79 (d, J = 14,2 Hz, 1H), 8,00–8,12 (m, 2H), 8,80 (s, 1H), 10 ,25 (s, 1H), 12,44 (s, 1H). BP tert-butyl-(R)-(1-(6-amino-pyridin-3-yl)piperidin-3-yl)carbamate PB1 I-298 N-(4-(4-((5-(2,2- Dimethyl-5-oxo-pyrrolidin-1- yl)pyridin-2- yl)amino)-5-oxo-5,6-di-hidro- 1, 6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexancarboxamide Method: C3, m/z = 569.73 [M+H]+, Ret. tempo = 1.57 min. RMN 1H (400 MHz, DMSO-d6) δ 1,23 (s 6H), 1,22–1,38 (m, 4H), 1,44 (q, J = 12,1 Hz, 2H), 1, 68 (d, J = 11,9 Hz, 1H), 1,82 (dd, J = 26,6, 12,2 Hz, 4H), 2,03 (t, J = 7,9 Hz, 2H), 2,40 (d, J = 11,7 Hz, 1H), 6,71 (d, J = 7,1 Hz, 1H), 7,27 (s, 1H), 7,52 (d, J = 8 ,6 Hz, 1H), 7,57–7,69 (m, 1H), 7,83 (d, J = 14,0 Hz, 1H), 7,99 (d, J = 8,8 Hz, 1H ). ), 8,21 (s, 1H), 9,06 (s, 1H), 10,33 (s, 1H), 12,08 (s, 1H), 13,12 (s, 1H). BP 1-(6-amino-pyridin-3-yl)-5,5-dimethylpyrrolidin-2-on PB1 I-299 (S)-N-(3-Fluoro-4-(4-((5-(3-hydroxy-piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro- 1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide Method: C3, m/z = 557.5 [M+H]+, Ret. tempo = 1.59 min. 1H-RMN (400 MHz, DMSO-d6) δ 1,25–1,31 (m, 5H), 1,39 (q, J = 12,2 Hz, 2H), 1,57 (s, 1H), 1,68 (d, J = 11,1 Hz, 1H), 1,77-1,81 (m, 5H), 2,34 (t, J = 12 Hz, 1H), 2,58 (dd, J = 11,4, 8,5 Hz, 1H), 2,72 (t, J = 11,9 Hz, 1H), 3,47 ( d, J = 11,8 Hz, 1H), 3,53–3 ,66 (m, 2H), 4,86 ​​(d, J = 4,6 Hz, 1H), 6,58 (d, J = 7,4 Hz, 1H), 7,05 (t, J = 11,9 Hz, 1H), 7,39-7,50 (m, 3H), 7,77 (d, J = 14,2 Hz, 1H), 7,96-8,09 (m, 2H) ), 8,74 (s, 1H), 10,24 (s, 1H), 11,66 (s, 1H), 12,42 (s, 1H). BP (S)-1-(6-aminopyridin-3-yl)piperidin-3-ol PB1 I-300 N-(4-(4-((5-(3,6-Diazabicyclo[3.1.1]heptan-3-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1, 6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexancarboxamide Method: C3, m/z = 554.88 [M+H]+, Ret. tempo = 1.34 min. RMN 1H (400 MHz, DMSO-d6) δ 0,87 (s, 1H), 1,22–1,31 (m, 4H), 1,43 (d, J = 12,0 Hz, 2H), 1 ,56 (s, 1H), 1,74–1,85 (m, 2H), 1,83–1,94 (m, 2H), 2,35–2,38 (m, 2H), 2,88 (s, 1H), 3,71 (d, J = 11,5 Hz, 1H), 3,84 (d, J = 11,6 Hz). , 1H), 4,44 (s, 2H), 6,58 (d, J = 7,2 Hz, 1H), 6,84 (s, 1H), 7,09 (s, 1H), 7,15 (d, J = 8,7 Hz, 1H), 7,34 (d, J = 8,9 Hz, 1H), 7,39–7,49 (m, 2H), 7,79 (d, J = 14,4 Hz, 1H), 7,97–8,07 (m, 1H), 8,65 (s, 1H), 10,28 (s, 1H), 11,64 (s, 1H), 12, 33 (s, 1H) PA PA4 PB1 I-301 N-(4-(4-((5-(3,8-Diazabicyclo[3.2.1]octan-3-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1, 6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexancarboxamide Method: H, m/z = 568 [M + H]+, Ret. time = 3.71 min. RMN 1H (400 MHz, DMSO-d6) δ 1,16–1,36 (m, 4H), 1,43 (q, J = 11,7, 11,2 Hz, 2H), 1,58–1, 88 (m, 8H), 2,26–2,39 (m, 2H), 2,72-2,87 (m, 2H), 3,50 (s, 4H), 4,24 (s, 1H) , 6,53 (d, J = 7,1 Hz, 1H), 6,99 (d, J = 9,0 Hz, 1H), 7,33 (dt, J = 9,0, 5,0 Hz, 1H), 7,44 (dd, J = 10,0, 8,0 Hz, 2H), 7,74 (dd, J = 14,2, 2,4 Hz, 1H), 7,90–8,02 (m, 2H), 8,62 (d, J = 3,8 Hz, 1H), 10,24 (s, 1H), 11,59 (s, 1H), 12,64 (s, 1H). PA PA5 PB1 I-302 N-(3-Fluor-4-(5-Oxo-4-((5-(THF-3-yl)Pyridin-2-yl)Amino)-5,6-Dihidro-1,6-Naftiridin-2- yl)phenyl)cyclohexanocarboxamide Method: C3, m/z = 528.82 [M+H]+, Ret. tempo = 1.57 min. 1H-RMN (400 MHz, DMSO-d6) δ 1,17–1,29 (m, 3H), 1,37–1,43 (m, 2H), 1,67 (d, J = 11,3 Hz , 1H), 1,81 (dd, J = 25,6–12,3 Hz). , 1H), 1,87–2,02 (m, 4H), 2,34 (ddd, J = 12,3, 8,4, 4,5 Hz, 2H), 3,41 (q, J = 7 ,8 Hz, 1H), 3,56 (t, J = 7,9 Hz, 1H), 3,81 (q, J = 7,8 Hz, 1H), 3,92-4,08 (m, 2H ), 6,61 (d, J = 7,2 Hz, 1H), 7,07 (d, J = 8,4 Hz, 1H), 7,39- 7,50 (m, 2H), 7,68 -7,82 BP PA3 PB1 (m, 2H), 8.02 (t, J = 8.8 Hz, 1H), 8,31 (d, J = 2,5 Hz, 1H), 9,04 (s, 1H), 10,24 (s, 1H), 11,72 (s, 1H), 12,69 (s, 1H) I-303 (R)-N-(3-Fluoro-4-(4-((5-(3-hydroxy-piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro- 1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide Method: C3, m/z = 558.38 [M+H]+, Ret. tempo = 1.54 min. RMN 1H (400 MHz, DMSO-d6) δ 1,16–1,41 (m, 4H), 1,43 (d, J = 11,9 Hz, 2H), 1,55 (d, J = 12, 0 Hz, 1H), 1,67 (d, J = 11,0 Hz, 1H), 1,73–1,93 (m, 5H), 2,32–2,38 (m, 1H), 2,470– 2,58 (m, 2H), 2,67–2,72 (m, 1H), 3,34–3,46 (m, 1H), 3,54-3,61 (m, 2H), 4, 84 (s, 1H), 6,57 (d, J = 7,2 Hz, 1H), 7,02 (d, J = 8,9 Hz, 1H), 7,37-7,45 (m, 3H ), 7,75 (dd, J = 14,2, 2,0 Hz, 1H), 7,95–8,04 (m, 2H), 8,72 (s, 1H), BP (R)-1-(6-aminopyridin-3-yl)piperidin-3-ol PB1 10,22 (s, 1H), 11,62 (d, J = 5,7 Hz, 1H), 12,37 (s, 1H) I-304 N-(3-Fluoro-4-(4-((5-((3aR,6aS)-hexahidro-pyrano[3,4-c]pyrrol-2(1H)-yl)pyridin-2-yl)amino) - 5-oxo-5,6-dihidro-1,6-nafthyridin-2-yl)phenyl)cyclohexanocarboxamide Method: C3, m/z = 568.2 [M+H]+, Ret. tempo = 1.64 min. 1H-NMR (400 MHz, DMSO-d6) δ 1,26 (dq, J = 22,8, 12,1, 11,6 Hz, 3H), 1,42 (q, J = 12,11, 11, 5 Hz, 2H), 1,67 (d, J = 11,7 Hz, 1H), 1,80 (dd, J = 24,4, 12,2 Hz, 4H), 2,32–2,38 ( m, 1H), 2,96 (d, J = 10,8 Hz, 2H), 3,04 (s, 2H), 6,56 (d, J = 7,3 Hz, 1H), 7,06 ( d, J = 8,8 Hz, 1H), 7,21 (dd, J = 8,8, 3,0 Hz, 1H), 7,36-7,47 (m, 2H), 7,77 (dd , J = 14,3, 2,0 Hz, 1H), 7,85 (d, J = 3,0 Hz, 1H), 8,01 (t, J = 8,8 Hz, 1H), BP PA34 PB1 8,28 (s, 1H), 8,64 (s, 1H), 10,25 (s, 1H), 12,30 (s, 1H). I-305 N-(4-(4-((5-(5-Azaspiro[2.4]heptan-5-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihidro-1,6-nafthyridin- 2-yl)-3-fluorophenyl)cyclohexane-carboxamide Method: C3, m/z = 554.13 [M+H]+, Ret. tempo = 1.90 min. RMN 1H (400 MHz, DMSO-d6) δ 0,66 (d, J = 8,2 Hz, 3H), 1,23–1,34 (m, 4H), 1,37–1,48 (m, 2H), 1,69 (s, 1H), 1,77– 1,85 (m, 4H), 1,94 (t, J = 6,7 Hz, 2H), 2,34-2,40 (m , 1H), 3,22 (s, 2H), 3,45 (t, J = 6,7 Hz, 2H), 6,56 (d , J = 7,3 Hz, 1H), 7,06 (s , 1H), 7,39–7,46 (m, 2H), 7,75 (d, J = 14,1 Hz, 2H), 8,01 (t, J = 8,8 Hz, 1H), 8 ,22 (s, 1H), 8,54 (s, 1H), 10,23 (s, 1H), 11,60 (s, 1H), 12,22 (s, 1H). BP 5-(5-Azaspiro[2.4]heptan-5-yl)pyridin-2-amine PB1 I-306 N-(4-(4- ((5-(1,4- diazepan-1- yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hidro-1,6-naphthyridine- 2-yl)-2-fluorophenyl)cyclohexane-carboxamide Method: J, m/z = 556.2 [M + H]+, Ret. Time = 3.37 min. RMN 1H (400 MHz, DMSO-d6) δ 1,19–1,32 (m, 3H), 1,39–1,44 (m, 2H), 1,69 (s, 1H), 1,77– 1,86 (m, 3H), 2,08 (s, 1H). ), 2,38 (s, 1H), 3,18 (s, 2H), 3,29 (s, 2H), 3,57 (t, J = 6,1 Hz, 3H), 3,77 (t , J = 6,1 Hz, 3H), 6,75 (d , J = 7,5 Hz, 1H), 7,25 (d, J = 9,4 Hz, 1H), 7,41 (s, 1H ), 7,55 (d, J = 8,9 Hz, 1H), 7,85 (dd, J = 28,7, 11,5 Hz). , 2H), 8,07 (d, J = 3,0 Hz, 1H), 8,60 (d, J = 5,7 Hz, 1H), 8,77 (s, 1H), 10,44 (s , 1H), 12,44 (s, 1H), 13,05 (s, 1H). PA PA7 PB1 I-307 Example 25: N-(4-(4-((5-((1R,4R))-2,5- Diazaspiro[2.2.1]heptan-2- yl)pyridin-2- yl)amino)- 5- . oxo-5,6-dihydro-1,6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexanecarboxamide Method: C3, m/z = 554.2 [M+H]+, Ret. tempo = 1.65 min. 1H-NMR (400 MHz, DMSO-d6) δ 1,25 (q, J = 12,1, 11,4 Hz, 3H), 1,35–1,46 (m, 2H), 1,67 (d , J = 11,2 Hz, 1H), 1,75–1,90 ( m, 5H), 2,34–2,37 (m, 3H), 2,96 (q, J = 10,3 Hz, 2H), 3,06 (d, J = 9,3 Hz, 1H), 3,56 (d, J = 8,8 Hz, 1H), 3,90 (s, 1H), 4,49 (s, 1H), 6,55 (d, J = 7,3 Hz, 1H), 7,03 (d, J = 8,9 Hz, 1H), 7,15 (dd, J = 8,8, 3,0 hz, 1H). ), 7,39 (d, J = 7,3 Hz, 1H), 7,46 (d, J = 9,0 Hz, 1H), 7,72–7,79 (m, tert-Butyl BP (1R,4R)-5-(6-amino-pyridin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate PB1 2H), 7.99 (t, J=8.8Hz, 1H), 8,60 (s, 1H), 10,30 (s, 1H), 11,60 (s, 1H), 12,27 (s, 1H) I-308 (R)-N-(3- Fluor-4- (4-((5-(4- hidroxy-3,3- dimethyl-piperidin-1- yl)pyridin-2- yl)amino)- 5-oxo- 5,6-Dihidro-1,6-nafthyridin-2-yl)phenyl)cyclohexancarboxamide Method: C3, m/z = 586.19 [M+H]+, Ret. tempo = 1.60 min. 1H-NMR (400 MHz, DMSO-d6) δ 0,94 (d, J = 7,6 Hz, 6H), 1,26 (dq, J = 23,2, 12,3 Hz, 3H), 1, 43 (q, J = 11,6, 11,1 Hz, 2H), 1,61-1,67 (m, 2H), 1,71-1,84 (m, 4H), 2,32-2, 38 (m, 1H), 2,50-5,54 (m, 2H), 2,74-2,81 (m, 1H), 3,16-3,25 (m, 2H), 3,47 ( d, J = 12,3 Hz, 1H), 4,63 (d, J = 4,6 Hz, 1H), 6,56 (d, J = 7,2 Hz, 1H), 7,00 (d, J = 8,9 Hz, 1H), 7,37–7,45 (m, 3H), 7,75 (dd, J = 14,3, 2,0 Hz, 1H), 7,95– BP PA6 PB1 8,04 (m, 2H), 8,73 (s, 1H), 10,22 (s, 1H), 11,61 (d, J = 5,8 Hz, 1H), 12,37 (s, 1H). I-309 N-(3-Fluor-4-(4-((5-(4-(methylamino)piperidin-1-y)pyridin-2-yl)amino)-5-oxo-5,6-di-hydro-1 ,6 -Naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 570.88 [M+H]+, Ret. time = 1.35 min. RMN 1H (400 MHz, DMSO-d6) δ 1,11–1,32 (m, 3H), 1,37–1,46 (m, 2H), 1,49–1,60 (m, 2H), 1,67 (d, J = 11,1 Hz, 1H), 1,75–1,84 (m, 4H), 2,01 (d, J = 12,3 Hz, 2H), 2,33–2 ,38 (m, 2H), 2,75 (t, J = 11,9 Hz, 3H), 2,97 (s, 2H), 3,76 (d, J = 12,5 Hz, 2H), 6 ,57 (d, J = 7,3 Hz, 1H), 7,04 (d, J = 8,9 Hz, 1H), 7,37–7,47 (m, 2H), 7,51 (dd, J = 8,9, 3,1 Hz, 1H), 7,78 (dd, J = 14,3, 2,0 Hz, 1H), 8,00 (t, J = 8,9 Hz, PA PA8 PB1 1H), 8,10 (d, J = 3,1 Hz, 1H), 8,77 (s, 1H), 10,24 (s, 1H), 12,42 (s, 1H) I-310 N-(4-(4-((5-(3,8-Diazaespiro[3.2.1]octan-8-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1, 6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexancarboxamide Method: C3, m/z = 568.2 [M+H]+, Ret. tempo = 1.69 min. RMN de 1H (400 MHz, DMSO-d6) δ 1,22–1,34 (m, 3H), 1,38–1,47 (m, 2H), 1,66–1,69 (m, 1H) , 1,77–1,86 (m, 4H), 2,04–2,09 (m, 4H), 2,37 (d, J = 12,3 Hz, 1H), 3,04-3,14 (m, 4H), 4,51 (s, 2H), 6,74 (s, 1H), 7,28 (s, 1H), 7,53 (d, J = 8,9 Hz, 2H), 7 ,87 (d, J = 13,4 Hz, 2H), 8,19 (s, 1H), 8,72 (s, 1H), 8,99 (s, 1H), 10,41 (s, 1H) , 12,37 (s, 1H), 13,04 (s, 1H) BP tert-butyl-8-(6-amino-pyridin-3-yl)-3,8-diazaspiro[3.2.1]octan-3-carboxylate PB1 I-311 (S)-N-(4- (4-((5-(3-amino-piperidin-1- yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hidro-1, 6- Naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: C3, m/z = 556.5 [M+H]+, Ret. tempo = 1.47 min. RMN 1H (400 MHz, DMSO-d6) δ 1,17–1,32 (m, 3H), 1,37–1,45 (m, 3H), 1,60–1,75 (m, 2H), 1,80–1,88 (m, 7H), 2,37 (dd , J = 13,3, 9,9 Hz, 1H), 2,88 (q, J = 8,1 Hz, 2H), 3 ,20 (dt, J = 8,1 Hz, 2H), 3,20 (dt, J = 11,0, 5,4 Hz, 2H), 6,58 (d, J = 7,3 Hz, 1H) , 7,07 (d, J = 8,8 Hz, 1H), 7,39-7,47 (m, 3H), 7,77 (d, J = 14,2 Hz, 1H), 7,96- 8,11 (m, 2H), 8,32 (s, 1H), 8,80 (s, 1H), 10,28 (s, 1H), 12,44 (s, 1H) BP tert-butyl-(S)-(1-(6-amino-pyridin-3-yl)piperidin-3-yl)carbamate PB1 I-312 N-(3-Fluoro-4-(4-((6-Fluoro-2-methyl-1,2,3,4-tetrahydro-isoquinolin-7-yl)amino)-5-oxo-5,6-dihydro -1,6-naphthyridine-2-yl)phenyl)cyclohexanecarboxamide Method: C3, m/z = 542.99 [M+H]+, Ret. tempo = 1.39 min. RMN 1H (400 MHz, DMSO-d6) δ 1,15–1,28 (m, 4H), 1,35–1,41 (m, 2H), 1,63–1,82 (m, 5H), 2,36 (d, J = 12,9 Hz, 1H), 2,43 (s, 2H), 2,74 (s, 2H), 2,88 (s, 2H), 3,60 (s, 2H) ), 6,56 (d, J = 7,3 Hz, 1H), 7,04 (s, 1H), 7,19 (d , J = 11,2 Hz, 1H), 7,32 (d, J = 8,0 Hz, 1H), 7,42 (d, J = 7,7 Hz, 2H), 7,72 (d, J = 14,6 Hz, 1H), 7,93-8,06 BP PA9 PB1 (m, 1H), 10,23 (s, 1H), 11,38 (s, 1H), 11,62 (s, 1H) I-313 N-(4-(4- ((5-((1R,4R)-2,5-Diazabicyclo[2.2.1]heptan-2-yl)pyridin-2-yl)amino)-5-oxo-5, 6-Dihidro-1,6-naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: C3, m/z = 554.2 [M+H]+, Ret. tempo = 1.65 min. RMN 1H (400 MHz, DMSO-d6) δ 1,19–1,33 (m, 3H), 1,39–1,47 (m, 2H), 1,67 (d, J = 9,8 Hz, 2H), 1,78–1,85 (m, 5H), 2,34-2,40 (m, 1H), 2,55 (s, 1H), 2,78-2,96 (m, 3H) , 3,52-3,54 (m, 1H), 3,62 (s, 1H), 4,40 (s, 1H), 6,56 (d, J = 7,3 Hz, 1H), 7, 02 (d, J = 8,8 Hz, 1H), 7,11 (dd, J = 8,8, 3,0 Hz, 1H), 7,36-7,49 (m, 2H), 7,76 (dd, J = 17,7, 2,7 Hz, 2H), 8,00 (t, J = 8,8 Hz, 1H), BP (1R,4R)-terc.-butyl-5-(6-amino-pyridin-3-yl)-2,5-diaza-bicyclo[2.2.1]heptan-2-carboxylate PB1 8,58 (s, 1H), 10,23 (s, 1H), 11,62 (s, 1H), 12,24 (s, 1H). I-314 (S)-N-(3- Fluor-4- (4-((5-(4- hidroxy- 3,3-dimethyl-piperidin-1- yl)pyridin-2- yl)amino)- 5-oxo- 5,6-dihydro-1,6-naphthyridin-2-yl)phenyl)cyclohexancarboxamide Method: C3, m/z = 585.93 [M+H]+, Ret. tempo = 1.56 min. RMN 1H (400 MHz, DMSO-d6) δ 0,94 (s, 3H), 0,96 (s, 3H), 1,13–1,31 (m, 3H), 1,39–1,48 ( m, 2H), 1,66–1,68 (m, 2H). ), 1,74–1,86 (m, 4H), 2,34–2,40 (m, 3H), 2,75 (t, J = 11,9 Hz, 1H), 3,18–3, 26 (m, 2H), 3,46–3,49 (m, 1H), 4,64 (s, 1H), 6,57 (d, J = 7,3 Hz, 1H), 7,04 (d , J = 8,9 Hz, 1H), 7,37–7,47 (m, 3H), 7,74–3,77 (m, 1H), 7,98 -8,05 (m, 2H), 8,73 (s, 1H), 10,24 (s, 1H), 11,62 (s, 1H), 12,42 BP PA33 PB1 (s, 1H) I-315 N-(4-(4-((5-(4-Oxa-7-azaspiro[2.5]octan-7-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1, 6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexancarboxamide Method: F, m/z = 569.3 [M + H]+, Ret. Time = 7.44 min. RMN 1H (400 MHz, DMSO-d6) δ 0,61–0,76 (m, 4H), 1,17–1,26 (m, 3H), 1,32 (ddt, J = 60,4, 23 ,0 12,1 Hz, 2H), 1,80 (dd, J = 24,7, 12,3 Hz, 4H), 3,11–3,24 (m, 4H), 3,81 (d, J = 9,6 Hz, 2H), 6,57 (d, J = 7,2 Hz, 1H), 7,05 (d, J = 9,0 Hz, 1H), 7,36–7,47 (m , 1H), 7,47 (dd, J = 9,1, 2,8 Hz, 2H), 7,75 (dd, J = 14,3, 2,0 Hz, 1H), 7,95–8, 09 (m, 2H), 8,79 (s, 1H), 10,23 (s, 1H), 11,63 (d, J = 5,4). BP PA10 PB1 Hz, 1H), 12,42 (s, 1H) I-316 N-(4-(4-((5-(3-(Dimethylamino)pyrrolidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridine - 2 -yl)-3-fluorphenyl)cyclohexane-carboxamide Method: F, m/z = 570.3 [M + H]+, Ret. Time = 6.56 min. RMN 1H (400 MHz, DMSO-d6) δ 1,16–1,33 (m, 4H), 1,40 (dd, J = 24,7, 13,3 Hz, 2H), 1,68 (d, J = 11,6 Hz, 2H), 1,81 (dd, J = 24,7, 12,0 Hz, 4H), 2,24 (s, 1H), 2,40 (s, 6H), 3, 22–3,34 (m, 2H), 3,49 (dt, J = 31,6, 8,6 Hz, 2H), 6,56 (d , J = 7,2 Hz, 1H), 7,02 –7,16 (m, 2H), 7,35–7,49 (m, 2H), 7,72–7,82 (m, 1H), 8,01 (t, J = 8,9 Hz, 1H ), 8,60 (s, 1H), 10,24 (s, 1H), 11,60 (d, BP 5-(3-(Dimethylamino)pyrrolidin-1-yl)pyridin-2-amine PB1 J = 5,9 Hz, 1H), 12,26 (s, 1H). I-317 N-(4-(4-((5-(2-Azaspiro[4.4]nonan-2-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridine- 2-yl)-3-fluorophenyl)cyclohexane-carboxamide Method: H, m/z = 581.45 [M + H]+, Ret. time = 5.77 min. RMN de 1H (400 MHz, DMSO-d6) δ 1,26 (h, J = 11,7 Hz, 3H), 1,43 (q, J = 11,8 Hz, 2H), 1,63 (dd, J = 31,2, 6,7 Hz, 9H), 1,74 - 1,92 (m, 7H), 2,39 (d, J = 12,1 Hz, 1H), 3,16 (s, 3H ), 6,55 (d, J = 7,3 Hz, 1H), 7,03 (s, 2H), 7,35-7,49 (m, 2H), 7,74 (d, J = 17, 6 Hz, 2H), 8,00 (t, J = 8,8 Hz, 1H), 8,53 (s, 1H), 10,23 (s, 1H), 11,58 (d, J = 5, 9 Hz, 1H), 12,19 (s, 1H). BP PA11 PB1 I-318 N-(3- Fluor-4- (4-((5-(4- isopropyl-piperazin-1- yl)pyridin-2- yl)amino)- 5-oxo-5,6-dihydro-1,6- Naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: H, m/z = 584.40 [M + H]+, Ret. time = 4.27 min. RMN 1H (400 MHz, DMSO-d6) δ 1,09–1,31 (m, 10H), 1,39–1,48 (dd, 4H), 1,68 (d, 2H), 1,77– 1,86 (m, 5H), 2,37 (m, 2H). ), 3,14 (s, 4H), 6,59 (d, J = 7,4 Hz, 1H), 7,06 (d, J = 9,0 Hz, 1H), 7,40–7,50 (m, 3H), 7,78 (d, J = 14,3 Hz). , 1H), 8,01 (t, J = 8,8 Hz, 1H), 8,11 (s, 1H), 8,80 (s, 1H), 10,25 (s, 1H), 11,65 (d, J = 5,8 Hz, 1H), 12,46 (s, 1H) BP 5-(4-Isopropil-piperazin-il)piridin-2-amina PB1 I-319 N-(3-Fluoro-4-(4-((3-Methoxy-7-Methyl-5,6,7,8-Tetrahydro-Naphthalen-2-yl)Amino)-5-Oxo-5,6-Dihydro -1,6-naphthyridin-2-yl)phenyl)cyclohexane Method: J, m/z = 556.2 [M + H]+, Ret. Time = 3.68 min. RMN 1H (400 MHz, DMSO-d6) δ 1,15–1,45 (m, 6H), 1,68 (d, J = 11,1 Hz, 1H), 1,81 (t, J = 17, 1 Hz, 2H), 2,37 (t, 2H), 2,94 (s, 3H), 3,13 (s, 2H), 3,36 (s, 2H), 3,87 (s, 3H) , 4,26 (s, 1H), 4,46 (d, J = 15,2 Hz, 1H), 6,72 (s, 1H), 6,90 (s, 1H), 7,15 (s, 1H), 7,41 (s, 1H), 7,49 (d, J = 8,7 Hz, 1H), 7,84 (d, J = 14,0 Hz, 3H), 10,14 (s, 1H), 10,41 (s, 1H), 12,11 (s, 1H) BP PA12 PB1 I-320 N-(4-(4-((5-cyclohexylpyridin-2-yl)amino)-5-oxo-5,6-dihidro-1,6-naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: H, m/z = 540.40 [M + H]+, Ret. time = 5.73 min. RMN 1H (400 MHz, DMSO-d6) δ 1,23–1,43 (m, 11H), 1,69 (d, 2H), 1,80 (d, J = 12,0 Hz, 8H), 2 ,35 (t, 1H), 6,59 (d, J = 7,3 Hz, 1H), 7,02 (d, J = 8,4 Hz, 1H), 7,41-7,46 (m, 2H), 7,66 (dd, J = 8,5, 2,5 Hz, 1H), 7,75 (d, J = 14,0 Hz, 1H), 8,00 (t, J = 8,8 Hz, 1H), 8,24 (d, J = 2,4 Hz, 1H), 9,01 (s, 1H), 10,23 (s, 1H), 11,68 (s, 1H), 12, 62 (s, 1H) PA PA13 PB1 I-321 N-(4-(4- ((5-(2,6-Diazaspiro[3.3]heptan-2-yl)pyridin-2-yl)amino)-5-oxo-5,6-di-hidro-1, 6- Naftiridin-2-yl)-2-fluorophenyl)cyclohexancarboxamida Method: C3, m/z = 554.98 [M+H]+, Ret. tempo = 1.39 min. RMN 1H (400 MHz, DMSO-d6) δ 1,17–1,47 (m, 5H), 1,68 (d, J = 11,7 