SUBSTITUTED PHENYL-1H-PYRROLO[2,3-c] PYRIDINE DERIVATIVES

Abstract
The present invention relates to pharmaceutical agents useful for therapy and/or prophylaxis in a mammal, pharmaceutical composition comprising such compounds, and their use as menin/MLL protein/protein interaction inhibitors, useful for treating diseases such as cancer, including but not limited to leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN); and diabetes.
Description
FIELD OF THE INVENTION

The present invention relates to pharmaceutical agents useful for therapy and/or prophylaxis in a mammal, pharmaceutical composition comprising such compounds, and their use as menin/MLL protein/protein interaction inhibitors, useful for treating diseases such as cancer, including but not limited to leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN); and diabetes.


BACKGROUND OF THE INVENTION

Chromosomal rearrangements affecting the mixed lineage leukemia gene (MLL; MLL1; KMT2A) result in aggressive acute leukemias across all age groups and still represent mostly incurable diseases emphasizing the urgent need for novel therapeutic approaches. Acute leukemias harboring these chromosomal translocations of MLL represent as lymphoid, myeloid or biphenotypic disease and constitute 5 to 10% of acute leukemias in adults and approximately 70% in infants (Marschalek, Br J Haematol 2011. 152(2), 141-54; Tomizawa et al., Pediatr Blood Cancer 2007. 49(2), 127-32).


MLL is a histone methyltransferase that methylates histone H3 on lysine 4 (H3K4) and functions in multiprotein complexes. Use of inducible loss-of-function alleles of Mll1 demonstrated that Mll1 plays an essential role in sustaining hematopoietic stem cells (HSCs) and developing B cells although its histone methyltransferase activity is dispensable for hematopoiesis (Mishra et al., Cell Rep 2014. 7(4), 1239-47).


Fusion of MLL with more than 60 different partners has been reported to date and has been associated with leukemia formation/progression (Meyer et al., Leukemia 2013. 27, 2165-2176). Interestingly, the SET (Su(var)3-9, enhancer of zeste, and trithorax) domain of MLL is not retained in chimeric proteins but is replaced by the fusion partner (Thiel et al., Bioessays 2012. 34, 771-80). Recruitment of chromatin modifying enzymes like Dot1 L and/or the pTEFb complex by the fusion partner leads to enhanced transcription and transcriptional elongation of MLL target genes including HOXA genes (e.g. HOXA9) and the HOX cofactor MEIS1 as the most prominent ones. Aberrant expression of these genes in turn blocks hematopoietic differentiation and enhances proliferation.


Menin which is encoded by the Multiple Endocrine Neoplasia type 1 (MEN1) gene is expressed ubiquitously and is predominantly localized in the nucleus. It has been shown to interact with numerous proteins and is, therefore, involved in a variety of cellular processes. The best understood function of menin is its role as an oncogenic cofactor of MLL fusion proteins. Menin interacts with two motifs within the N-terminal fragment of MLL that is retained in all fusion proteins, MBM1 (menin-binding motif 1) and MBM2 (Thiel et al., Bioessays 2012. 34, 771-80). Menin/MLL interaction leads to the formation of a new interaction surface for lens epithelium-derived growth factor (LEDGF). Although MLL directly binds to LEDGF, menin is obligatory for the stable interaction between MILL and LEDGF and the gene specific chromatin recruitment of the MLL complex via the PWWP domain of LEDGF (Cermakova et al., Cancer Res 2014. 15, 5139-51; Yokoyama & Cleary, Cancer Cell 2008. 8, 36-46). Furthermore, numerous genetic studies have shown that menin is strictly required for oncogenic transformation by MLL fusion proteins suggesting the menin/MLL interaction as an attractive therapeutic target. For example, conditional deletion of Men1 prevents leukomogenesis in bone marrow progenitor cells ectopically expressing MLL fusions (Chen et al., Proc Natl Acad Sci 2006. 103, 1018-23). Similarly, genetic disruption of menin/MLL fusion interaction by loss-of-function mutations abrogates the oncogenic properties of the MLL fusion proteins, blocks the development of leukemia in vivo and releases the differentiation block of MLL-transformed leukemic blasts. These studies also showed that menin is required for the maintenance of HOX gene expression by MLL fusion proteins (Yokoyama et al., Cell 2005. 123, 207-18). In addition, small molecule inhibitors of menin/MLL interaction have been developed suggesting druggability of this protein/protein interaction and have also demonstrated efficacy in preclinical models of AML (Borkin et al., Cancer Cell 2015. 27, 589-602; Cierpicki and Grembecka, Future Med Chem 2014. 6, 447-462). Together with the observation that menin is not a requisite cofactor of MLL1 during normal hematopoiesis (Li et al., Blood 2013. 122, 2039-2046), these data validate the disruption of menin/MLL interaction as a promising new therapeutic approach for the treatment of MLL rearranged leukemia and other cancers with an active HOX/MEI gene signature. For example, an internal partial tandem duplication (PTD) within the 5′region of the MLL gene represents another major aberration that is found predominantly in de novo and secondary AML as well as myeloid dysplasia syndromes. Although the molecular mechanism and the biological function of MLL-PTD is not well understood, new therapeutic targeting strategies affecting the menin/MLL interaction might also prove effective in the treatment of MLL-PTD-related leukemias. Furthermore, castration-resistant prostate cancer has been shown to be dependent on the menin/MLL interaction (Malik et al., Nat Med 2015. 21, 344-52).


MLL protein is also known as Histone-lysine N-methyltransferase 2A (KMT2A) protein in the scientific field (UniProt Accession #Q03164).


Several references describe inhibitors targeting the menin-MLL interaction: WO2011029054, J Med Chem 2016, 59, 892-913 describe the preparation of thienopyrimidine and benzodiazepine derivatives; WO2014164543 describes thienopyrimidine and thienopyridine derivatives; Nature Chemical Biology March 2012, 8, 277-284 and Ren, J.; et al. Bioorg Med Chem Lett (2016), 26(18), 4472-4476 describe thienopyrimidine derivatives: J Med Chem 2014, 57, 1543-1556 describes hydroxy- and aminomethylpiperidine derivatives; Future Med Chen 2014, 6, 447-462 reviews small molecule and peptidomimetic compounds; WO2016195776 describes furo[2,3-d]pyrimidine, 9H-purine, [1,3]oxazolo[5,4-d]pyrimidine, [1,3]oxazolo[4,5-d]pyrimidine, [1,3]thiazolo[5,4-d]pyrimidine, thieno[2,3-b]pyridine and thieno[2,3-d]pyrimidine derivatives; WO2016197027 describes 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidine, 5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine, pyrido[2,3-d]pyrimidine and quinoline derivatives; and WO2016040330 describes thienopyrimidine and thienopyridine compounds. WO2017192543 describes piperidines as Menin inhibitors. WO2017112768, WO2017207387, WO2017214367, WO2018053267 and WO2018024602 describe inhibitors of the menin-MLL interaction. WO2017161002 and WO2017161028 describe inhibitors of menin-MLL. WO2018050686, WO2018050684 and WO2018109088 describe inhibitors of the menin-MLL interaction. WO2018226976 describes methods and compositions for inhibiting the interaction of menin with MLL proteins. WO2018175746 provides methods of treatment for hematological malignancies and Ewing's sarcoma. WO2018106818 and WO2018106820 provide methods of promoting proliferation of a pancreatic cell. WO2018153312 discloses azaspiro compounds relating to the field of medicinal chemistry. WO2017132398 discloses methods comprising contacting a leukemia cell exhibiting an NPM1 mutation with a pharmacologic inhibitor of interaction between MLL and Menin. WO2019060365 describes substituted inhibitors of menin-MLL. WO2020069027 describes the treatment of hematological malignancies with inhibitors of menin. Krivtsov et al., Cancer Cell 2019. No. 6 Vol. 36, 660-673 describes a menin-MLL inhibitor.


WO2014199171 discloses compounds as VAP1 inhibitors. WO2011113798 and WO2013037411 disclose compounds as SSAO inhibitors. WO2011056440 discloses compounds as CCR1 inhibitors.


WO2021060453 describes a crosslinking-type optically-active secondary amine derivative. WO2021121327 describes substituted straight chain spiro derivatives and their use as menin/MLL protein/protein interaction inhibitors.