Hz, 1H), 1,77–1,86 (m, 4H), 2,34 (s, 1H), 3,64 (s, 5H), 4,08 (s, 4H), 4,19 (t, J = 6,2 Hz, 4H), 6,72 ( d, J = 7,3 Hz, 1H), 7,08 (dd, J = 8,7, 3,0 Hz, 1H), 7,20 (d, J = 8,8 Hz, 1H), 7, 53 (d, J = 8,6 Hz, 1H), 7,75 (d, J = 2,9 Hz, 2H), 7,86 (q, J = 9,5, 8,4 Hz, 2H), 8,60 (d, J = 17,7 Hz, 3H), 10,41 (s, 1H), 12,34 (s, 1H), 12,94 (s, 1H) BP terc-butyl-6-(6-amino-pyridin-3-yl)-2,6-diazaspiro[3,3]heptano-2-carboxylate PB1 I-322 N-(3- Fluor-4- (4-((5-(3- Metoxi-pyrrolidin- 1-yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hydro-1, 6- Naftiridin-2-yl)phenyl)cyclohexane-carboxamida Method: C3, m/z = 557.83 [M+H]+, Ret. tempo = 1.65 min. RMN 1H (400 MHz, DMSO-d6) δ 1,19–1,30 (m, 3H), 1,39–1,47 (m, 2H), 1,67 (d, J = 11,5 Hz, 1H), 1,76–1,85 (m, 3H), 2,06-2,11 (m, 2H), 2,37 (t, J = 11,5 Hz, 1H), 3,28 (s , 3H), 3,31-3,35 (m, 3H), 3,43-3,47 (m, 2H), 4,11 (s, 1H), 6,56 (d, J = 7,3 , 1H), 7,03–7,11 (m, 2H), 7,37–7,46 (m, 2H), 7,73–7,76 (m, 2H), 8,00 (t, J = 8,8 Hz, 1H), 8,56 (s, 1H), 10,23 (s, 1H), 11,59 (d, J = 5,9 Hz, 1H), 12,23 (s, 1H ) BP 5-(3-Methoxypyrrolidin-1-yl)pyridin-2-amine PB1 I-323 N-(3-Fluoro-4-(4-((6-methoxy-1,2,3,4-tetrahydro-isoquinolin-7-yl)amino)-5-oxo-5,6-dihydro-1,6 - Naphthyridine-2-yl)phenyl)cyclohexane-carboxamide Method: C3, m/z = 542.4 [M+H]+, Ret. tempo = 1.48 min. RMN 1H (400 MHz, DMSO-d6) δ 1,21–1,29 (m, 3H), 1,37–1,46 (q, J = 11,8 Hz, 2H), 1,67 (d, J = 10,9 Hz, 1H), 1,80 (t, J = 16,7 Hz, 4H), 2,34 (s, 1H), 2,77 (s, 3H), 3,05 (t, J = 5,8 Hz, 2H), 3,80 (s, 3H), 3,88 (s, 1H), 6,52 (d, J = 7,2 Hz, 1H), 6,89 (s, 1H), 7,16 (dd, 2H), 7,39 (dd, J = 12,2, 8,1 Hz, 2H), 7,72 (d, J = 14,4 Hz, 1H), 8, 01 (t, J = 8,9 Hz, 1H), 10,21 (s, 1H), 11,27 (s, 1H), 11,48 (s, 1H) BP PA14 PB1 I-324 N-(4-(4-((5-cyclopentylpyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: C3, m/z = 526.66 [M+H]+, Ret. tempo = 2.05 min. RMN 1H (400 MHz, DMSO-d6) δ 1,17–1,32 (m, 3H), 1,38–1,46 (m, 2H), 1,53 (d, J = 9,7 Hz, 2H), 1,65 (t, 3H), 1,78– 1,84 (m, 6H), 2,02 (s, 2H), 2,36 (t, J = 8,4 Hz, 1H), 2,98 (t, J = 8,3 Hz, 1H), 6,60 (d, J = 7,2 Hz, 1H), 7,04 (d, J = 8,4 Hz, 1H), 7, 41–7,46 (q, 2H), 7,68–7,78 (dd, 2H), 8,00 (t, J = 8,8 Hz, 1H), 8,27 (s, 1H), 9 ,00 (s, 1H), 10,23 (s, 1H), 11,71 (s, 1H), 12,65 (s, 1H) BP PA15 PB1 I-325 N-(4-(4-((5-(1-oxa-7-azaspiro[3.5] nonan-7- yl)pyridin-2- yl)amino)-5-oxo-5,6-di-hydro- 1, 6-naphthyridin-2-yl)-3-fluorophenyl)cyclohexancarboxamide Method: C3, m/z = 583.88 [M+H]+, Ret. tempo = 1.53 min. 1H-NMR (400 MHz, DMSO-d6) δ 0,86 (s, 1H), 1,19–1,34 (m, 5H), 1,39–1,47 (m, 2H), 1,56 (s, 1H), 1,68 (d, J = 11,1). Hz, 1H), 1,77-1,86 (m, 4H), 2,19 (s, 3H), 3,47-3,54 (m, 2H), 3,67 (s, 2H), 4 ,50 (s, 1H), 5,54 (s, 1H), 6,61 (d, J = 7,4 Hz, 1H), 7,09 (d, J = 9,0 Hz, 1H), 7 ,48 (d, J = 9,1 Hz, 3H), 7,79 (d, J = 13,9 Hz, 1H), 7,95 (t, 1H), 8,10 (s, 1H), 8 ,70 (s, 1H), 10,29 (s, 1H), 11,79 (s, 1H), 12,56 (s, 1H) BP 5-(1-Oxa-7-Azaspiro[3.5]Nonan-7-yl)pyridin-2-amine PB1 I-326 (R)-N-(4- (4-((5-(3-amino-pyrrolidin-1- yl)pyridin-2- yl)amino)- 5-oxo-5,6-di-hydro-1, 6- Naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: C3, m/z 542.73 [M+H]+, Ret. tempo = 1.36 min. RMN 1H (400 MHz, DMSO-d6) δ 1,16–1,31 (m, 4H), 1,37–1,45 (m, 3H), 1,64–1,83 (m, 6H), 2,05 (m, 1H), 2,35 (t, 1H). ), 2,90 (t, 1H), 3,27 (t, 1H), 3,41 (t, 1H), 3,57 (d, 1H), 6,53 (d, J = 6,9 Hz , 1H), 7,08 (s, 2H), 7,37 (d, 1H), 7,43 (d, J = 7,3 Hz, 1H), 7,71 (t, 2H), 7,98 (t, 1H), 8,49 (s, 1H), 11,22 (s, 1H), 12,18 (s, 1H) PA PA16 PB1 I-327 Synthesis of N-(3- Fluor-4-(5-oxo-4- ((5-(piperidin- 3-yl)pyridin- 2-yl)amino)-5,6-di-hydro- 1,6- naftiridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 541.7 [M+H]+, Ret. tempo = 1.36 min. RMN 1H (400 MHz, DMSO-d6) δ 1,26–1,32 (m, 3H), 1,40–1,45 (m, 2H), 1,70–1,94 (m, 7H), 2,92–2,97 (m, 2H), 3,12 (d , J = 11,4 Hz, 1H), 3,38 (m, 5H verschmolzen mit Feuchtigkeit), 6,70 (s, 1H), 7,23 (s, 1H), 7,48-7,60 (m, 2H), 7,84-7,98 (m, 2H), 8,40 (s, 1H), 8,47 (d, 1H), 8,80 (d, J = 11,1 Hz, 1H), 9,05 (s, 1H), 10,33 (s, 1H), 11,93 (s, 1H), 13,00 ( s, 1H) BP PA17 PB1 I-328 (R)-N-(3-Fluoro-4-(4-((5-(3-hydroxy-pyrrolidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro- 1,6-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide Method: C3, m/z = 543.90 [M+H]+, Ret. time = 1.515 min. RMN 1H (400 MHz, DMSO-d6) δ 1,19–1,47 (m, 7H), 1,69 (d, 1H), 1,76–1,93 (m, 6H), 2,06 ( m, 1H), 2,37 (m, 1H), 2,12 (d, 1H), 3,46 (m, 1H), 4,43 (s, 1H), 5,00 (s, 1H), 6,56 (d, J = 7,5 Hz, 1H), 7,06 (s, 2H), 7,45 (m , 2H), 7,75 (d, J = 13,0 Hz, 2H), 7,98 (s, 1H), 8,52 (s, 1H), 10,23 (s, 1H), 11,61 (s, 1H), 12,21 (s, 1H) BP (R)-1-(6-aminopyridin-3-yl)pyrrolidine-3-ol PB1 I-329 (S)-N-(4- (4-((5-(3-amino-pyrrolidin-1- yl)pyridin-2- yl)amino)-5-oxo-5,6-dihydro-1,6- Naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: C3, m/z = 542.83 [M+H]+, Ret. tempo = 1.387 min. 1 H RMN (400 MHz, DMSO-d6) ö 0,86 (s, 1H), 1,24 (s, 4H), 1,29 (d, J = 11,9 Hz, 2H), 1,43 ( d, J = 11,9 Hz, 2H), 1,67 (s, 1H), 1,82-1,92 (m, 6H), 2,08 (s, 1H), 3,16 (s, 1H) ), 3,54 (s, 2H), 6,55 (d, J = 7,2 Hz, 1H), 7,04 (s, 1H), 7,39 (d, 1H), 7,45 (d , 1H), 7,71–7,79 (m, 3H), 8,00 (t, J = 8,8 Hz, 1H), 8,52 (d, J = 10,3 Hz, 1H). ), 10,22 (s, 1H), 12,20 (d, J = 11,2 Hz, 1H) BP(S)-(1-(6-amino-pyridin-3-yl)pyrrolidin-3-yl)carbamate of tert-butyl PB1 I-330 (S)-N-(3-Fluor-4-(4-((5-(3-(methyl-amino)piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6 - Dihydro-1,6-naftiridin-2-yl)phenyl)cyclohexane-carboxamide Method: C3, m/z = 270.37 [M+H]+, Ret. tempo = 1.381 min. RMN 1H (400 MHz, DMSO-d6) δ 1,10–1,29 (m, 4H), 1,37–1,66 (m, 2H), 1,54 (s, 2H), 1,75 ( s, 1H), 1,81–1,88 (m, 6H). ), 2,33 (s, 4H), 2,69 (d, J = 11,0 Hz, 2H), 3,51 (d, J = 12,4 Hz, 2H), 3,66 (d, J = 10,5 H,z 1H), 6,56 (d, J = 7,3 Hz, 1H), 7,01 (d, J = 8,9 Hz, 2H), 7,38–7,45 ( m, 3H), 7,75 (d, J = 13,9 Hz, 1H), 7,97–8,04 (m, 2H), 8,73 (s, 1H), 10,24 (s, 1H ), 12,39 (s, 1H) BP PA18 PB1 I-331 (S)-N-(3-Fluor-4-(4-((5-(3-(hidroxymethyl)pyrrolidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6 - Dihidro -1,6-nafthyridin-2-yl)phenyl)cyclohexane-carboxamida Method: C3, m/z = 577.78 [M+H]+, Ret. time = 1.519 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,12-1,22 (m, 2H), 1,28-1,34 (m, 2H), 1,44 (q, J = 12,3 Hz, 2H), 1,59-1, 86 (m, 4H), 2,02-2,08 (m, 1H), 2,34–2,43 (m, 2H), 3,04 (dd, J = 9,3, 6,1 Hz, 2H), 3,16–3,32 (m, 3H), 3,27–3,52 (m, 2H), 4,76 (s, 1H), 6,32 ( d, J = 6,0 Hz , 1H), 6,87 (d, J = 8,8 Hz, 1H), 7,05 (dd, J = 8,8, 3,1 Hz, 1H), 7,41 (dd, J = 8, 6, 2,0 Hz, 1H), 7,61 (d, J = 6,0 Hz, 1H), 7,65-7,75 (m, 2H), 7,90 (m, 1H), 8, 34 (s, 2H), 10,18 (s, 1H), 14,64 (s, 1H) PA PA19 PB1 I-332 (R)-N-(3-Fluor-4-(4-((5-(3-(methyl-amino)piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6 - Dihydro-1,6-naftiridin-2-yl)phenyl)cyclohexane-carboxamide Method: C3, m/z = 570.8 [M+H]+, Ret. Time = 1.354 min Chiral HPLC: Method A3 Ret. time: 5.5 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,21–1,29 (m, 4H), 1,40–1,43 (m, 2H), 1,54–1,57 (m, 2H), 1,64–1,75 (m , 2H), 1,81–1,90 (m, 5H), 2,36–2,71 (m, 5H), 3,34-3,43 (m, 2H), 3,66 (d, J = 11,6 Hz, 2H), 6,56 (d, J = 7,3 Hz, 1H), 7,02 (d, J = 8,9 Hz, 1H), 7,39–7,45 (m , 3H), 7,75 (d, J = 14,1 Hz, 1H), 7,97–8,06 (m, 3H), 8,74 (s, 1H), 10,23 (s, 1H) , 12,39 (s, 1H). ) BP PA20 PB1 I-333 (S)-N-(3-Fluoro-4-(4-((5-(3-hydroxy-pyrrolidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro- 6H-naphthyridin-2-yl)phenyl)cyclohexanecarboxamide Method: H, m/z = 543.40 [M + H]+, Ret. time = 3,668 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,90–1,34 (m, 3H), 1,39–1,48 (m, 2H), 1,66–1,69 (d, J = 10,4 Hz, 1H), 1, 72–1,93 (m, 6H), 2,02–2,08 (m, 1H), 2,34–2,40 (m, 2H), 3,11–3,13 (m, 1H), 3,46 (dd, J = 10,1, 4,9 Hz, 1H), 4,43 (s, 1H), 4,98–5,04 (m, 1H), 6,55 (d, J = 7,3 Hz, 1H), 7,42 (dd, J = 25,0, 7,9 Hz, 2H), 7,75 (d, J = 13,3 Hz, 2H), 8,00 (m, 1H), 8,53 (s, 1H). ), 10,23 (s, 1H), 11,58 (s, 1H), 12,21 (s, 1H). BP (S)-1-(6-amino-pyridin-3-yl)pyrrolidin-3-ol PB1 I-334 (S)-N-(4-(4-((5-(3-Amino-piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6- naftiridine - 2-yl)-3-fluorophenyl)cyclohexan)carboxamida Method: C3, m/z = 556.78 [M+H]+, Ret. tempo = 1.372 min. RMN 1H (400 MHz, DMSO-d6) δ 0,75–0,78 (m, 3H), 1,72 (s, 3H), 1,89 (dt, J = 11,8, 7,3 Hz, 6H), 2,33–2,41 (m, 5H), 2,62-2,66 (m, 3H), 2,78 (s, 2H), 6,57 (d, J = 7,3 Hz , 1H), 7,02 (d, J = 8,9 Hz, 1H), 7,39-7,45 (m, 2H), 7,55 (d, J = 8,5 Hz, 1H), 7 ,81 (d, J = 14,1 Hz, 1H), 7,97-8,04 (m, 2H), 8,74 (s, 1H), 9,74 (s, 1H), 12,38 ( s, 1H) BP PA23 PB4 I-335 N-(3- Fluoro-4-(5-oxo-4-((5-(pyrrolidin- 3-yl)pyridin- 2-yl)amino)-5,6-dihydro-1,6-naphthyridin-yl) . Phenyl) cyclohexane-carboxamide Method: C3, m/z = 527.3 [M+H]+, Ret. tempo = 1.359 min. RMN 1H (400 MHz, DMSO-d6) δ 1,23–1,29 (m, 5H), 1,40–1,43 (m, 3H), 1,67–1,75 (m, 2H), 1,82–1,84 (m, 5H), 2,14–2,45 (m, 1H), 3,13-3,16 (m, 2H), 3,50-3,67 (m, 1H ), 6,57 (d, J = 7,1 Hz, 1H), 7,02 (d, J = 8,5 Hz, 1H), 7,46 ( s, 2H), 7,69 (d, J = 14,3 Hz, 1H), 7,97-7,99 (m, 1H), 8,27 (s, 1H), 8,99 (s, 1H), 10,32 (s, 1H), 12 ,95 (segundos, 1H) BP tert-butyl-3-(6-amino-pyridin-3-yl)pyrrolidine-1-carboxylate PB1 I-336 N-(3- Fluor-4- (4-((5-(3- isopropyl-pyrrolidin-1- yl)pyridin-2- yl)amino)- 5-oxo-5,6-di-hydro-1, 6- Naftiridin-2-yl)phenyl)cyclohexane-carboxamida Method: C3, m/z = 569.49 [M+H]+, Ret. time = 2.060 min. RMN 1H (400 MHz, DMSO-d6) δ 0,97 (d, J = 6,6 Hz, 6H), 1,22–1,34 (m, 3H), 1,39–1,47 (m, 2H), 1,53–1,69 (m, 3H), 1,85 (d, J = 12,9 Hz, 4H), 1,91-2,03 (m, 1H), 2,09-2 ,17 (m, 1H), 2,38 (dd, J = 13,4, 9,7 Hz, 1H), 2,93 (t, J = 9,3 Hz, 1H), 3,16–3, 32 (m, 1H), 3,45 (m, 2H), 6,62 (d, J = 7,2 Hz, 1H), 7,09 (s, 2H), 7,45–7,56 (m , 2H), 7,74–7,84 (m, 2H), 7,92 (t, J = 8,7). BP PA24 PB1 Hz, 1H), 8,52 (s, 1H), 10h30 (seg, 1h), 11h91 (seg, 1H), 12,54 (s, 1H) I-337 N-(3- Fluor-4- (4-((5-(1- methyl-piperidin-3- yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hidro-1, 6- Naftiridin-2-yl)phenyl)cyclohexane-carboxamida Method: C3, m/z = 555.8 [M+H]+, Ret. time = 1.435 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,25-1,30 (m, 5H), 1,39-1,51 (m, 3H), 1,69-1,77 (m, 2H), 1,78-1,86 (m , 6H), 2,40 (s, 3H), 2,87-3,01 (s , 3H), 6,62 (d, J = 7,2 Hz, 1H), 7,08 (d, J = 8,5 Hz, 1H), 7,40-7,50 (m, 2H), 7,71-7,81 (m, 2H), 8,01-8,05 (m, 1H), 8,32 (s, 1H), 9,06 (s, 1H), 10,26 (s, 1H), 11,71 (d, J = 6,0 Hz, 1H), 12,69 (s, 1H) DPPA22 PB1 I-338 N-(3- Fluor-4- (4-((5-(3- Fluor-piperidin-1- yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hydro-1, 6- Naftiridin-2-yl)phenyl)cyclohexane-carboxamida Method: C3, m/z 559.8 [M+H]+, Ret. time = 1.731 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,18–1,32 (m, 3H), 1,38–1,46 (m, 2H), 1,62–1,67 (m, 2H), 1,75–1,85 (m , 6H), 1,90–1,93 (m, 1H), 2,33–2,37 (m, 1H), 3,16–3,17 (m, 3H), 3,39–3,45 (m, 1H), 4,75–4,87 (m, 1H), 6,56 (d, J = 7,2 Hz, 1H), 7,02 (d, J = 8,9 Hz, 1H) , 7,38– 7,51 (m, 3H), 7,75 (dd, J = 14,3, 2,0 Hz, 1H), 7,97– 8,01 (m, 1H), 8,08 (d, J = 3,1 Hz, 1H), 8,75 (s, 1H), 10,22 (s, 1H), 11,62 (d, J = 5,7 Hz, 1H), 12,41 (s, 1H) PA PA21 PB1 I-339 N-(3- Fluor-4-(5-oxo-4-((5-(piperidin-4- yl)pyridin- 2-yl)amino)- 5,6-dihydro- 1,6- naphthyridin- 2- yl)phenyl)cyclohexanocarboxamide Method: H, m/z = 539.35 [M + H]+, Ret. time = 3.365 min. 1H NMR (400 MHz, DMSO-d6) δ 1.12–1.29 (m, 3H), 1.37–1.54 (m, 4H), 1.67–1.84 (m, 7H), 2.38 (d, J=11.7Hz, 2H), 2.58 (d, J=12.1Hz, 2H), 3.02 (d, J=12.0Hz, 2H), 6, 59 (d, J=7.2Hz, 1H), 7.03 (d, J=8.4Hz, 1H), 7.44 (dd, J=13.5, 7.9Hz, 2H), 7.66 (d, J=8.5Hz, 1H), 7.77 (d, J=14.2Hz, 1H), 8.00 (t, J=8.8Hz, 1H), 8, 25 (s, 1H), 9.01 (s, 1H), 10.25 (s, 1H), 12.65 (s, 1H) BP terc.-butyl-4-(6-amino-pyridin-3-yl)piperidine-1-carboxylate PB1 I-340 N-(4-(4- ((5-((2S,6R)-2,6- Dimethyl-piperazin-1- yl)pyridin-2- yl)amino)-5-oxo-5,6- dihydro- 1,6-naphthyridin-2-yl)-3-fluorophenyl)cyclo-hexane-carboxamide Method: H, m/z = 568.40 [M + H]+, Ret. time = 3.583 min. 1H NMR (400 MHz, DMSO-d6) δ 0.75 (d, J=6.0 Hz, 5H), 1.17–1.29 (m, 4H), 1.37–1.43 (m, 2H), 1.66 (d, J=11.9Hz, 1H), 1.75-1.84 (m, 3H), 2.72 (d, J=11.0Hz, 2H), 3, 16 ( d, J=4.8Hz, 4H), 4.09-4.17 (m, 2H), 6.61 (d, J=7.2Hz, 1H), 7.13 (d, J=8, 7Hz, 1H), 7.45 (d, J=8.4Hz, 2H), 7.59–7.67 (m, 1H), 7.80 (d, J=14.0Hz, 1H) . ), 8.01 (t, J=8.8Hz, 1H), 8.09 (s, 1H), 9.03 PA PA27 PB1 (s, 1H), 10,28 (s, 1H), 11,73 (s, 1H), 12,77 (s, 1H) I-341 (R)-N-(3-Fluor-4-(4-((5-(3-(hidroximethyl)pyrrolidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6 - Dihidro -1,6-nafthyridin-2-yl)phenyl)cyclohexane-carboxamide Method: C3, m/z = 557.6 [M+H]+, Ret. tempo = 1.566 min. RMN 1H (400 MHz, DMSO-d6) δ 1,17–1,32 (m, 3H), 1,37–1,45 (m, 2H), 1,67–1,84 (m, 6H), 2,04 (m, 1H), 2,38 (d, J = 11,7 Hz, 3H), 3,02 (d, J = 12,0 Hz, 1H), 3,16 (m, 2H), 3,04 (m, 2H), 4,74 (s, 1H), 6,59 (d, J = 7,2 Hz, 1H). ), 7,03 (d, J = 8,4 Hz, 2H), 7,44 (dd, J = 13,5, 7,9 Hz, 2H), 7,66 (d, J = 8,5 Hz , 1H), 7,77 (d, J = 14,2 Hz, 1H). ), 8,00 (t, J = 8,8 Hz, 1H), 8,51 (s, 1H), PA PA26 PB1 10,22 (s, 1H), 11,58 (s, 1H), 12,23 (s, 1H) I-342 (R)-N-(4-(4-((5-(3-amino-piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-di-hydro-1,1 6- Naphthyridin-2-yl)-3-fluorophenyl)-1-ethyl-cyclobutane-1-carboxamide Method: C3, m/z = 556.78 [M+H]+, Ret. time = 1.399 min. RMN 1H (400 MHz, DMSO-d6) δ 0,77 (t, J = 7,3 Hz, 3H), 1,13 (d, J = 12,3 Hz, 2H), 1,56 (s, 2H ), 1,85–1,92 (m, 6H), 2,76 (s, 2H), 3,16 (m, 1H), 3,55 (dd, J = 30,8, 11,8 Hz, 3H), 6,57 (d, J = 7,2 Hz, 1H), 7,02 (d, J = 8,9 Hz, 1H), 7,42 (dd, J = 16,5, 7,6 Hz, 2H), 7,55 (d, J = 9,0 Hz, 1H), 7,81 (d, J = 14,7 Hz, 1H), 7,94-8,07 (m, 2H), 8,74 (s, 1H), 9,74 (s, 1H), 12,38 (s, 1H) BP PA25 PB4 I-343 N-(3- Fluor-4- (4-((5-(1- methyl-2- oxo-pyrrolidin-3- yl)pyridin- 2-yl)amino)- 5-oxo-5,6- dihydro- 1,6-nafthyridin-2-yl)phenyl)cyclohexancarboxamida Method: H, m/z = 555.40 [M + H]+, Ret. time = 3.749 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,19-1,34 (m, 3H), 1,37-1,43 (m, 2H), 1,56 (s, 1H), 1,68 (d, J = 11,1 Hz, 1H), 1,82 (dd, J = 25,9, 12,1 Hz, 4H), 2,07 (dq, J = 12,7, 8,7 Hz, 1H), 2,35–2,50 (m, 1H), 2,82 (s, 3H), 3,38–3,51 (m, 2H), 3,69 (t, J = 9,2 Hz). , 1H), 6,63 (d, J = 7,2 Hz, 1H), 7,09 (d, J = 8,5 Hz, 1H), 7,41-7,52 (m, 2H), 7 ,67 (dd, J = 8,4, 2,5 Hz, 1H), 7,79 (dd, J = BP PA28 PB1 14,3, 2,1 Hz, 1H), 8,03 (d, J = 2,4 Hz, 1H), 9,07 (s, 1H), 10,26 (s, 1H), 11,74 (s, 1H), 12,73 (s, 1H) I-344 (R)-N-(4-(4-((5-(3-(Dimethylamino)piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6 -Naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: J, m/z = 584.4 [M + H]+, Ret. Tempo = 3.710 min. RMN 1H (400 MHz, DMSO-d6) δ 1,18–1,37 (m, 3H), 1,43 (q, J = 12,0 Hz, 2H), 1,56–1,59 (m, 1H), 1,67 (d, J = 10,8 Hz, 1H), 1,82–1,92 (m, 6H), 2,26 (s, 6H), 2,37 (d, J = 9 ,0 Hz, 3H), 2,55–2,66 (m, 2H), 3,61 (d, J = 12,2 Hz, 1H). ), 3,72 (d, J = 12,0 Hz, 1H), 6,58 (dd, J = 7,3, 1,8 Hz, 1H), 7,02 (d, J = 8,9 Hz , 1H), 7,38–7,53 (m, 3H), 7,77 (d, J = 14,2 Hz, 1H), 8,00 (t, J = 8,8 Hz, 1H), 8 ,08 (d, J = 2,8 PA PA29 PB1 Hz, 1H), 8,74 (s, 1H), 10,27 (s, 1H), 11,67 (s, 1H), 12,40 (s, 1H) I-345 N-(3- Fluor-4- (4-((5-(4- hidroxy-1- methyl-piperidin-4- yl)pyridin- 2-yl)amino)- 5-oxo-5,6- dihidro- 1,6-nafthyridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 571.6 [M+H]+, Ret. tempo = 1.458 min. RMN 1H (400 MHz, DMSO-d6) δ 1,20–1,35 (m, 3H), 1,45 (q, J = 12,3, 11,5 Hz, 2H), 1,64–1,677 ( m, 2H), 1,78–1,87 (m, 4H ), 1,98 (dt, J = 12,7, 6,5 Hz, 2H), 2,23 (s, 3H), 2,38 (t, J = 11,5 Hz, 3H), 2,56-2,69 (m, 3H), 4,96 (s, 1H), 6,63 (d, J = 7,3 Hz, 1H) , 7,07 (d, J = 8,6 Hz, 1H), 7,41–7,53 (m, 2H), 7,79 (dd, J = 14,2, 2,1 Hz, 1H), 7,88 (dd, J = 8,6, 2,6 Hz, 1H), 8,04 (t, J = 8,8 Hz, 1H), 8,49 (d, J = 2,5 Hz, DP 4-(6-amino-pyridin-3-yl)-1-methyl-piperidin-4-ol PB1 1H), 9,07 (s, 1H), 10,26 (s, 1H), 11,71 (s, 1H), 12,70 (s, 1H) I-346 (S)-N-(4-(4-((5-(3-(Dimethylamino)piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6 -Naftiridin-2-yl)-3-fluorphenyl)cyclohexancarboxamida Method: C3, m/z = 584.9 [M+H]+, Ret. tempo = 1,400 min. 