DESCRIPTION OF THE INVENTION

The present invention concerns novel compounds of Formula (I),




embedded image


and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R8; —C(═O)—O—C1-4alkyl-NR22aR22b; —C(═O)—O—C1-4alkyl;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;

    • R19 represents hydrogen or C1-6alkyl;
    • or R18 and R19 are taken together to form —(CH2)3—, —(CH2)4— or —(CH2)5—;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three O-, S- or N-atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, —C1-4alkyl-OH, halo, CF3, C3-6cycloalkyl, Het3, and NR11c—R11d;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, cyano, and C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo and OR23;

    • or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, cyano, and C1-4alkyl substituted with one, two or three substituents each independently selected from the group consisting of halo and OR23;
    • R23 represents hydrogen or C1-4alkyl optionally substituted with one, two or three halo;


R1b represents hydrogen, F, Cl, or —O—C1-4alkyl;

    • R2 represents halo, C3-6cycloalkyl, C1-4alkyl, —O—C1-4alkyl, cyano, or C1-4alkyl substituted with one, two or three halo substituents;
    • R2 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n1 is selected from 1 and 2;


n2 is selected from 1, 2, 3 and 4;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen, C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b, —C(═O)—Het6a, —C(═O)—Het6b, —NR10c—C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4; or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl-S(═O)2—C1-4alkyl, and cyano;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; —(C═O)—C1-4alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl, —C(═O)—NR10aR10b, and —NR10c—C(═O)—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three substituents each independently selected from the group consisting of C1-4alkyl, halo, —O—C1-4alkyl, —CF3, —OH, —S(═O)2—C1-4alkyl, and —C(═O)—NR10aR10b;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2 wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, pyridazinyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a;


R6 and R6a are each independently selected from the group consisting of


Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a; —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b, Cy1, —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—N(C1-4alkyl)2, —C(═O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and —NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl,


—C(═O)—N(C1-4alkyl)2, —NH—S(═O)2—C1-4alkyl, and C7-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents hydrogen, —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, —S(═O)2—C1-4alkyl, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b, or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b, —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b.


Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl, cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b;


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—NR10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl, —O—C1-4alkyl, cyano,




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C7-4alkyl-Het3a; —C(═O)—R14;


C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and


C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R15a, R15b, R17a, R17b, R20a, R20b, R22a and R22b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a, R10b and R10c are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R10d and R10e are each independently selected from the group consisting of C1-4alkyl, —O—C1-4alkyl and C3-6cycloalkyl;


R14 represents Het5a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═O)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8;


and the pharmaceutically acceptable salts and the solvates thereof.


It should be clear that substituents R21 and —Y—R3 in Formula (I) can be attached to any carbon or nitrogen atom of the ring to which they are attached, thereby replacing hydrogens on the same atom or they may replace hydrogen atoms on different atoms in the moiety (including the N-atom). Lines drawn from substituents into ring systems indicate that the bond may be attached to any of the suitable ring atoms.


The present invention also concerns novel compounds of Formula (A),




embedded image


and the tautomers and the stereoisomeric forms thereof wherein


L is absent or represents —CH2— or —CH2—CH2—;


Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18; —C(═O)—O—C1-4alkyl-NR22aR22b; —C(═O)—O—C1-4alkyl;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


or R18 and R19 are taken together to form —(CH2)3—, —(CH2)4— or —(CH2)5—;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three O-, S- or N-atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, halo or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, —C1-4alkyl-OH, halo, CF3, C3-6cycloalkyl, Het3, and NR11cR11d; or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, cyano, and C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo and OR23;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, cyano, and C1-4alkyl substituted with one, two or three substituents each independently selected from the group consisting of halo and OR23;


R23 represents hydrogen or C1-4alkyl optionally substituted with one, two or three halo;


R1b represents hydrogen, F, Cl, or —O—C1-4alkyl;


R2 represents halo, C3-6cycloalkyl, C1-4alkyl, —O—C1-4alkyl, cyano, or C1-4alkyl substituted with one, two or three halo substituents;


R2a represents hydrogen or C1-4alkyl;


R21 represents hydrogen or —Ya—R3a; provided that when R2′ represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n3 is selected from 0 and 1;


n4 is selected from 0, 1, 2 and 3;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen, C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b, —C(═O)—Het6a, —C(═O)—Het6b, —NR10c—C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4;


or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl-S(═O)2—C1-4alkyl, and cyano;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; —(C═O)—C1-4alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl,


—C(═O)—NR10aR10b, and —NR10c—C(═O)—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three substituents each independently selected from the group consisting of C1-4alkyl, halo, —O—C1-4alkyl, —CF3, —OH, —S(═O)2—C1-4alkyl, and —C(═O)—NR10aR10b.


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2 wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, pyridazinyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a;


R6 and R6a are each independently selected from the group consisting of


Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a; —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b, Cy1, —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—N(C1-4alkyl)2, —C(═O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and


—NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—N(C1-4alkyl)2, —NH—S(═O)2—C1-4alkyl, and C1-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═—O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents hydrogen, —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, —S(═O)2—C1-4alkyl, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b, —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH4—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH1—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b. Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b;


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—N10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b, and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl, —O—C1-4alkyl, cyano,




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C1-4alkyl-Het3a; —C(═O)—R14; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and


C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R5a, R15b, R17a, R17b, R20a, R20b, R22a, and R22b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a, R10b and R10c are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R10d and R10e are each independently selected from the group consisting of C1-4alkyl, —O—C1-4alkyl and C3-6cycloalkyl;


R14 represents Het5a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═O)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8;


R24 represents hydrogen or C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


It should be clear that substituents R21, R24 and —Y—R3 in Formula (A) can be attached to any carbon or nitrogen atom of the ring to which they are attached, thereby replacing hydrogens on the same atom or they may replace hydrogen atoms on different atoms (including the N-atom) in the moiety. Lines drawn from substituents into ring systems indicate that the bond may be attached to any of the suitable ring atoms.


The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof.


The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, and a pharmaceutically acceptable carrier or excipient.


Additionally, the invention relates to a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, for use as a medicament, and to a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, for use in the treatment or in the prevention of cancer, including but not limited to leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN); and diabetes.


In a particular embodiment, the invention relates to a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, for use in the treatment or in the prevention of cancer.


In a specific embodiment said cancer is selected from leukemias, lymphomas, myelomas or solid tumor cancers (e.g. prostate cancer, lung cancer, breast cancer, pancreatic cancer, colon cancer, liver cancer, melanoma and glioblastoma, etc.). In some embodiments, the leukemias include acute leukemias, chronic leukemias, myeloid leukemias, myelogeneous leukemias, lymphoblastic leukemias, lymphocytic leukemias, Acute myelogeneous leukemias (AML), Chronic myelogenous leukemias (CML), Acute lymphoblastic leukemias (ALL), Chronic lymphocytic leukemias (CLL), T cell prolymphocytic leukemias (T-PLL), Large granular lymphocytic leukemia, Hairy cell leukemia (HCL), MLL-rearranged leukemias, MLL-PTD leukemias, MLL amplified leukemias, MLL-positive leukemias, leukemias exhibiting HOX/MEIS1 gene expression signatures etc.


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of leukemias, in particular nucleophosmin (NPM1)-mutated leukemias, e.g. NPM1c.


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may have improved metabolic stability properties.


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may have extended in vivo half-life (T½).


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may have improved oral bioavailability.


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may reduce tumor growth e.g., tumours harbouring MLL (KMT2A) gene rearrangements/alterations and/or NPM1 mutations.


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may have improved PD properties in vivo during a prolonged period of time, e.g. inhibition of target gene expression such as MEIS1 and upregulation of differentiation marker over a period of at least 16 hours.


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may have an improved safety profile (e.g. reduced hERG inhibition; improved cardiovascular safety).


In an embodiment, compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, may be suitable for Q.D. dosing (once daily).


The invention also relates to the use of a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, in combination with an additional pharmaceutical agent for use in the treatment or prevention of cancer, including but not limited to leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN); and diabetes.


Furthermore, the invention relates to a process for preparing a pharmaceutical composition according to the invention, characterized in that a pharmaceutically acceptable carrier is intimately mixed with a therapeutically effective amount of a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof.


The invention also relates to a product comprising a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, and an additional pharmaceutical agent, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of cancer, including but not limited to leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN); and diabetes.


Additionally, the invention relates to a method of treating or preventing a cell proliferative disease in a warm-blooded animal which comprises administering to the said animal an effective amount of a compound of Formula (I), a pharmaceutically acceptable salt, or a solvate thereof, as defined herein, or a pharmaceutical composition or combination as defined herein.


Any aspects of the invention and embodiments described herein for the compounds of formula (I) as listed herein, also hold for the compounds of formula (A).





BRIEF DESCRIPTION OF THE DRAWINGS

The summary, as well as the following detailed description, is further understood when read in conjunction with the appended drawings. For the purpose of illustrating the invention, there are shown in the drawings exemplary embodiments of the invention; however, the invention is not limited to the specific disclosure of the drawings. In the drawings:



FIG. 1 is an X-ray powder diffraction (XRPD) pattern of Compound 51 as a crystalline free base Form.



FIG. 2 is an X-ray powder diffraction (XRPD) pattern of Compound 51a as a crystalline HCl salt Form.



FIG. 3 is a Dynamic vapor sorption (DVS) isotherm plot of Compound 51a as a crystalline HCl salt Form.



FIG. 4 is a Dynamic vapor sorption (DVS) change in mass plot of Compound 51a as a crystalline HCl salt Form.





DETAILED DESCRIPTION OF THE INVENTION

The term ‘halo’ or ‘halogen’ as used herein represents fluoro, chloro, bromo and iodo.


The prefix ‘Cx-y’ (where x and y are integers) as used herein refers to the number of carbon atoms in a given group. Thus, a C1-6alkyl group contains from 1 to 6 carbon atoms, and so on.