1RMN-H (400 MHz, DMSO-d6) δ0 1,19-1,34 (m, 3H), 1,44 (q, J = 11,6 Hz, 2H), 1,56-1,59 (m, 1H) 1,68 (d, J = 11,2 Hz, 1H), 1,77-1,92 (m, 6H ), 2,26 (s, 6H), 2,32–2,43 (m, 3H), 2,53–2, 66 (m, 2H), 3,61 (d, J = 12,3 Hz, 1H), 3,68–3,77 (m, 1H), 6,58 (dd, J = 7,3, 1, 8 Hz, 1H), 7,03 (d, J = 8,9 Hz, 1H), 7,38–7,53 (m, 3H), 7,77 (d, J = 14,2 Hz, 1H) , 8,00 (t, J = 8,8 Hz, 1H), 8,08 (d, J = 3,0 Hz, 1H), BP PA30 PB1 8,75 (s, 1H), 10,27 (s, 1H), 11,67 (s, 1H), 12,41 (s, 1H). I-347 N-(3- Fluor-4- (4-((5-(1- methyl-pyrrolidin-3- yl)pyridin- 2-yl)amino)- 5-oxo-5,6-di-hydro-1, 6- Naftiridin-2-yl)phenyl)cyclohexane-carboxamida Method: C3, m/z = 541.4 [M+H]+, Ret. Tiempo = 1.384 min.. 1RMN-H (DMSO-d6, 400 MHz): 12,66 (s, 1H), 11,70 (s, 1H), 10,26 (s, 1H), 9,05 (s, 1H), 8,29 (s, 1H), 8,03 (t, J = 8,8, 1H), 7,80-7,74 (m, 2H), 7,48 (d, J = 14,4 Hz, 1H), 7,46 (d, J = 8,0 Hz, 1H), 7,06 (d, J = 8,4 Hz, 1H), 6,62 (d, J = 6,8 Hz, 1H), 2,85–2,8 ( m, 1H), 2,69–2,65 (m, 2H), 2,46–2,42 (m, 2H), 2,38–2,34 (m, 2H), 2,31 (s, 3H), 1,86-1,78 (s, 4H), 1,69 (br s, DP 5-(1-Methylpyrrolidin-3-yl)pyridin-2-amine PB1 1H), 1,45-1,39 (M, 2H), 1,31-1,25 (m, 4H). I-348 N-(3-Fluor-4-(5-oxo-4-((5,6,7,8-tetrahydro-1,6-naphthyridin-2-yl)amino)-5,6-dihydro-2-yl )phenyl)cyclohexanecarboxamide Method: H, m/z = 513.55 [M + H]+, Ret. time = 3.603 min. 1RMN-H (DMSO-d6, 400 MHz): 12,63 (s, 1H), 11,73 (s, 1H), 10,27 (s, 1H), 9,32 (s, 1H), 8,3 (s, 1H), 8,11 (t, J = 8,8, 1H), 7,86 (d, J = 14,4 Hz, 1H), 7,57 (d, J = 8,0 Hz, 1H), 7,43 –7,38 (m, 2H), 6,89 (d, J = 8,4 Hz, 1H), 6,62 (d, J = 6,8 Hz, 1H), 3,96 (br s, 1H ), 3,2 (s, 2H), 2,87 (s, 2H), 2,37 (m, 1H), 1,86-1,77 (m, 4H), 1,69- BP tert.-butyl-2-amino-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate PB1 1,66 (m, 1H), 1,45-1,42 (m, 2H), 1.34-1.24 (m, 4H). I-349 4-((5-(4-Hydroxypiperidin-1-yl)pyridin-2-yl)amino)-2-(2-methyl-4-(oxazol-2-yl)phenyl)-1,6-naphthyridin-5 (6H)-ein Method: C3 m/z = 495.47 [M+H]+, Ret. tempo = 1.321 min. 1RMN-H (DMSO-d6, 400 MHz): 12,49 (s, 1H), 11,67 (s, 1H), 8,5 (s, 1H), 8,28 (s, 1H), 8,06-8,05 (d , J = 4,8 Hz, 1H), 7,96 (s , 1H), 7,93–7,91 (d, J = 4,0 Hz, 1H), 7,6 (d, J = 4, 0 Hz, 1H), 7,48–7,42 (m, 3h), 7,01 c(d, J = 8,0 Hz, 1H). ), 6,57 (d, J = 7,8 Hz, 1H), 3,61 (br s, 1H), 3,51-3,48 (m, 1H), 2,84 (s, J = 10 Hz, 1H), 2,46 (s, 3H), 1,83–1,80 (brd, 2H), 1,51–1,47 (m, 2H). CP 1-(6-amino-pyridin-3-yl)piperidin-4-ol 2-(3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2 ).-yl)phenyl)oxazole I-350 4-(3-(4-((5-(4- hidroxy- piperidin-1- yl)pyridin-2- yl)amino)- 5-oxo-5,6-di-hidro-1,6- naphthyridine- 2- the) phenyl) morpholin-3-on Method: C3, m/z = 513.67 [M+H]+, Ret. Time = 1,290 minutes. 1RMN-H (DMSO-d6, 400 MHz): 12,46 (s, 1H), 11,67 (s, 1H), 8,84 (s, 1H), 8,15–8,12 (m, 2H), 7,92 (d , J = 7,8 Hz, 1H), 7,61–7,57 (m, 1H), 7,54-7,48 (m, 1H), 7,43 (d, J = 7,2 Hz, 1H), 7,05 (d, J = 8,8 Hz, 1H), 6,62 (d, J = 7,2 Hz, 1H), 4,72 (d, J = 3,2 Hz, 1H) , 4,27 (s, 2H), 4,39 (t, J = 4,4 Hz, 2H), 3,83 (t, J = 4,4 Hz, 2H), 3,66–3,65 ( m, 1H), 3,56-3,53 (m, 2H), 2,89 CP 1-(6-amino-pyridin-3-yl)piperidin-4-ol PB2 (t, J = 10Hz, 2H), 1,83- 1.80 (per d, 2H), 1.51–1.47 (m, 2H). I-351 N-(3- Fluor-4- (4-((5-(oxetan-3- yl)pyridin-2- yl)amino)- 5-oxo-5,6-di-hydro-1,6- naphthyridine- 2-yl)phenyl)cyclohexanocarboxamide Method: C3, m/z = 514.57 [M+H]+, Ret. time = 1.539 min. 1RMN-H (DMSO-d6, 400 MHz): 12,73 (s, 1H), 11,73 (s, 1H), 10,25 (s, 1H), 9,015 (s, 1H), 8,37 (s, 1H), 8, 02 (t, J = 8,4 Hz, 1H). ), 7,97–7,94 (m, 1H), 7,80–7,76 (m, 1H), 7,72–7,70 (m, 1H), 7,47–7,36 (m , 2H), 7,14 (d, J = 8,4 Hz, 1H), 6,62 (d, J = 7,2 Hz, 1H), 4,97–4,93 ​​​​​​(m, 2H) , 4,65 (t, J = 6,4 Hz, 2H), 4,33–4,25 (m, 1H), 1,86–1,77 (m, 4H) , 1,66 (s , 1H ), 1,45-1,38 (m, 2H), 1,34-1,24 (m, 4H) BP PA31 PB1 I-352 N-(3- Fluor-4-(4-((5-(1- methyl-2- oxopiperidin- 4-yl)pyridin- 2-yl)amino)-5-oxo-5,6-di-hydro- 1, 6-nafthyridin-2-yl)phenyl)cyclohexancarboxamida Method: C3, m/z = 567.61 [M+H]+, Ret. time = 1.455 min. 1RMN-H (DMSO-d6, 400 MHz): 12,69 (s, 1H), 11,71 (s, 1H), 10,25 (s, 1H), 9,015 (s, 1H), 8,31 (s, 1H), 8, 02 (t, J = 8,4 Hz, 1H). ), 7,79-7,37 (m, 2H), 7,48-7,43 (m, 2H), 7,07 (d, J = 8,4 Hz, 1H), 6,61 (d, J = 7,2 Hz, 1H), 3,38 3,3 (m, 2H), 3,08 (s, 1H), 2,83 (s, 3H), 2,37-2,33 (m, 3H), 2,01 (s, 2H), 1,86-1,77 (m, 4H), 1,66 (s, 1H), 1,45- 1,38 (m, 2H), 1,34 -1,24 (metro, 4H) BP PA32 PB1 I-353 N-(4-(4-((5-(Cyanomethyl)yl)pyridin-2-yl)amino)-5-oxo-5,6-dihydro-1,6-naphthyridin-2-yl)-3 - Fluorphenyl )cyclohexancarboxamide Method: C3, m/z = 597.39 [M+H]+, Ret. Time = 1,580 minutes. 1RMN-H (DMSO-d6, 400 MHz): 13,26 (s, 1H), 12,41 (s, 1H), 10,40 (s, 1H), 8,96 (s, 1H), 8,43 (s, 1H), 7,89–7,85 (m, 3H), 7,47 (s, 1H), 7,55 (d, J = 8,0 Hz, 1H), 7,34 (s, 1H), 6,77 (d, J = 72 Hz, 1H), 4,21 (s, 2H), 2,42–2,36 (m, 1H), 1,87–1,77 (m, 4H), 1,69– 1,67 (m, 1H), 1,48–1,39 (m, 2H), 1,34–1,25 (m, 4H). EP 2-(6-Brompiridin-3-il)acetonitrilo PB1 I-354 N-(3-Fluoro-4-(4-((6-methyl-5,6,7,8-tetrahidro-1,6-nafthyridin-2-yl)amino)-5-oxo-5,6-dihidro -1,6-naphthyridine-2-yl)phenyl)cyclohexanocarboxamida Method: C3 02, m/z = 527.5 [M+H]+, Ret. tempo = 1.421 min. 1RMN-H (DMSO-d6, 400 MHz): 12,72 (s, 1H), 11,73 (s, 1H), 10,26 (s, 1H), 9,32 (s, 1H), 8,11 (t, J = 8 ,8, 1H), 7,87 (d, J = 14,4 Hz, 1H), 7,57 (d, J = 8,0 Hz, 1H), 7,43-7,38 (m, 2H) , 6,95 (d, J = 8,4 Hz, 1H), 6,61 (d, J = 6,8 Hz, 1H), 4,06 (br s, 1H), 3,06 (s, 2H ), 2,66 (s, 3H), 1,90–1,65 (m, 6H), 1,43–1,4 (m, 2H), 1,29–1,73 (m, 2H). DP 6- methyl- 5,6,7,8- tetrahydro- 1,6- naphthyridin-2- amine PB1 I-355 6-Amino-2-fluor-3-(4-((5-(4-hidroxy-piperidin-1-yl)pyridin-2-yl)amino)-5-oxo-5,6-di-hidro-1 ,6 - Naftiridin-2-yl)-N,N-dimethyl-benzamide Method: C3 04, m/z = 518.44 [M+H]+, Ret. time = 1.333 min. 1H-NMR (400 MHz, DMSO-d6) δ 12,25 (s, 1H), 11,33 (s, 1H), 8,68 (s, 1H), 7,98 (d, J = 2,4 Hz, 1H), 7,7 (s, 1H), 7,6 (d, J = 8,0 Hz, 1H), 7,45–7,41 (m, 2H), 7,25 (t, J = 6,4 Hz, 1H), 7,00–6,93 (m, 2H), 6,23 (d, J = 6,8). Hz, 1H), 4,70 (d, 1H), 4,52 (t, J = 5,2 Hz, 1H), 3,62–3,61 (m, 1H), 3,49–3,42 (m, 4H), 2,9 (s, 3H), 2,88 (s, 3H), 2,83–2,80 (m, 1H), 1,84–1,81 (m, 2H), 1,50–1,48 (m, 2H). EP 1-(6-amino-pyridin-3-yl)piperidin-4-ol PB3 I-356 (R)-8-((6-((Dimethylamino)methyl)-5-(THF-3-yl)pyridin- 2-yl)amino)- 5-(7-fluorimidazo[1,2-a]pyridin- 3-yl)isoquinoline-1(2H)-on LCMS method J m/z = 500.2 [M+H]+, Ret. tempo = 2.57 min Chiral HPLC method A1: Ret. time = 6.63 RMN 1H (400 MHz, DMSO-d6) δ 12,81 (s, 1H), 11,61 (d, J = 6,0 Hz, 1H), 8,89 (d, J = 8,7 Hz, 1H), 7,93 (dd, J = 7,5, 5,8 Hz, 1H), 7,77–7,63 (m, 3H), 7,55 (dd, J = 10,2, 2, 6 Hz, 1H), 7,15 (dd, J = 7,3, 5,8 Hz, 1H), 7,01–6,88 (m, 2H), 5,89 (dd, J = 7,3 , 1,3 Hz, 1H), 4,05–3,93 (m, 2H), 3,81 (q, J = 7,7 Hz, 2H), 3,81 (q, J = 8,2, 6,8 Hz, 1H), 2,36-2,18 (m, 6H), 1,90 (dq, J = 12,1, 7,7 Hz, 1H). FP PA35 3-Bromo-7- Fluorimidazo [1,2-a] Pyridine I-357 (S)-8-((6-((Dimethylamino)methyl)-5-(THF-3-yl)pyridin-2-yl)amino) 5-(7- fluorimidazo[1,2-a]pyridin-3 -yl)isoquinoline-1(2H)-on LCMS method J m/z = 500.2 [M+H]+, Ret. tempo = 2.56 min Chiral HPLC Method A1: Ret. time = 6.71 1H-NMR (400 MHz, DMSO-d6) δ 12,81 (s, 1H), 11,63 (d, J = 5,8 Hz, 1H), 8,80 (s, 1H), 7,93 ( dd, J = 7,6, 5,8 Hz, 1H), 7,77-7,63 (m, 3H), 7,56 (dd, J = 10,1, 2,6 Hz, 1H), 7 ,23-7,11 (m, 1H), 7,02 (d, J = 8,3 Hz, 1H), 6,93 (td, J = 7,6, 2,6 Hz, 1H), 5, 97–5,81 (m, 1H), 4,15–3,93 (m, 3H), 3,82 (q, J = 7,7 Hz, 2H), 3,73 (s, 1H), 3 ,55 (t, J = 7,6 Hz, 1H), 3,49 (t, J = 5,3 Hz, 1H), 3,42 (t, J = 5,2 Hz, 1H), 2,31 (s, 2H), 1,96-1,86 (m, 1H), 1,25 (d, J = 8,0 Hz, 1H). FP PA35 3-Bromo-7- Fluorimidazo [1,2-a] Pyridine I-358 8-((5-(4-Methyl-piperazin-1-yl)pyridin-2-yl)amino)-5-(pyridin-4-yl)isoquinolin-1(2H)-one LCMS method J m/z = 418.1 [M+H]+, Ret. time = 2.09 minutes RMN 1H (400 MHz, DMSO-d6) δ 12,51 (s, 1H), 8,70–8,54 (m, 3H), 8,00 (d, J = 3,0 Hz, 1H), 7 ,54 (d, J = 8,7 Hz, 1H), 7,50-7,39 (m, 3H), 7,17 (d, J = 7,4 Hz, 1H), 6,96 (d, J = 8,9 Hz, 1H), 6,47 (d, J = 7,5 Hz, 1H), 3,13 (t, J = 5,0 Hz, 4H), 2,5 (s, 4H, em DMSO-Peak eingetaucht), 2,26 (s, 3H). GP 5-(4-methylpiperazin-1-yl)pyridin-2-aminopyridin-4-borosaure I-359 8-((5-(4-(Methylamino)piperidin-1-yl)pyridin-2-yl)amino)-5-(pyridin-4-yl)isoquinolin-1(2H)-ona By LCMS method J m/z = 438.1 [M+H]+, Ret. time = 2.31 minutes 1H-NMR (400 MHz, DMSO-d6) δ 12,59 (s, 1H), 11,54 (s, 1H), 8,66 (t, J = 6,8 Hz, 3H), 8,35 ( s, 1H), 8,04 (d, J = 3,0 Hz, 1H), 7,55 (d, J = 8,7 Hz, 1H), 7,50-7,38 (m, 3H), 7,18 (d, J = 7,4 Hz, 1H), 6,95 (d, J = 8,9 Hz, 1H), 6,46 (d, J = 7,4 Hz, 1H), 3, 68 (d, J = 4,1 Hz, 2H), 2,85 (td, J = 10,2, 9,7, 5,2 Hz, 1H), 2,77-2,69 (m, 2H) , 2,48 (s, 3H), 2,02 (d, J = 12,0 Hz, 2H), 1,56 (tt, J = 12,1, 6,2 Hz, 2H). GP PA36 pyridine-4-boronic acid I-360 5-(7- Fluorimidazo[1,2-a]pyridin-3-yl)-8-((5-(4-methyl-piperazin-1-yl)pyridin-2-yl)amino)isoquinoline-1 (2H )-eins By LCMS method J m/z = 471.2 [M+H]+, Ret. time = 2.31 minutes 1H-RMN (400 MHz, DMSO-d6) δ 12,59 (s, 1H), 11,56 (s, 1H), 8,75–8,59 (m, 1H), 8,27 (s, 1H ) ), 8,03 (d, J = 3,1 Hz, 1H), 7,97–7,86 (m, 1H), 7,71–7,60 (m, 2H), 7,56 (dd , J = 10,1, 2,8 Hz, 1H), 7,47 (dd, J = 9,0, 3,0 Hz, 1H), 7,14 (dd, J = 7,6, 3,3 Hz , 1H), 7,05–6,85 (m, 2H), 5,88 (d, J = 7,3 Hz, 1H), 3,14 (t, J = 4,9 Hz, 4H), 2 ,48 (d, J = 5,1 Hz, 3H), 2,25 (s, 3H). FP 5-(4-Methyl-piperazin-1-yl)pyridin-2-amine 3-Bromo-7-fluorimidazo[1,2-a]pyridine I-361 8-((5-(4- hidroxy- piperidin-1- yl)pyridin-2- yl)amino)-5-(1-methyl- 1H-pyrrolo [2,3-b] pyridin-4-yl)- 2,6-nafthyridin-1(2H)-on By LCMS method J m/z = 468.4 [M+H]+, Ret. time = 2.57 minutes 1H-RMN (400 MHz, DMSO-d6) δ 11,41 (s, 1H), 9,33 (s, 1H), 8,32–8,07 (m, 2H), 8,04 (d, J = 3,1 Hz, 1H), 7,97–7,77 (m, 2H), 7,51–7,37 (m, 2H), 7,32 (t, J = 6,4 Hz, 1H) , 7,18 (d, J = 7,4 Hz, 1H), 6,92 (d, J = 4,9 Hz, 1H), 6,07 (d, J = 3,5 Hz, 1H), 4 ,70 (s, 1H), 3,84 (s, 3H), 3,71–3,58 (m, 1H), 3,49 (dd, J = 11,2, 6,2 Hz, 2H), 2,93 -2,76 (m, 2H), 1,97-1,75 (m, 2H), 1,51 (q, J = 11,1, 9,7 Hz, 2H). IP 1-(6-amino-pyridin-3-yl)piperidin-4-ol 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl) -1H-pyrrolo[2,3-b]pyridine I-362 5-(1-Methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)-8-((5-morpholinopyridin-2-yl)amino)-2,6-naphthyridin-1(2H) - eins By LCMS method J m/z = 454.3 [M+H]+, Ret. time = 2.39 minutes 1H-NMR (400 MHz, DMSO-d6) δ 11,43 (s, 1H), 9,39 (s, 1H), 8,31–8,16 (m, 1H), 8,06 (d, J = 3,0 Hz, 1H), 7,99 (d, J = 9,1 Hz, 1H), 7,90 (s, 1H), 7,50–7,37 (m, 2H), 7,33 (t, J = 6,5 Hz, 1H), 7,19 (d, J = 7,3 Hz, 1H), 6,93 (d, J = 4,9 Hz, 1H), 6,08 (d , J = 3,5 Hz, 1H), 3,86 (s, 3H), 3,78 (t, J = 4,7 Hz, 3H), 3,13 (t, J = 4,8 Hz, 4H ). JP 5- morpholino- pyridin-2- amine 1- methyl-4-(4,4,5,5- tetramethyl- 1,3,2-dioxa-borolan-2- yl)-1H- pyrrolo [2,3- b] pyridine I-363 8-((6-((Dimethylamino)methyl)-5-(tetrahydro- 2H-pyran-4-yl)pyridin- 2-yl)amino)- 5-(7- fluorimidazo[1,2-a]pyridin- 3-yl)isoquinolin-1(2H)-one By LCMS method J m/z = 513.2 [M+H]+, Ret. time = 2.52 minutes 1H-RMN (400 MHz, DMSO-d6) δ 12,80 (s, 1H), 11,64 (s, 1H), 8,94 (d, J = 8,7 Hz, 1H), 8,41 ( s, 2H), 7,94 (t, J = 6,6 Hz, 1H), 7,78-7,60 (m, 3H), 7,16 (d, J = 7,3 Hz, 1H), 6,94 (d, J = 8,4 Hz, 2H), 5,89 (d, J = 7,3 Hz, 1H), 3,98 (d, J = 11,1 Hz, 3H), 3, 58 (s, 3H), 3,21 (s, 2H), 2,23 (s, 6H), 1,78–1,61 (m, 4H), 1,25 (s, 2H). FP PA37 3-Bromo-7-fluoroimidazo[1,2-a]pyridine I-364 (R)-5-(7-Fluoro-imidazo[1,2-a]pyridin-3-yl)-8-((5-(2-(2-hydroxy-propan-2-yl)morpholino)pyridin- 2-yl)amino)isoquinoline-1(2H)-one LCMS method J m/z = 545.2 [M+H]+, Ret. tempo = 2.89 min Chiral HPLC method A2: Ret. Hour = 19.69 RMN 1H (400 MHz, DMSO-d6) ö 12,61 (d, J = 7,5 Hz, 1H), 11,56 (d, J = 5,6 Hz, 1H), 8,71 (dd , J = 24,1, 8,7 Hz, 1H), 8,05 (d, J = 3,3 Hz, 1H), 7,97–7,86 (m, 1H), 7,77–7, 60 (m, 2H), 7,55 (dd, J = 10,2, 2,8 Hz, 1H), 7,47 (dt, J = 11,3, 5,6 Hz, 1H), 7,14 (t, J = 6,3 Hz, 1H), 7,01 (t, J = 8,2 Hz, 1H), 6,93 (td, J = 7,6, 2,7 Hz, 1H), 5 ,88 (d, J = 7,3 Hz, 1H), 4,49 (s, 1H), 4,07–3,96 (m, 1H), 3,75–3,59 (m, 2H), 3,49 (d, J = 11,7 Hz, 1H), 1,16 (d, J = 22,8 Hz, 6H). FP PA38 3-Bromo-7-fluoroimidazo[1,2-a]pyridine I-365 (S)-5-(7-fluoro-imidazo[1,2-a]pyridin-3-yl)-8-((5-(2-(2-hydroxy-propan-2-yl)morpholino)pyridin- 2-yl)amino)isoquinoline-1(2H)-one LCMS method J m/z = 515.1 [M+H]+, Ret. tempo = 2.87 min Chiral HPLC method A1: Ret. Time = 8.42 1H-RMN (400 MHz, DMSO-d6) δ 12,62 (s, 1H), 11,60 (s, 1H), 8,68 (d, J = 8,7 Hz, 1H), 8,51 ( s, 2H), 8,05 (d, J = 3,0 Hz, 1H), 7,98–7,88 (m, 1H), 7,71–7,60 (m, 2H), 7,51 (ddd, J = 29,1, 9,6, 2,9 Hz, 2H), 7,13 (d, J = 5,7 Hz, 1H). ), 7,00 (d, J = 8,9 Hz, 1H), 6,93 (td, J = 7,5, 2,7 Hz, 1H), 5,88 (d, J = 7,3 Hz , 1H), 4,50 (s, 1H), 4,06–3,95 (m, 1H), 3,71-3,59 (m, 2H), 1,16 (d, J = 22,8 Hz, 6H). FP PA38 3-Bromo-7-fluoroimidazo[1,2-a]pyridine I-366 5-(1-Methyl-1H-imidazol-5-yl)-8-((5-(piperazin-1-yl)pyridin-2-yl)amino)isoquinoline-1(2H)-on By LCMS method J m/z = 402.21 [M+H]+, Ret. time = 2.14 minutes 1H-NMR (400 MHz, DMSO-d6) δ 12,52 (s, 1H), 11,50 (s, 1H), 8,60 (d, J = 8,7 Hz, 1H), 8,27 ( s, 1H), 8,01 (d, J = 3,0 Hz, 1H), 7,76 (s, 1H), 7,51–7,43 (m, 2H), 7,15 (d, J = 5,7 Hz, 1H), 6,93 (d, J = 8,9 Hz, 1H), 6,91 (td, J = 7,5, 2,7 Hz, 1H), 6,14 (d , J = 7,2 Hz, 1H), 3,36 (s, 3H), 3,26 (s, 4H), 3,17 (s, 4H). HP tert-Butyl 4-(6-amino-pyridin-3-yl)piperazine-carboxylate 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl )-1H-imidazole I-367 8-((6-((Dimethylamino)methyl)-5-(4-hidroxy-piperidin-1- yl)pyridin- 2-yl)amino)- 5-(1-methyl-1H-pyrrol[2, 3- b]Pyridin-4-yl)-2,6-naphthyridin-1(2H)-on By LCMS method J m/z = 525.2 [M+H]+, Ret. time = 2.58 minutes 1H-NMR (400 MHz, DMSO-d6) δ 11.43 (d, J=5.9 Hz, 1H), 9.33 (s, 2H), 8.28–8.13 (m, 2H), 7.89 (s, 1H), 7.84 (d, J=8.8Hz, 1H), 7.55 (d, J=8.8Hz, 1H), 7.41 (d, J=3 .5 Hz, 1H), 733 (t, J = 6.6 Hz, 1H), 7.17 (d, J = 7.3 Hz, 1H), 6.92 (d, J = 4.9 Hz, 1H), 6.06 (d, J=3.5Hz, 1H), 3.84 (s, 3H), 2.72 (d, J=10.8Hz, 3H), 2.31 (s, 6H), 1.86 (d, J=13.6Hz, 2H), 1.63–1.52 (m, 2H). IP PA40 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl)-1H-pyrrolo [2,3-b] pyridine I-368 5-(7-fluor-imidazo[1,2-a]pyridin-3-yl)-8-((5-(3-(2-hidroxy-propan-2-yl)piperidin-1-yl)pyridin- 2-yl)amino)isoquinoline-1(2H)-on LCMS method C m/z = 513.8 [M + H]+, Ret. Time = 0.98 minutes RMN 1H (400 MHz, DMSO-d6) ö 12,56 (s, 1H), 11,55 (s, 1H), 8,65 (d, J = 8,7 Hz, 1H), 8,01 (d, J = 3,0 Hz, 1H), 7,91 (dd, J = 7,6, 5,8 Hz, 1H), 7,72–7,58 (m, 2H), 7,55 ( dd, J = 10,1, 2,7 Hz, 1H), 7,43 (dd, J = 8,9, 3,1 Hz, 1H), 7,13 (d, J = 7,3 Hz, 1H) ), 7,02–6,83 (m, 2H), 5,87 (d, J = 7,3 Hz, 1H), 4,28 (s, 1H), 3,76 (d, J = 12, 3Hz, 1H). ), 3,64 (d, J = 11,8 Hz, 1H), 3,17 (d, J = 5,0 Hz, 1H), 1,84 (d, J = 12,8 Hz, 1H), 1,75 (d, J = 13,1 Hz, 1H), 1,60–1,49 (m, 2H), 1,24 (s, 1H), 1,11 (d, J = 10,1 Hz) , 6H). FP PA39 3-Bromo-7-fluoroimidazo[1,2-a]pyridine I-369 8-((6-((Dimetilamino)metil)-5-morpholinopiridin-2-il)amino)-5-(1-metil-1H-pirrolo[2,3-b]piridin-4-il)-2, 6-naftiridin-1(2H)-on By LCMS method J m/z = 510.9 [M+H]+, Ret. time = 2.38 minutes RMN 1H (400 MHz, DMSO-d6) δ 9,40 (s, 1H), 8,24 (d, J = 4,9 Hz, 1H), 7,91–7,74 (m, 2H), 7 ,45 (dd, J = 8,2, 4,9 Hz, 2H), 6,97 (dd, J = 16,2, 6,1 Hz, 2H), 4,51 (s, 1H), 3, 86 (s, 2H), 3,81-3,74 (m, 2H), 2,95 (s, 2H), 2,85 (t, J = 4,5 Hz, 2H). JP PA41 1-Methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolan-2-yl)-1H-pyrrolo[2,3-b]pyridine I-370 8-((6-((Dimethylamino)methyl)-5-(4-hidroxy-4-(methoxymethyl)piperidin-1-yl)pyridin-2-yl)amino)-5-(7-fluor - Imidazo[1 ,2-a]pyridin-3-yl)isoquinoline-1(2H)-on By LCMS method J m/z = 572.2 [M+H]+, Ret. time = 2.55 minutes RMN 1H (400 MHz, DMSO-d6) δ 12,72 (s, 1H), 11,57 (d, J = 5,6 Hz, 1H), 8,86 (d, J = 8,8 Hz, 1H), 7,92 (t, J = 6,8 Hz). , 1H), 7,66-7,53 (m, 3H), 7,35 (t, J = 6,4 Hz, 1H), 6,96-6,90 (m, 2H), 5,86 ( d, J = 7,2 Hz, 1H), 4,36 (s, 1H), 3,56 (s, 2H), 3,3 (s, 3H), 3,21 (s, 2H), 2, 97 (d, J = 6,0 Hz, 2H), 2,33 (s, 6H), 3,81–3,74 (m, 2H). ), 1,80-1,76 (metro, 2H), 1,54-1,51 (metro, 2H) FP PA42 3-Bromo-7-fluoroimidazo[1,2-a]pyridine

HPLC conditions

Chirale A1 method of analysis instrumentation Detector Agilent Serie 1260 HPLC y PDA To share Chiralpak IH (250 x 4.6 mm), 5 microns kept at temperature mobile phone, cell phone 0.1% DEA in n-hexane Fase A mobile phone, cell phone 0.1% DEA in Propan-2-ol: Acetonitrile (70:30) Fase B Flow 1.0ml/min time (minutes) % A %B Gradient 0,01 Sixty-five 35 program 25 Sixty-five 35 ultraviolet 308nm

Chiral analysis method A2 instrumentation Detector Agilent Serie 1260 HPLC y PDA To share Chiralpak-IC (250 x 4,6 mm), 5 micrones kept at temperature mobile phone, cell phone 0.1% DEA in n-hexane Fase A mobile phone, cell phone 0.1% DEA in Propan-2-ol: Acetonitrile (50:50) Fase B Flow 1.0ml/min time (minutes) % A %B Gradient 0,01 80 20 program 5 45 55 10 30 70 20 30 70 ultraviolet 276 nanometers

Chirale A3 method of analysis instrumentation Waters CFS Investigator and PDA Detector To share Chiralcel OJ-H (250 x 4,6 mm), 5 micras mobile phone, cell phone liquid carbon dioxide Fase A mobile phone, cell phone 0.1% DEA in methanol Fase B Flow 4.0ml/min time (minutes) % A %B Gradient 0,01 95 5 program 6 50 50 12 50 50 ultraviolet 290 nanometers

LCMS Conditions

Method: AcHSSC18 instrumentation UPLC + DAD Water + SQD2 Water, einzelnes Quadrapol-UPLC-MS To share Acquity UPLC HSS C18 1,8 um 100 × 2,1 mm. (Protection Plus cartridge), stored at room temperature mobile phone, cell phone Water (high purity via PureLab Option unit) Fase A with 0.1% formic acid mobile phone, cell phone Acetonitrile (Far Ultraviolet Grade) 0.1% Fase B (V/V) formic acid Flow 0,4ml/min time (minutes) % A %B Gradient 0,0 95 05 program 0,4 95 05 6.0 05 95 6.8 05 95 7,0 95 05 8.0 95 05 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

Method: 10 cm_Formic_AQ instrumentation UPLC + DAD Water + SQD2 Water, einzelnes Quadrapol-UPLC-MS To share Acquity UPLC HSS C18 1,8 um 100 × 2,1 mm. (Protection Plus cartridge), stored at room temperature mobile phone, cell phone Water (high purity via PureLab Option unit) Fase A with 0.1% formic acid mobile phone, cell phone Acetonitrile (Far Ultraviolet Grade) 0.1% Fase B (V/V) formic acid Flow 0,5ml/min time (minutes) % A %B Gradient 0,0 95 05 program 1.2 95 05 3.5 0 100 4.9 0 100 5 95 05 6 95 05 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

Method: 10 cm_Formic_ACE-AR_AQ instrumentation UPLC + DAD Water + SQD2 Water, einzelnes Quadrapol-UPLC-MS To share ACE-AR ACE excel 2um C18-AR, kept at temperature mobile phone, cell phone Water (high purity via PureLab Option unit) Fase A with 0.1% formic acid mobile phone, cell phone Acetonitrile (Far Ultraviolet Grade) 0.1% Fase B (V/V) formic acid Flow 0,5ml/min time (minutes) % A %B Gradient 0,0 95 05 program 1.2 95 05 3.5 0 100 4.9 0 100 5 95 05 6 95 05 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

Method: BicarbBEHC18 instrumentation UPLC + DAD Water + SQD2 Water, einzelnes Quadrapol-UPLC-MS To share Acquity UPLC BEH Shield RP18 1,7um 100×2,1 millimeter (Protection Plus cartridge), stored at room temperature mobile phone, cell phone Water (high purity via PureLab Option unit) Fase A 10 mM ammonium bicarbonate (ammonium hydrogen carbonate) mobile phone, cell phone Acetonitrile (Far UV Grade) Fase B Flow 0,4ml/min time (minutes) % A %B Gradient 0,0 95 05 program 0,4 95 05 6.0 05 95 6.8 05 95 7,0 95 05 8.0 95 05 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