The term ‘C1-4alkyl’ as used herein as a group or part of a group represents a straight or branched chain saturated hydrocarbon radical having from 1 to 4 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl, 1-butyl and the like.


Similar, the term ‘C1-6alkyl’ as used herein as a group or part of a group represents a straight or branched chain saturated hydrocarbon radical having from 1 to 6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl, t-butyl, n-pentyl, n-hexyl and the like.


Similar, the term ‘C1-8alkyl’ as used herein as a group or part of a group represents a straight or branched chain saturated hydrocarbon radical having from 1 to 8 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl, t-butyl, n-pentyl, n-hexyl,




embedded image


and the like.


The term ‘C3-6cycloalkyl’ as used herein as a group or part of a group defines a saturated, cyclic hydrocarbon radical having from 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.


The term ‘C3-7cycloalkyl’ as used herein as a group or part of a group defines a saturated, cyclic hydrocarbon radical having from 3 to 7 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.


It will be clear for the skilled person that S(═O)2 or SO2 represents a sulfonyl moiety.


It will be clear for the skilled person that CO or C(═O) represents a carbonyl moiety.


It will be clear for the skilled person that a group such as —NR— represents




embedded image


An example of such a group is —NRq—.


Non-limiting examples of ‘monocyclic 5- or 6-membered aromatic rings containing one, two or three nitrogen atoms and optionally a carbonyl moiety’, include, but are not limited to pyrazolyl, imidazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl or 1,2-dihydro-2-oxo-4-pyridinyl.


The skilled person will understand that a monocyclic 5- or 6-membered aromatic ring containing one, two or three nitrogen atoms and a carbonyl moiety includes, but is not limited to




embedded image


The term ‘monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N’, defines a fully or partially saturated, cyclic hydrocarbon radical having from 4 to 7 ring members and containing at least 1 nitrogen atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, which is attached to the remainder of the molecule of formula (I) via a nitrogen atom. Examples are N-linked azetidinyl, N-linked pyrrolidinyl, N-linked morpholinyl, N-linked thiomorpholinyl, N-linked piperazinyl, N-linked 1,4-diazepanyl, N-linked piperidinyl, and N-linked 1,2,3,6-tetrahydro-pyridinyl. Two R groups taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, are defined similar.


The term ‘monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N’, defines a fully or partially saturated, cyclic hydrocarbon radical having from 4 to 7 ring members and containing one, two or three heteroatoms each independently selected from O, S, and N, such as for example C-linked azetidinyl, C-linked pyrrolidinyl, C-linked morpholinyl, C-linked tetrahydrofuranyl, C-linked thiolanyl, C-linked oxetanyl, C-linked thietanyl, C-linked tetrahydropyranyl, C-linked tetrahydrothiopyranyl, C-linked piperidinyl, C-linked azepanyl, C-linked 1,3-dioxolanyl, and C-linked 1,2,3,6-tetrahydro-pyridinyl.


For clarity, the 4- to 7-membered fully or partially saturated heterocyclyls have from 4 to 7 ring members including the heteroatoms.


Non-limiting examples of ‘monocyclic C-linked 5- or 6-membered aromatic rings containing one, two or three heteroatoms each independently selected from O, S, and N’, include, but are not limited to C-linked pyrazolyl, C-linked imidazolyl, C-linked pyridinyl, C-linked triazolyl, C-linked pyridazinyl, C-linked pyrimidinyl, C-linked oxazolyl, C-linked furanyl, C-linked isothiazolyl, C-linked thiazolyl, C-linked thiadiazolyl, C-linked oxadiazolyl, or C-linked pyrazinyl.


Within the context of this invention, bicyclic 6- to 11-membered fully or partially saturated heterocyclyl groups, include fused, spiro and bridged bicycles.


Fused bicyclic groups are two cycles that share two atoms and the bond between these atoms. Spiro bicyclic groups are two cycles that are joined at a single atom.


Bridged bicyclic groups are two cycles that share more than two atoms.


Examples of bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, include, but are not limited to




embedded image


embedded image


embedded image


and the like.


Examples of bicy clic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, include, but are not limited to




embedded image


embedded image


embedded image


and the like.


Two R groups taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, are defined similar.


Examples of fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N, include but are not limited to




embedded image


embedded image


and the like.


As used herein ‘5- to 12-membered saturated carbobicyclic’ systems define saturated fused, spiro and bridged bicyclic hydrocarbon systems having from 5 to 12 carbon atoms. Examples of 5- to 12-membered saturated carbobicyclic’ systems include, but are not limited to




embedded image


and the like.


Whenever substituents are represented by chemical structure, such as for example




embedded image


‘----’ represents the bond of attachment to the remainder of the molecule of Formula (I).


When any variable occurs more than one time in any constituent, each definition is independent.


When any variable occurs more than one time in any formula (e.g. Formula (I)), each definition is independent.


It will be clear for a skilled person that when a moiety (for example a heterocyclyl or monocyclic 5- or 6-membered aromatic ring) is substituted with two or more substituents (for example one, two or three substituents) selected from a group, each substituent can be selected independently from said group, even if not explicitly mentioned.


In general, whenever the term ‘substituted’ is used in the present invention, it is meant, unless otherwise indicated or clear from the context, to indicate that one or more hydrogens, in particular from 1 to 4 hydrogens, more in particular from 1 to 3 hydrogens, preferably 1 or 2 hydrogens, more preferably 1 hydrogen, on the atom or radical indicated in the expression using ‘substituted’ are replaced with a selection from the indicated group, provided that the normal valency is not exceeded, and that the substitution results in a chemically stable compound, i.e. a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture (isolation after a reaction e.g. purification by silica gel chromatography). In a particular embodiment, when the number of substituents is not explicitly specified, the number of substituents is one.


Combinations of substituents and/or variables are permissible only if such combinations result in chemically stable compounds. ‘Stable compound’ is in this context meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture (isolation after a reaction e.g. purification by silica gel chromatography).


The skilled person will understand that the term ‘optionally substituted’ means that the atom or radical indicated in the expression using ‘optionally substituted’ may or may not be substituted (this means substituted or unsubstituted respectively).


When two or more substituents are present on a moiety they may, where possible and unless otherwise indicated or clear from the context, replace hydrogens on the same atom or they may replace hydrogen atoms on different atoms in the moiety.


Within the context of this invention ‘saturated’ means ‘fully saturated’, if not otherwise specified.


Unless otherwise specified or clear from the context, aromatic rings and heterocyclyl groups, can be attached to the remainder of the molecule of Formula (I) through any available ring carbon atom (C-linked) or nitrogen atom (N-linked).


Unless otherwise specified or clear from the context, aromatic rings and heterocyclyl groups, may optionally be substituted, where possible, on carbon and/or nitrogen atoms according to the embodiments. A skilled person will understand that in such a case hydrogens on the carbon and/or nitrogen atoms are replaced by such substituents.


Unless otherwise specified or clear from the context, variable R21 and —Y—R3 can be attached to any carbon or nitrogen atom of the ring to which they are attached, provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring.


For example in case R21 represents hydrogen, and —Y—R3 is attached to the nitrogen atom of the ring in Formula (I), a compound of subformula (I-x) is obtained:




embedded image


In case Y represents a covalent bond in Formula (I), a compound of subformula (I-y) is obtained:




embedded image


In case Y represents




embedded image


in Formula (I), a compound of subformula (I-z) is obtained:




embedded image


The term “subject” as used herein, refers to an animal, preferably a mammal (e.g. cat, dog, primate or human), more preferably a human, who is or has been the object of treatment, observation or experiment.


The term “therapeutically effective amount” as used herein, means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medicinal doctor or other clinician, which includes alleviation or reversal of the symptoms of the disease or disorder being treated.


The term “composition” is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts.


The term “treatment”, as used herein, is intended to refer to all processes wherein there may be a slowing, interrupting, arresting or stopping of the progression of a disease, but does not necessarily indicate a total elimination of all symptoms.


The term “compound(s) of the (present) invention” or “compound(s) according to the (present) invention” as used herein, is meant to include the compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof.


As used herein, any chemical formula with bonds shown only as solid lines and not as solid wedged or hashed wedged bonds, or otherwise indicated as having a particular configuration (e.g. R, S) around one or more atoms, contemplates each possible stereoisomer, or mixture of two or more stereoisomers.


Hereinbefore and hereinafter, the term “compound(s) of Formula (I)” is meant to include the tautomers thereof and the stereoisomeric forms thereof.


The terms “stereoisomers”, “stereoisomeric forms” or “stereochemically isomeric forms” hereinbefore or hereinafter are used interchangeably.


The invention includes all stereoisomers of the compounds of the invention either as a pure stereoisomer or as a mixture of two or more stereoisomers.


Enantiomers are stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a racemate or racemic mixture.


Atropisomers (or atropoisomers) are stereoisomers which have a particular spatial configuration, resulting from a restricted rotation about a single bond, due to large steric hindrance. All atropisomeric forms of the compounds of Formula (I) are intended to be included within the scope of the present invention.


Diastereomers (or diastereoisomers) are stereoisomers that are not enantiomers, i.e. they are not related as mirror images. If a compound contains a double bond, the substituents may be in the E or the Z configuration.


Substituents on bivalent cyclic saturated or partially saturated radicals may have either the cis- or trans-configuration; for example if a compound contains a disubstituted cycloalkyl group, the substituents may be in the cis or trans configuration.


Therefore, the invention includes enantiomers, atropisomers, diastereomers, racemates, E isomers, Z isomers, cis isomers, trans isomers and mixtures thereof, whenever chemically possible.


The meaning of all those terms, i.e. enantiomers, atropisomers, diastereomers, racemates, E isomers, Z isomers, cis isomers, trans isomers and mixtures thereof are known to the skilled person.


The absolute configuration is specified according to the Cahn-Ingold-Prelog system. The configuration at an asymmetric atom is specified by either R or S. Resolved stereoisomers whose absolute configuration is not knon can be designated by (+) or (−) depending on the direction in which they rotate plane polarized light. For instance, resolved enantiomers whose absolute configuration is not known can be designated by (+) or (−) depending on the direction in which they rotate plane polarized light.


When a specific stereoisomer is identified, this means that said stereoisomer is substantially free, i.e. associated with less than 50%, preferably less than 20%, more preferably less than 10%, even more preferably less than 5%, in particular less than 2% and most preferably less than 1%, of the other stereoisomers. Thus, when a compound of Formula (I) is for instance specified as (R), this means that the compound is substantially free of the (S) isomer; when a compound of Formula (I) is for instance specified as E, this means that the compound is substantially free of the Z isomer; when a compound of Formula (I) is for instance specified as cis, this means that the compound is substantially free of the trans isomer.


Some of the compounds according to Formula (I) may also exist in their tautomeric form. Such forms in so far as they may exist, although not explicitly indicated in the above Formula (I) are intended to be included within the scope of the present invention. It follows that a single compound may exist in both stereoisomeric and tautomeric form.


Pharmaceutically acceptable salts include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form with one or more equivalents of an appropriate base or acid, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.


The pharmaceutically acceptable salts as mentioned hereinabove or hereinafter are meant to comprise the therapeutically active non-toxic acid and base salt forms which the compounds of Formula (I) and solvates thereof, are able to form.


Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e. ethanedioic), malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form.


The compounds of Formula (I) and solvates thereof containing an acidic proton may also be converted into their non-toxic metal or amine salt forms by treatment with appropriate organic and inorganic bases.


Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, cesium, magnesium, calcium salts and the like, salts with organic bases, e.g. primary, secondary and tertiary aliphatic and aromatic amines such as methylamine, ethylamine, propylamine, isopropylanine, the four butylamine isomers, dimethylamine, diethylamine, diethanolamine, dipropylamine, diisopropylamine, di-n-butylamine, pyrrolidine, piperidine, morpholine, trimethylamine, triethylamine, tripropylamine, quinuclidine, pyridine, quinoline and isoquinoline; the benzathine, N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. Conversely the salt form can be converted by treatment with acid into the free acid form.


The term “prodrug” includes any compound that, following oral or parenteral administration, in particular oral administration, is metabolised in vivo to a (more) active form in an experimentally-detectable amount, and within a predetermined time (e.g. within a dosing interval of between 0.5 and 24 hours, or e.g. within a dosing interval of between 6 and 24 hours (i.e. once to four times daily)). For the avoidance of doubt, the term “parenteral” administration includes all forms of administration other than oral administration, in particular intravenous (IV), intramuscular (IM), and subcutaneous (SC) injection.


Prodrugs may be prepared by modifying functional groups present on a compound in such a way that the modifications are cleaved in vivo when such prodrug is administered to a mammalian subject. The modifications typically are achieved by synthesising the parent compound with a prodrug substituent. In general, prodrugs include compounds wherein a hydroxyl, amino, sulfhydryl, carboxy or carbonyl group is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, sulfhydryl, carboxy or carbonyl group, respectively.


Examples of prodrugs include, but are not limited to, esters and carbamates of hydroxy functional groups, esters groups of carboxyl functional groups, N-acyl derivatives and N-Mannich bases. General information on prodrugs may be found e.g. in Bundegaard, H. “Design of Prodrugs” p. 1-92, Elesevier, New York-Oxford (1985).


The term solvate comprises the solvent addition forms as well as the salts thereof, which the compounds of Formula (I) are able to form. Examples of such solvent addition forms are e.g. hydrates, alcoholates and the like.


The compounds of the invention as prepared in the processes described below may be synthesized in the form of mixtures of enantiomers, in particular racemic mixtures of enantiomers, that can be separated from one another following art-known resolution procedures. A manner of separating the enantiomeric forms of the compounds of Formula (I), and pharmaceutically acceptable salts, and solvates thereof, involves liquid chromatography using a chiral stationary phase. Said pure stereochemically isomeric forms may also be derived from the corresponding pure stereochemically isomeric forms of the appropriate starting materials, provided that the reaction occurs stereospecifically. Preferably if a specific stereoisomer is desired, said compound would be synthesized by stereospecific methods of preparation. These methods will advantageously employ enantiomerically pure starting materials.


The term “enantiomerically pure” as used herein means that the product contains at least 80% by weight of one enantiomer and 20% by weight or less of the other enantiomer. Preferably the product contains at least 90% by weight of one enantiomer and 10% by weight or less of the other enantiomer. In the most preferred embodiment the term “enantiomerically pure” means that the composition contains at least 99% by weight of one enantiomer and 1% or less of the other enantiomer.


The present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (or the most abundant one found in nature).


All isotopes and isotopic mixtures of any particular atom or element as specified herein are contemplated within the scope of the compounds of the invention, either naturally occurring or synthetically produced, either with natural abundance or in an isotopically enriched form. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15O, 17O, 18O, 32P, 33P, 35S, 18F, 36C, 122I, 123I, 125I, 131I, 75Br, 76Br, 77Br and 82Br. Preferably, the isotope is selected from the group of 2H, 3H, 11C, 13C and 18F. Preferably, the isotope is selected from the group of 2H, 3H, 11C and 18F. More preferably, the isotope is 2H, 3H or 13C. More preferably, the isotope is 2H or 13C. More preferably, the isotope is 2H. In particular, deuterated compounds and 13C-enriched compounds are intended to be included within the scope of the present invention. In particular, deuterated compounds are intended to be included within the scope of the present invention.


Certain isotopically-labeled compounds of the present invention (e.g., those labeled with 3H and NC) may be useful for example in substrate tissue distribution assays. Tritiated (H) and carbon-14 (14C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 15O, 13N, 11C and 18F are useful for positron emission tomography (PET) studies. PET imaging in cancer finds utility in helping locate and identify tumours, stage the disease and determine suitable treatment. Human cancer cells overexpress many receptors or proteins that are potential disease-specific molecular targets. Radiolabelled tracers that bind with high affinity and specificity to such receptors or proteins on tumour cells have great potential for diagnostic imaging and targeted radionuclide therapy (Charron, Carlie L. et al. Tetrahedron Lett. 2016, 57(37), 4119-4127). Additionally, target-specific PET radiotracers may be used as biomarkers to examine and evaluate pathology, by for example, measuring target expression and treatment response (Austin R. et al. Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.008).


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy— or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; halo; —C(═O)—NRxaRxb; —S(═O)2—R18;


—C(═O)—O—C1-4alkyl; or




embedded image


R18 represents C1-6alkyl;


R19 represents hydrogen or C1-6alkyl;


or R18 and R19 are taken together to form —(CH2)3—, —(CH2)4— or —(CH2)5—;


Rxa and Rxb are each independently selected from the group consisting of hydrogen;


Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and —C1-4alkyl-OH;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, —O—C1-4alkyl, and C1-4alkyl substituted with one, two or three OR23;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —OH substituents;


R23 represents hydrogen or C1-4alkyl;


R1b represents F or —O—C1-4alkyl;


R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R1 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


R5 represents hydrogen;


n1 is selected from 1 and 2;


n2 is selected from 1, 2 and 3;


Ry represents hydrogen;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)-Het6a, —C(═O)-Het6b, —NR10c—C(═O)—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl, and —S(═O)2—C1-4alkyl;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; —(C═O)—C1-4alkyl; and C1-6alkyl substituted with one, two or three —O—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three substituents each independently selected from the group consisting of C1-4alkyl and —C(═O)—NR10aR10b;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, C1-4alkyl, oxo and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, pyridazinyl or triazolyl;


R6 is selected from the group consisting of Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a; —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het6a, Het6b, and —OH;


R8 represents hydrogen, —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, cyano, —S(═O)2—C1-4alkyl, and Het3a;


Het3 and Het3a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen atom with —(C═O)—C1-4alkyl;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl and —C(═O)—NR10aR10b:


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo and —S(═O)2—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a —C(═O)—C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three —OH;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, and C1-4alkyl;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; —C(═O)—C1-4alkyl; —S(═O)2—C1-4alkyl; and —C(═O)—R14;