Method: 10 cm_Bicarb_AQ instrumentation UPLC + DAD Water + SQD2 Water, einzelnes Quadrapol-UPLC-MS To share Acquity UPLC BEH Shield RP18 1,7um 100×2,1 millimeter (Protection Plus cartridge), stored at room temperature mobile phone, cell phone Water (high purity via PureLab Option unit) Fase A 10 mM ammonium bicarbonate (ammonium hydrogen carbonate) mobile phone, cell phone Acetonitrile (Far UV Grade) Fase B Flow 0,5ml/min time (minutes) % A %B Gradient 0,0 95 05 program 1.2 95 05 3.5 0 100 4.9 0 100 5 95 05 6 95 05 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

Method: PC instrumentation AGUAS ACQUETY class H with PDA and DETECTOR Q To share BEH C18 (50 × 2.1 mm) 1.7 μm maintained at temperature mobile phone, cell phone Ammonium acetate 2 mM Fase A mobile phone, cell phone 0.1% formic acid in acetonitrile Fase B Flow 0,55ml/min time (minutes) % A %B Gradient 0,0 98 02 program 0,3 98 2 0,6 50 50 1.10 25 75 2 0 100 2.7 0 100 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

GP method instrumentation RRLC Agilent 1290 Infinity con Agilent Connected 6120 Ground Detector and Diode Array Detector To share YMC Triart C18 (150 x 4.6 mm), 5 μm kept at room temperature mobile phone, cell phone 10 mM ammonium acetate Fase A mobile phone, cell phone 100% Acetonitrila Fase B Flow 1.0ml/min time (minutes) % A %B Gradient 0,01 100 0 program 7,0 50 50 9,0 0 100 11 0 100 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

FP method instrumentation RRLC Agilent 1290 Infinity con Agilent Connected 6120 Ground Detector and Diode Array Detector To share YMC Triart C18 (150 x 4.6 mm), 5 μm kept at room temperature mobile phone, cell phone 10 mM ammonium acetate Fase A mobile phone, cell phone 100% Acetonitrila Fase B Flow 1.0ml/min time (minutes) % A %B Gradient 0,01 90 10 program 5,0 10 90 7,0 0 100 11 0 100 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

HP method instrumentation Shimadzu Nexera High Pressure UHPLC e LCMS-2020 To share Puente X C18 (50*4,6mm), 3,5um kept at temperature mobile phone, cell phone 5 mM ammonium bicarbonate Fase A mobile phone, cell phone 100% Acetonitrila Fase B Flow 1.0ml/min time (minutes) % A %B Gradient 0,01 95 05 program 5,0 10 90 5.8 5 95 7.2 5 95 7.21 95 5 10 95 5 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

JP method instrumentation AGUAS ACQUETY class H with PDA and DETECTOR Q To share BEH C18 (50 x 2.1 mm) 1.7 µm held at room temperature mobile phone, cell phone Ammonium acetate 2 mM Fase A mobile phone, cell phone 0.1% formic acid in acetonitrile Fase B Flow 0,45ml/min time (minutes) % A %B Gradient 0,01 98 2 program 0,5 98 2 5,0 10 90 6.0 5 95 7,0 5 95 Al final 0.5-2 µl (concentration ~ 0.2-1 mg/ml). o detector UV, diode array 210nm-400nm 1.2nm resolution Other wavelength traces are extracted from the DAD data MS, mass 100-700 (or -1500 for the HM method) me ES+ & ES− 300 μl/min divided in MS)

HPLC preparative conditions

After synthesis, all compounds were analyzed by reverse-phase HPLC using a Waters Fractionlynx preparative HPLC system (2525 pump, 2996/2998 UV/VIS detector, 2767 liquid controller) or a Gilson preparative HPLC system (322 pump, 2767 detector). UV/VIS 155, Liquid Handler GX-281).

A Waters Sunfire OBD, Phenomenex Luna Phenyl Hexyl, or Waters Xbridge Phenyl at 10 µm 19 × 150 mm was used as a column for preparative purification of compounds.

Appropriate focused gradients were selected based on acetonitrile and methanol solvent systems under acidic or basic conditions. The standard gradient used was 5% ACN to 20% in 1 min, 2.5 min hold, to 80% ACN in 12.5 min, 7.5 min hold. Followed by 3 min of reequilibration to initial conditions. A flow rate of 20 ml/min is used.

All compounds were analyzed analytically before the purification step. Each sample was processed under acidic and basic conditions (2 µl injection, 5/95 gradient over 2.25 minutes). The analyst then made a decision on which pH and gradient to use based on where the desired product was being eluted and what separation was being achieved.

Modifiers used under acidic/basic conditions were formic acid (0.1% v/v) or ammonium bicarbonate (10 mM) or TFA (0.1% v/v) if method development was required.

Purification was monitored by monitoring the Waters FractionLynx software at 210-400 nm and triggered a collection threshold at 260 nm and the presence of target molecular ions observed under ESI conditions. The collected fractions were analyzed by LCMS (Waters Acquity Systems with Waters SQD). Fractions containing the desired product were dried overnight by Genevac lyophilization and then dried using commercial lyophilizers from BioPharma. Some of the compounds may have gone through a second purification process to achieve the required purity due to complex mixtures. For more challenging separations (eg method development), more focused gradients or isocratic conditions may have been used.

SFC Preliminary Conditions

When indicated, post-synthesis compounds were analyzed by supercritical fluid chromatography (SFC) using the Waters Thar Prep100 preparative SFC system (P200 CO2 pump, 2545 modifier pump, 2998 UV/VIS detector, 2767 liquid controller with injection stacked) or Waters Thar clean semi-prepared test system (Waters Fluid Delivery Module, 2998 UV/VIS Detector, Waters Fraction Collection Module). When the Waters 2767 Liquid Handler was used, it acted as an autosampler and fraction collector.

Compounds were purified using an appropriate column (YMC Amylose-C, YMC Cellulose-C, YMC Cellulose-SC, Phenomenex LUX Cellulose-3 or Phenomenex LUX Cellulose-4) unless otherwise indicated.

Appropriate isocratic processes were selected based on methanol, ethanol or isopropanol solvent systems under basic or unmodified conditions. The standard procedure used was modifier/CO2, 100 mL/min (or as appropriate), 120 bar back pressure, 40°C column temperature, with the specific modifier composition as indicated in method development.

All compounds were analyzed analytically before the purification step. Each sample was run in ethanol, methanol, and isopropanol under basic and unmodified conditions (2.0 µL injection, 5/55 gradient over 2.25 minutes). If necessary, a secondary screen can also be checked through advanced solvents such as acetonitrile, ethyl acetate and THE. The analyst then made a decision on which pH and isocratic conditions to use based on where the desired product was eluting and what separation was achieved.

The modifier used under basic conditions was diethylamine (0.1% v/v). Alternative modifiers such as formic acid (0.1% v/v), acetic acid (0.1% v/v), etc. It can be used as acid modifiers.

Purification was monitored by monitoring Waters FractionLynx or Waters ChromScope software at 210-400 nm and triggering a collection threshold at a suitable wavelength. The collected fractions were analyzed by SFC (Waters/Thar SFC with Waters SQD or Waters UPCC with Waters QDa systems). Fractions containing the desired product were concentrated by vacuum centrifugation and subsequently dried using standard Biopharma lyophilizers.

All samples were pre-purified with achiral systems and their purity was verified prior to chiral SFC purification.

Some of the compounds may have gone through a second purification process to achieve the required %ee or %purity.

Example 33. Biochemical assay of the enzyme HPK1

HPK1 Enzyme Biochemical Assay: HPK1 enzyme inhibition was measured using a microfluidic mobility shift assay. Reactions were performed in a 384-well plate containing 1.5 nM HPK1 (Invitrogen) in assay buffer (Carna Biosciences; pH 7.4). Test compounds were titrated on ten-point curves (3 µM final upper assay concentration) and preincubated with an enzyme/substrate mixture for 30 min before the reaction was started by addition of ATP (1 mM final concentration). ) and substrate (1 mM final concentration). final concentration). Carna Biosciences), diluted in assay buffer supplemented with MgCl2(final assay concentration 5 mM). After incubating at room temperature for 60 min, the reaction was terminated by adding 60 µl/well stop buffer (Carna Biosciences) and determining the signal using a Caliper EZ Reader (Perkin Elmer, UK).

Table 3 shows the activity of selected compounds of this invention in the biochemical assay for the HPK1 enzyme. The compound numbers correspond to the compound numbers in Table 1. Compounds with an activity designated "A" gave a CI50≤100nM; Compounds with a designated "B" activity provided a CI50>100 nM and ≤1000 nM; Compounds with an activity designated "C" provided a CI50>1000 nM. Tabla 3

TISCH 3 HPK1 1000UMATP HPK1 1000UMATP Brake Caliper IC50 (nM) Brake Caliper IC50 (nM) A <100 nM A <100 nM B 100-1000 nm B 100-1000 nm Connection C > 1000 nM Connection C > 1000 nM I-4 A I-5 B I-6 A I-7 B I-8 B I-9 B I 10 B I-11 B I-12 B I-13 B I-14 A I-15 B I-16 B I-17 B I-18 C I-19 B I-20 B I-21 B I-22 A I-23 B I-24 B I-25 B I-26 C I-28 B I-29 B I-30 B I-31 B I-32 B I-33 B I-34 B I-35 B I-36 A I-37 B I-38 A I-39 C I-40 B I-4I C I-42 B I-43 A I-44 A I-45 A I-46 B I-47 A I-48 B I-49 B I-50 B I-51 B I-52 B I-53 A I-54 B I-55 B I-56 C I-57 A I-58 A I-59 B I-60 B I-61 B I-62 B I-63 B I-64 B I-65 B I-66 B I-67 B I-68 B I-69 B I-70 B I-71 B I-72 B I-73 B I-74 A I-75 A I-76 B I-77 B I-78 C I-79 B I-80 B I-81 A I-82 B I-83 B I-84 B I-85 B I-86 B I-87 B I-88 A I-89 C I-90 B I-91 A I-92 B I-93 A I-94 A I-95 B I-96 B I-97 B I-98 A I-99 B I-100 A I-101 A I-102 A I-103 A I-104 C I-105 C I-106 A I-107 B I-108 A I-109 A I-110 A I-111 C I-112 C I-113 B I-114 A I-115 A I-116 B I-117 B I-118 B I-119 B I-120 B I-121 A I-122 B I-123 A I-124 B I-125 A I-126 B I-127 B I-128 B I-129 B I-130 A I-131 B I-132 B I-133 A I-134 A I-135 A I-136 A I-137 A I-138 B I-139 A I-140 A I-141 C I-142 A I-143 C I-144 C I-145 C I-146 B I-147 B I-148 C I-149 B I-150 B I-151 A I-152 B I-153 B I-154 B I-155 C I-156 B I-157 C I-158 A I-159 A I-160 A I-161 A I-162 A I-163 C I-164 A I-165 B I-166 A I-167 A I-168 A I-169 B I-170 B I-171 B I-172 C I-173 B I-174 C I-175 B I-176 A I-177 A I-178 B I-179 B I-180 A I-181 C I-182 C I-183 A I-184 C I-185 B I-186 A I-187 B I-188 A I-189 B I-190 B I-191 A I-192 B I-193 B I-194 A I-195 C I-196 B I-197 B I-198 B I-199 B I-200 A I-201 A I-202 A I-203 A I-204 A I-205 A I-206 A I-207 A I-208 B I-209 B I-210 A I-211 C I-212 B I-213 B I-214 B I-215 B I-216 C I-217 B I-218 A I-219 A I-220 A I-221 B I-222 A I-223 A I-224 B I-225 C I-226 A I-227 A I-228 A I-229 B I-230 A I-231 B I-232 A I-233 A I-234 A I-235 A I-236 B I-237 B I-238 B I-239 B I-240 B I-241 B I-242 B I-243 B I-244 A I-245 A I-247 A I-248 A I-249 A I-250 A I-251 A I-255 C I-256 C I-257 C I-258 B I-259 A I-260 B I-261 A I-262 C I-263 A I-264 A I-265 A I-266 A I-267 A I-268 C I-269 B I-270 A I-271 A I-273 A I-274 A I-275 A I-276 C I-277 A I-278 B I-279 A I-280 C I-281 A I-282 C I-283 A I-284 C

Top Articles
Latest Posts
Article information

Author: Rob Wisoky

Last Updated: 03/31/2023

Views: 6447

Rating: 4.8 / 5 (48 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Rob Wisoky

Birthday: 1994-09-30

Address: 5789 Michel Vista, West Domenic, OR 80464-9452

Phone: +97313824072371

Job: Education Orchestrator

Hobby: Lockpicking, Crocheting, Baton twirling, Video gaming, Jogging, Whittling, Model building

Introduction: My name is Rob Wisoky, I am a smiling, helpful, encouraging, zealous, energetic, faithful, fantastic person who loves writing and wants to share my knowledge and understanding with you.