R10a, R10b and R10c are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R10d and R10c are each independently selected from the group consisting of C1-4alkyl and —O—C1-4alkyl;


R14 represents —O—C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(O)—NRxaRxb; —S(═O)2—R18;


—C(═O)—O—C1-4alkyl; or




embedded image


R18 represents C1-6alkyl;


R19 represents hydrogen or C1-6alkyl;


or R18 and R19 are taken together to form —(CH2)3—, —(CH2)4— or —(CH2)5—;


Rxa and Rxb are each independently selected from the group consisting of hydrogen;


Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and —C1-4alkyl-OH;

    • or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, —O—C1-4alkyl, and C1-4alkyl substituted with one, two or three OR23;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —OH substituents;


R23 represents hydrogen or C1-4alkyl;


R1b represents F or —O—C1-4alkyl;


R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya represent a covalent bond;


n1 is selected from 1 and 2;


n2 is selected from 1, 2 and 3;


Ry represents hydrogen;


R3 and R3a are each independently selected from the group consisting of Het1; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—Het6a, —C(═O)—Het6b, —NR10c—C(═O)—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl, and —S(═O)2—C1-4alkyl;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; —(C═O)—C1-4alkyl; and C1-6alkyl substituted with one, two or three —O—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three substituents each independently selected from the group consisting of C1-4alkyl and —C(═O)—NR10aR10b;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, C1-4alkyl, oxo and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, pyridazinyl or triazolyl; R6 is selected from the group consisting of Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a; —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het6a, Het6b, and —OH;


R8 represents hydrogen, —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, cyano, —S(═O)2—C1-4alkyl, and Het3a;


Het3 and Het3a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen atom with —(C═O)—C1-4alkyl;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl and —C(═O)—NR10aR10b;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo and —S(═O)2—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a —C(═O)—C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three —OH;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6, Het6b, —NR9aR9b, —OH, and C1-4alkyl;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; —C(═O)—C1-4alkyl; —S(═O)2—C1-4alkyl; and —C(═O)—R14;


R10a, R10b and R10c are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R10d and R10e are each independently selected from the group consisting of C1-4alkyl and —O—C1-4alkyl;


R14 represents —O—C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb; —S(═O)2—R18;


—C(═O)—O—C1-4alkyl; or




embedded image


R18 represents C1-6alkyl;


R19 represents hydrogen or C1-6alkyl;


or R18 and R19 are taken together to form —(CH2)3—;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and —C1-4alkyl-OH;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, —O—C1-4alkyl, and C1-4alkyl substituted with one, two or three OR2;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —OH substituents;


R23 represents hydrogen or C1-4alkyl;


R1b represents F or —O—C1-4alkyl;


R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond;


n1 is selected from 1 and 2:


n2 is selected from 1, 2 and 3:


Ry represents hydrogen;


R3 and R3a are each independently selected from the group consisting of Het1; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)-Het6a, —C(═O)—Het6b, —NR10c—C(═O)—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl, and —S(═O)2—C1-4alkyl;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; —(C═O)—C1-4alkyl; and C1-6alkyl substituted with one, two or three —O—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three —C(═O)—NR10aR10b.


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, C1-4alkyl, oxo and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, or pyridazinyl;


R6 is selected from the group consisting of Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a; —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het6a, Het6b, and —OH;


R8 represents hydrogen, —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, cyano and Het3a;


Het3 and Het3a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen atom with —(C═O)—C1-4alkyl;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b.


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four halo; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a —C(═O)—C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three —OH;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, and C1-4alkyl; R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; —C(═)—C1-4alkyl; —S(═O)2—C1-4alkyl; and —C(═O)—R14;


R10a, R10b and R10c are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R10d and R10e are each independently selected from the group consisting of C1-4alkyl and —O—C1-4alkyl;


R14 represents —O—C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy— or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; halo; —C(═O)—NRxaRxb; —S(═O)2—R18.


—C(═O)—O—C1-4alkyl;




embedded image


R18 represents C1-6alkyl;


R19 represents hydrogen or C1-6alkyl;


or R18 and R19 are taken together to form —(CH2)3—;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and —C1-4alkyl-OH;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, and C1-4alkyl substituted with one, two or three substituents selected from the group consisting of OR23;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —OH substituents;


R23 represents hydrogen or C1-4alkyl;


R1b represents F;


R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n1 is selected from 1 and 2;


n2 is selected from 1, 2 and 3;


Ry represents hydrogen;


R5 represents hydrogen;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—Het6a, —C(═O)—Het6b, —NR10c—C(═O)—C1-4alkyl,


—NRxcRxd, —NR8aR8b, —CF3, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; and C1-6alkyl substituted with one —O—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three substituents each independently selected from the group consisting of C1-4alkyl and —C(═O)—NR10aR10b;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, C1-4alkyl, oxo, and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, pyridazinyl or triazolyl;


R6 is selected from the group consisting of Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a; —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two —OH substituents; and


C3-6cycloalkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, cyano, —S(═O)2—C1-4alkyl, and Het3a;


Het3 and Het3a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one carbon atom with oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with


—(C═O)—C1-4alkyl;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl and —C(═O)—NR10aR10b;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo and —S(═O)2—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a —C(═O)—C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three —OH;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, and C1-4alkyl;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; —C(═O)—C1-4alkyl; —S(═O)2—C1-4alkyl; and —C(═O)—R14;


R10a, R10b and R10c are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R10d and R10e are each independently selected from the group consisting of C1-4alkyl and —O—C1-4alkyl;


R14 represents —O—C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three nitrogen atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and NR11cR11d;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


R1b represents hydrogen, F or Cl;


R2 represents halo, C3-6cycloalkyl, C1-4alkyl, —O—C1-4alkyl, cyano, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen, C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b, —NR10c—C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4; or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl-S(═O)2—C1-4alkyl, and cyano;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl, —C(═O)—NR10aR10b, and —NR10c—C(═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a;


R6 and R6a are each independently selected from the group consisting of


Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—RB; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b, Cy1, —CN, —OH, —O—C1-4alkyl,


—C(═O)—NH—C1-4alkyl, —C(═O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and —NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl,


—NH—S(═O)2—C1-4alkyl, and C1-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b, or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b, —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b; Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo,


—NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl, cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b;


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—NR10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b, and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl,




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C1-4alkyl-Het3a; —C(═O)—R14;


C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and


C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R15a, R15b, R17a, R17b, R20a, and R20b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R14 represents Het5a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═O)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three nitrogen atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and NR11cR11d;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


R1b represents hydrogen, F or Cl;


R2 represents halo, C3-6cycloalkyl, C1-4alkyl, —O—C1-4alkyl, cyano, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen, C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4;


or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl-S(═O)2—C1-4alkyl, and cyano;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl, and —C(═O)—NR10aR10b;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a;


R6 and R6a are each independently selected from the group consisting of


Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b, Cy1, —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and —NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl,


—NH—S(═O)2—C1-4alkyl, and C1-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2;


wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b, or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b, —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b;


Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl, cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b;


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—NR10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b, and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl,




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen;


C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C1-4alkyl-Het3a; —C(═O)—R14;


C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and


C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R15a, R15b, R17a, R17b, R20a and R20b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R14 represents Het5a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═O)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three nitrogen atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and NR11cR11d;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


R1b represents hydrogen, F or Cl;


R2 represents halo, C3-6cycloalkyl, C1-4alkyl, —O—C1-4alkyl, cyano, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen, C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b, —NR10c—C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4;


or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl-S(═O)2—C1-4alkyl, and cyano; R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl, —C(═O)—NR10aR10b, and —NR10c—C(═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a;


R6 and R6a are each independently selected from the group consisting of


Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—RB; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b, Cy1, —CN, —OH, —O—C1-4alkyl,


—C(═O)—NH—C1-4alkyl, —C(═O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and —NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl,


—NH—S(═O)2—C1-4alkyl, and C1-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2;


wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b, or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b, —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b;


Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl, cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b;


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—NR10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b, and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7Cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl, C1-4alkyl C1-4alkyl




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C1-4alkyl-Het3a; —C(═O)—R14;


C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R15a, R15b, R17a, R17b, R20a, and R20b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R14 represents Het5a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═O)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three nitrogen atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, and NR11cR11d;


or Rax and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


or Rax and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, —C1-4alkyl-O—C1-4alkyl, and cyano;


R1b represents hydrogen, F or Cl;


R2 represents C1-4alkyl; in particular R2 represents methyl;


R21 represents hydrogen or —Ya—R3a; provided that when R21 represents —Ya—R3a, one of —Ya—R3a and —Y—R3 is attached to the nitrogen atom of the ring;


Y and Ya each independently represent a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen, C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b,


—NR10c—C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4; or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl —S(═O)2—C1-4alkyl, and cyano;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl, —C(═O)—NR10aR10b, and —NR10c—C(═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a;


R6 and R6a are each independently selected from the group consisting of


Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—RB; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b, Cy1, —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and —NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl,


—NH—S(═O)2—C1-4alkyl, and C1-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b, or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b, —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b;


Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo,


—NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl, cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b;


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—NR10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b, and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl,




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C1-4alkyl-Het3a; —C(═O)—R14;


C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R15a, R15b, R17a, R17b, R20a, and R20b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R14 represents Het8a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═O)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8; and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; halo; —C(═O)—NRxaRxb; or




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


Rax and Rxb are each independently selected from the group consisting of hydrogen; Het3; and C1-6alkyl; wherein optionally said C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, and —OC1-4alkyl; or Rax and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, and —O—C1-4alkyl;


or Rax and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, and —O—C1-4alkyl;


R1b represents F;


R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen;


Y represents a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen;


R5 represents hydrogen;


R3 and R4 are each independently selected from the group consisting of Het1; Cy2;


C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, —NR8aR8b, —CF3, —OH, Het1, and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


R8a and R8b are each independently selected from the group consisting of C1-6alkyl; and C1-6alkyl substituted with one —O—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of oxo and —NR9aR9b;


R6 represents Het4; —C(═O)—NH—R8; —S(═O)2—C1-4alkyl; or C1-6alkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —O—C1-4alkyl, and cyano;


Het3 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two —S(═O)2—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b,




embedded image


R9a and R9b are each independently selected from the group consisting of hydrogen;


C1-4alkyl; —C(═O)—C1-4alkyl; and —S(═O)2—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;


R1a represents hydrogen; halo; or —C(═O)—NRxaRxb;


Rxa and Rxb are each independently selected from the group consisting of hydrogen and C1-6alkyl;


R1b represents F;


R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen;


Y represents a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen;


R5 represents hydrogen;


R3 and R4 are each independently selected from the group consisting of Het1; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, —NR8aR8b, Het1, and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


R8a and R8b are each independently selected from the group consisting of C1-6alkyl; and C1-6alkyl substituted with one —O—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8;


R6 represents Het4; —C(═O)—NH—R8; or —S(═O)2—C1-4alkyl;


R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —O—C1-4alkyl, and cyano;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two —S(═O)2—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, Het6a, Het6b, and —NR9aR9b;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; —C(═O)—C1-4alkyl; and —S(═O)2—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Ra and Rb represent C1-6alkyl;


R1b represents F;


R2 represents halo or C1-4alkyl;


R21 represents hydrogen;


Y represents a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen;


R5 represents hydrogen;


R3 is selected from the group consisting of Het1; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2;


R4 represents C1-6alkyl; in particular isopropyl;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8;


R6 represents Het4 or —C(═O)—NH—RB;


R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —O—C1-4alkyl, and cyano;


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, Het6a, Het6b, and —NR9aR9b; R9a and R9b are each independently selected from the group consisting of hydrogen; and —S(═O)2—C1-4alkyl;


R10a and R10b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Rxa and Rxb represent C1-6alkyl;


R1b represents F;


R2 represents C1-4alkyl;


R21 represents hydrogen;


Y represents a covalent bond or




embedded image


n1 and n2 are each independently selected from 1 and 2;


Ry represents hydrogen;


R5 represents hydrogen;


R3 is selected from the group consisting of Cy2; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2;


R4 represents C1-6alkyl; in particular isopropyl;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl; Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8;


R6 represents —C(═O)—NH—R8;


R8 represents C1-6alkyl;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6 and Het6a; and the pharmaceutically acceptable salts and the solvates thereof.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb,


Rxa and Rxb are C1-6alkyl optionally substituted with 1, 2 or 3 —OH;

    • R1b represents F;
    • R2 represents methyl;


R21 represents hydrogen or methyl;


Y represents a covalent bond;


n1 is 1;


n2 is selected from 1 and 2;


Ry represents hydrogen;


R3 is selected from C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxc, Het1 and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R; and wherein said heterocyclyl is optionally substituted on one carbon atom with oxo;


R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom night be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one Het6a;


and the pharmaceutically acceptable salts and the solvates thereof.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Q represents —CHRy— or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Q represents —CHRy—.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R1a represents hydrogen; Het; —C(═O)—NRxaRxb; —S(═O)2—R18;




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R1a represents Het; —C(—O)—NRxaRxb; —S(═O)2—R18;




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R1a represents —C(═O)—NRxaRxb; —S(═O)2—R18; or




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R1a represents —C(═O)—NRxaRxb; or




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R1a represents —C(═O)—NRxaRxb.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R18 represents C1-6alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxa and Rxb represent hydrogen or C1-6alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; and C1-6alkyl; wherein optionally said C1-8alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, and —OC1-4alkyl;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, and —O—C1-4alkyl;


or Rax and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, —OH, and —O—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; and C1-6alkyl; wherein optionally said C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, and —OC1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxa and Rxb represent C1-6alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxa and Rxb are taken together.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxa and Rxb are not taken together.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R1b represents F or Cl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R1b represents F.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R2 represents halo or C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R2 represents C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R2 represents methyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R2 represents methyl; and R1a represents —C(═O)—NRxaRxb.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Y and Ya represent a covalent bond.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R21 represents hydrogen.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R21 represents hydrogen or methyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R21 represents hydrogen; and


Y represents a covalent bond.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R21 represents hydrogen or methyl; and


Y represents a covalent bond.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R21 represents —Ya—R3a.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R21 represents hydrogen, C1-6alkyl, C3-6cycloalkyl, or C1-6alkyl substituted with 1 substituent selected from the group consisting of halo, —OH, —O—C1-4alkyl, —C(═O)—NR10aR10b, —NR10c—C(═O)—C1-4alkyl, and —S(═O)2—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R21 represents C1-6alkyl, C3-6cycloalkyl, or C1-6alkyl substituted with 1 substituent selected from the group consisting of halo, —OH, —O—C1-4alkyl, —C(═O)—NR10aR10b, —NR10c—C(═O)—C1-4alkyl, and —S(═O)2—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R10c is selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, and Cy2;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl, and —C(═O)—NR10aR10b.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3, R3a, and R4 are not C1-6alkyl substituted with —NR10b—C(═O)—C1-4alkyl;


R8a and R8b are not C1-6alkyl substituted with —NR10c—C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Y represents a covalent bond.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Ya represents a covalent bond.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Y represents




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Ya represents




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein n1 represents 1, and n2 represents 2.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R4 represents C1-6alkyl; oxetanyl; tetrahydropyranyl;




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Ry represents hydrogen.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R4 represents C1-6alkyl; oxetanyl; tetrahydropyranyl;




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R4 represents C1-6alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R4 represents isopropyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R4 represents C1-8alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R4 represents C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R5 represents hydrogen.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3 and R4 are each independently selected from the group consisting of Het1; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, —NR8aR8b, Het1, and Cy2.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3 is selected from the group consisting of Het1; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2; and


R4 represents C1-6alkyl; in particular isopropyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3 is selected from the group consisting of Cy2; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2; and


R4 represents C1-6alkyl; in particular isopropyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3 is selected from the group consisting of Het1; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R3 is selected from the group consisting of Cy2; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —NR9aR9b, and —OH.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;

    • or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxc and Rxd are taken together.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Rxc and Rxd are not taken together.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein fully or partially saturated heterocyclyl groups are limited to fully saturated heterocyclycl groups.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb.


R1b represents F;


R2 represents methyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R8a and R8b are each independently selected from the group consisting of C1-6alkyl; and C1-6alkyl substituted with one —O—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of oxo and —NR9aR9b.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R6 represents Het4 or —C(═O)—NH—R.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —O—C1-4alkyl, and cyano.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R8 represents C1-6alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R8 represents methyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b,




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, Het6a, Het6b, —NR9aR9b,




embedded image


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, Het6a, Het6b, and —NR9aR9b.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R9a and R9b are each independently selected from the group consisting of hydrogen; and —S(═O)2—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein R10a and R10b are each independently selected from the group consisting of hydrogen and C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein when Rxa and Rxb are taken together to form a monocyclic heterocyclyl they represent 1-pyrrolidinyl or 1-piperidinyl, each optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein when Rxa and Rxb are taken together to form a bicyclic heterocyclyl they represent




embedded image


each optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein when Rxc and Rxd are taken together to form a monocyclic heterocyclyl they represent 1-pyrrolidinyl, 1-piperidinyl, or 1-piperazinyl, each optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein when Rxc and Rxd are taken together to form a bicyclic heterocyclyl they represent




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het1 represents




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het1 represents




embedded image


optionally substituted on a nitrogen atom with —C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het1 represents




embedded image


substituted on a nitrogen atom with —C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a fused or spiro bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a fused or spiro bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of oxo and —NR9aR9b.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of oxo and —NR9aR9b.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het3 represents




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het4 represents C-linked pyrazinyl optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het6a represents




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het6a represents




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het6a represents




embedded image


substituted on a nitrogen atom with —C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het6b represents




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Het6b represents




embedded image


substituted on a nitrogen atom with —C(═O)—C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein Cy2 represents C3-7cycloalkyl,




embedded image


optionally substituted as defined in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein C1-8alkyl is limited to C1-6alkyl, in particular wherein C1-8alkyl is limited to C1-4alkyl.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein —Y—R3 is attached to the nitrogen atom of the ring.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein


R21 is hydrogen, and wherein —Y—R3 is attached to the nitrogen atom of the ring.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-x):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-x1):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-x2):




embedded image


wherein Q represents —CHRy—, —O—, —C(═O)— or —NRq—; and wherein the other variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


The present invention relates in particular to compounds of Formula (T-x2) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—, —O—, —C(═O)— or —NRq—;


R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18; —C(═O)—O—C1-4alkyl-NR22aR22b; —C(═O)—O—C1-4alkyl;




embedded image


R18 represents C1-6alkyl or C3-6cycloalkyl;


R19 represents hydrogen or C1-6alkyl;


or R18 and R19 are taken together to form —(CH2)3—, —(CH2)4— or —(CH2)—;


Het represents a monocyclic 5- or 6-membered aromatic ring containing one, two or three O-, S- or N-atoms and optionally a carbonyl moiety; wherein said monocyclic 5- or 6-membered aromatic ring is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, C3-6cycloalkyl, or cyano;


Rxa and Rxb are each independently selected from the group consisting of hydrogen; Het3; C3-6cycloalkyl; and C1-6alkyl; wherein optionally said C3-6cycloalkyl and C1-6alkyl are substituted with 1, 2 or 3 substituents each independently selected from the group consisting of —OH, —OC1-4alkyl, —C1-4alkyl-OH, halo, CF3, C3-6cycloalkyl, Het3, and NR11cR11d;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, cyano, and C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo and OR23;


or Rxa and Rxb are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of C1-4alkyl, halo, —OH, —O—C1-4alkyl, cyano, and C1-4alkyl substituted with one, two or three substituents each independently selected from the group consisting of halo and OR23;


R23 represents hydrogen or C1-4alkyl optionally substituted with one, two or three halo;


R1b represents hydrogen, F, Cl, or —O—C1-4alkyl;


R2 represents halo, C3-6cycloalkyl, C1-4alkyl, —O—C1-4alkyl, cyano, or C1-4alkyl substituted with one, two or three halo substituents;


R21 represents hydrogen or —Ya—R3a;


Y and Ya each independently represent a covalent bond or




embedded image


n1 is selected from 1 and 2;


n2 is selected from 1, 2, 3 and 4;


Ry represents hydrogen, —OH, C1-4alkyl, —C1-4alkyl-OH, or —C1-4alkyl-O—C1-4alkyl;


Rq represents hydrogen or C1-4alkyl;


R5 represents hydrogen. C1-4alkyl, or C3-6cycloalkyl;


R3, R3a, and R4 are each independently selected from the group consisting of Het1; Het2; Cy2; C1-8alkyl; and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —C(═O)—NR10aR10b. —C(═O)—Het6a, —C(═O)—Het6b, —NR10c—C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, —NRxcRxd, —NR8aR8b, —CF3, cyano, halo, —OH, —O—C1-4alkyl, Het1, Het2, Ar1, and Cy2;


Rxc represents Cy1; Het5; —C1-6alkyl-Cy1; —C1-6alkyl-Het3; —C1-6alkyl-Het4; or —C1-6alkyl-phenyl;


Rxd represents hydrogen; C1-4alkyl; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and cyano;


or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —(C═O)—C1-4alkyl-S(═O)2—C1-4alkyl, and cyano;


R8a and R8b are each independently selected from the group consisting of hydrogen; C1-6alkyl; —(C═O)—C1-4alkyl; and C1-6alkyl substituted with one, two or three substituents each independently selected from the group consisting of —OH, cyano, halo, —S(═O)2—C1-4alkyl, —O—C1-4alkyl,


—C(═O)—NR10aR10b, and —NR10c—C(═O)—C1-4alkyl;


Ar1 represents phenyl optionally substituted with one, two or three substituents each independently selected from the group consisting of C1-4alkyl, halo, —O—C1-4alkyl, —CF3, —OH, —S(═O)2—C1-4alkyl, and —C(═O)—NR10aR10b;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6, —C(═O)—Cy1, and —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, R6, Het6a, Het6b, C1-4alkyl, oxo, —NR9aR9b and —OH;


Het2 represents C-linked pyrazolyl, 1,2,4-oxadiazolyl, pyridazinyl or triazolyl; which may be optionally substituted on one nitrogen atom with R6a,


R6 and R6a are each independently selected from the group consisting of Het3; Het4; —C(═O)—NH—Cy1; —C(═O)—NH—R8; —C(═O)—Het6a, —C(═O)—NR10dR10e; —C(═O)—O—C1-4alkyl; —S(═O)2—C1-4alkyl;


C1-6alkyl optionally substituted with one or two substituents each independently selected from the group consisting of Het3, Het4, Het6a, Het6b. Cy1, —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—N(C1-4alkyl)2, —C(O)—NH—C1-4alkyl-C3-6cycloalkyl, —C(═O)—OH, —NR11aR11b, and —NH—S(═O)2—C1-4alkyl; and


C3-6cycloalkyl optionally substituted by one or two substituents each independently selected from the group consisting of —CN, —OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl, —C(═O)—N(C1-4alkyl)2, —NH—S(═O)2—C1-4alkyl, and C1-4alkyl optionally substituted with one substituent selected from the group consisting of OH, —O—C1-4alkyl, —C(═O)—NH—C1-4alkyl and —NH—S(═O)2—C1-4alkyl;


R8 represents hydrogen, —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl, halo, cyano, —NR11aR11b, —S(═O)2—C1-4alkyl, Het3a, and Het6a;


Het3, Het3a, Het5 and Het5a each independently represent a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one carbon atom with C1-4alkyl, halo, —OH, —NR11aR11b, or oxo; and wherein said heterocyclyl is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—C1-4alkyl;


Het4 and Het7 each independently represent a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl or —(C═O)—O—C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —OH, halo, C1-4alkyl, —O—C1-4alkyl, —NR11aR11b, C1-4alkyl-NR11aR11b; —NH—C(═O)—C1-4alkyl, cyano, —COOH, —NH—C(═O)—O—C1-4alkyl, —NH—C(═O)—Cy3, —NH—C(═O)—NR10aR10b, —(C═O)—O—C1-4alkyl, —NH—S(═O)2—C1-4alkyl, Het8a, —C1-4alkyl-Het8a, Het8b, Het9, and —C(═O)—NR10aR10b:


Het6a, Het8 and Het8a each independently represent a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one, two, three or four substituents each independently selected from the group consisting of halo, —OH, oxo,


—NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, —(C═O)—NR10aR10b, —O—C3-6cycloalkyl, —S(═O)2—C1-4alkyl, cyano, C1-4alkyl, —C1-4alkyl-OH, —O—C1-4alkyl, —O—(C═O)—NR10aR10b, and —O—(C═O)—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —S(═O)2—C1-4alkyl, and —(C═O)—NR10aR10b.


Het6b and Het8b each independently represent a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of C1-4alkyl, —OH, oxo, —(C═O)—NR10aR10b, —NH—C(═O)—C1-4alkyl, —NH—C(═O)—Cy3, and —O—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of —C(═O)—C1-4alkyl, —C(═O)—Cy3, —(C═O)—C1-4alkyl-OH, —C(═O)—C1-4alkyl-O—C1-4alkyl, —C(═O)—C1-4alkyl-NR11aR11b, and C1-4alkyl;


Het9 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one nitrogen atom with C1-4alkyl; and wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two substituents each independently selected from the group consisting of —Oil, halo, and C1-4alkyl;


Cy1 represents C3-6cycloalkyl optionally substituted with one, two or three substituents selected from the group consisting of —OH, —NH—C(═O)—C1-4alkyl, C1-4alkyl, —NH—S(═O)2—C1-4alkyl, —S(═O)2—C1-4alkyl, and —O—C1-4alkyl;


Cy2 represents C3-7cycloalkyl or a 5- to 12-membered saturated carbobicyclic system; wherein said C3-7cycloalkyl or said carbobicyclic system is optionally substituted with one, two, three or four substituents each independently selected from the group consisting of halo, R6, —C(═O)—Het6a, Het6a, Het6b, —NR9aR9b, —OH, C1-4alkyl, —O—C1-4alkyl, cyano,




embedded image


and


C1-4alkyl substituted with one or two substituents each independently selected from the group consisting of Het3a, Het6a, Het6b, and —NR9aR9b;


Cy3 represents C3-7cycloalkyl; wherein said C3-7cycloalkyl is optionally substituted with one, two or three halo substituents;


R9a and R9b are each independently selected from the group consisting of hydrogen; C1-4alkyl; C3-6cycloalkyl; —C(═O)—C1-4alkyl; —C(═O)—C3-6cycloalkyl; —S(═O)2—C1-4alkyl; Het5; Het7; —C1-4alkyl-R16; —C(═O)—C1-4alkyl-Het3a; —C(═O)—R14;


C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano; and


C1-4alkyl substituted with one, two or three substituents selected from the group consisting of halo, —OH, —O—C1-4alkyl, —NR11aR11b, and cyano;


R11a, R11b, R13a, R13b, R15a, R15b, R17a, R17b, R20a, R20b, R22a, and R22b are each independently selected from the group consisting of hydrogen and C1-4alkyl;


R11c and R11d are each independently selected from the group consisting of hydrogen, C1-6alkyl, and —C(═O)—C1-4alkyl;


R10a, R10b and R10C are each independently selected from the group consisting of hydrogen, C1-4alkyl, and C3-6cycloalkyl;


R10d and R10e are each independently selected from the group consisting of C1-4alkyl, —O—C1-4alkyl and C3-6cycloalkyl;


R14 represents Het5a; Het7; Het8a; —O—C1-4alkyl; —C(═O)NR15aR15b; C3-6cycloalkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl and halo; or C1-4alkyl substituted with one, two or three substituents selected from the group consisting of —O—C1-4alkyl, —NR13aR13b, halo, cyano, —OH, Het8a, and Cy1;


R16 represents —C(═)—NR17aR17b, —S(═O)2—C1-4alkyl, Het5, Het7, or Het8;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I-x2) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Rxa and Rxb are C1-6alkyl optionally substituted with 1, 2 or 3 —OH;


R1b represents F;


R2 represents methyl;


R21 represents hydrogen or methyl;


Y represents a covalent bond or




embedded image


R5 represents hydrogen;


n1 is 1;


n2 is selected from 1 and 2;


Ry represents hydrogen;


R3 and R4 are each independently selected from Het1, Cy2, and C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1 and Cy;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one carbon atom with oxo;

    • R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one Het6a;


and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I-x2) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Rxa and Rxb are C1-6alkyl optionally substituted with 1, 2 or 3 —OH;


R1b represents F;


R2 represents methyl;


R21 represents hydrogen or methyl;


Y represents a covalent bond or




embedded image


R5 represents hydrogen;


n1 is 1;


n2 is selected from 1 and 2;


Ry represents hydrogen;


R3 and R4 are each independently selected from Het1, Cy2, and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1 and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one carbon atom with oxo;


R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one Het6a; and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I-x2) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Rxa and Rxb are C1-6alkyl optionally substituted with 1, 2 or 3 —OH;


R1b represents F;

    • R2 represents methyl;


R21 represents hydrogen or methyl;


Y represents a covalent bond;


n1 is 1;


n2 is selected from 1 and 2:


Ry represents hydrogen;


R3 is selected from C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1 and Cy;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)O; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one carbon atom with oxo;


R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to forn S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one Het6a; and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I-x2) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Rxa and Rxb are C1-6alkyl;


R1b represents F;


R2 represents methyl;


R21 represents hydrogen;


Y represents a covalent bond;


n1 is 1;


n2 is selected from 1 and 2;


Ry represents hydrogen;


R3 is selected from C1-8alkyl substituted with one substituent selected from the group consisting of —NRxccRxd, Het1 and Cy2;


Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8;


R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;


Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;


Cy2 represents C3-7cycloalkyl optionally substituted with one Het6a; and the pharmaceutically acceptable salts and the solvates thereof.


The present invention relates in particular to compounds of Formula (I-x2) as defined herein, and the tautomers and the stereoisomeric forms thereof, wherein


Q represents —CHRy—;


R1a represents —C(═O)—NRxaRxb;


Rxa and Rxb are C1-8alkyl;


R1b represents F;


R2 represents methyl;


R21 represents hydrogen;


Y represents a covalent bond;


n1 is 1;


n2 is selected from 1 and 2;


R7 represents hydrogen;


R3 is selected from C1-4alkyl substituted with one Het1;


Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8;


R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;


and the pharmaceutically acceptable salts and the solvates thereof.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-y):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-y1):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-z):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-z1):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to those compounds of Formula (I) and the pharmaceutically acceptable salts, and the solvates thereof, or any subgroup thereof as mentioned in any of the other embodiments, wherein the compounds of Formula (I) are restricted to compounds of Formula (I-q):




embedded image


wherein the variables are as defined for the compounds of Formula (I) or any subgroup thereof as mentioned in any of the other embodiments.


In an embodiment, the present invention relates to a subgroup of Formula (I) as defined in the general reaction schemes.


In an embodiment the compound of Formula (I) is selected from the group consisting of any of the exemplified compounds, tautomers and stereoisomeric forms thereof, and the free bases, any pharmaceutically acceptable salts, and the solvates thereof.


In an embodiment the compound of Formula (I) is selected from the group consisting of compounds 4, 8, 8a, 9a, 10, 12, 18a, 18b, 20, 27a, 27d, 32a, 34a, 38b, 43, 51, 51a, 59, 60, 115, 117a, 125, 140, 157, 159, 169a, 207, 228, 258, 262 and 365b.


In an embodiment the compound of Formula (I) is selected from the group consisting of compounds 4, 8, 8a, 9a, 10, 12, 18a, 18b, 20, 27a, 27d, 32a, 34a, 38b, 43, 51, 51a, 59, 60, 115, 117a, 125, 140, 157, 159, 169a, 207, 228, 258, 262 and 365b;


tautomers and stereoisomeric forms thereof,


and the free bases, any pharmaceutically acceptable salts, and the solvates thereof.


The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier or excipient, wherein the compound of Formula (I) is selected from the group consisting of any of the exemplified compounds.


The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier or excipient, wherein the compound of Formula (I) is selected from the group consisting of any of the exemplified compounds, tautomers and stereoisomeric forms thereof, and the free bases, any pharmaceutically acceptable salts, and the solvates thereof.


The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier or excipient, wherein the compound of Formula (I) is selected from the group consisting of compounds 4, 8, 8a, 9a, 10, 12, 18a, 18b, 20, 27a, 27d, 32a, 34a, 38b, 43, 51, 51a, 59, 60, 115, 117a, 125, 140, 157, 159, 169a, 207, 228, 258, 262 and 365b.


The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier or excipient, wherein the compound of Formula (I) is selected from the group consisting of compounds 4, 8, 8a, 9a, 10, 12, 18a, 18b, 20, 27a, 27d, 32a, 34a, 38b, 43, 51, 51a, 59, 60, 115, 117a, 125, 140, 157, 159, 169a, 207, 228, 258, 262 and 365b; tautomers and stereoisomeric forms thereof, and the free bases, any pharmaceutically acceptable salts, and the solvates thereof.


In a particular embodiment, the solvate is a hydrate. In a particular embodiment, the pharmaceutically acceptable salt is a HCl salt. In a particular embodiment, the compound is a HCl salt hydrate.


In an embodiment the compound of Formula (I) is




embedded image


or a pharmaceutically acceptable salt or solvate thereof; in particular a HCl salt, solvate; more in particular a HCl salt, hydrate; more in particular a mono HCl salt, hydrate; even more in particular mono HCl salt, trihydrate.


All possible combinations of the above indicated embodiments are considered to be embraced within the scope of the invention.


Any aspects of the invention and embodiments described herein for the compounds of formula (I) as listed herein, also hold for the compounds of formula (A).


In an embodiment the invention relates to any of the intermediates described herein, tautomers and stereoisomeric forms thereof, and the free bases, any pharmaceutically acceptable salts, and the solvates thereof.


Methods for the Preparation of Compounds of Formula (I)

In this section, as in all other sections unless the context indicates otherwise, references to Formula (I) also include all other sub-groups and examples thereof as defined herein.


The general preparation of some typical examples of the compounds of Formula (I) is described hereunder and in the specific examples, and are generally prepared from starting materials which are either commercially available or prepared by standard synthetic processes commonly used by those skilled in the art of organic chemistry. The following schemes are only meant to represent examples of the invention and are in no way meant to be a limit of the invention.


Alternatively, compounds of the present invention may also be prepared by analogous reaction protocols as described in the general schemes below, combined with standard synthetic processes commonly used by those skilled in the art.


The skilled person will realize that in the reactions described in the Schemes, although this is not always explicitly shown, it may be necessary to protect reactive functional groups (for example hydroxy, amino, or carboxy groups) where these are desired in the final product, to avoid their unwanted participation in the reactions. In general, conventional protecting groups (PG) can be used in accordance with standard practice. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.


The skilled person will realize that in the reactions described in the Schemes, it may be advisable or necessary to perform the reaction under an inert atmosphere, such as for example under N2-gas atmosphere.


It will be apparent for the skilled person that it may be necessary to cool the reaction mixture before reaction work-up (refers to the series of manipulations required to isolate and purify the product(s) of a chemical reaction such as for example quenching, column chromatography, extraction).


The skilled person will realize that heating the reaction mixture under stirring may enhance the reaction outcome. In some reactions microwave heating may be used instead of conventional heating to shorten the overall reaction time.


The skilled person will realize that another sequence of the chemical reactions shown in the Schemes below, may also result in the desired compound of Formula (I).


The skilled person will realize that intermediates and final compounds shown in the Schemes below may be further functionalized according to methods well-known by the person skilled in the art. The intermediates and compounds described herein can be isolated in free form or as a salt, or a solvate thereof. The intermediates and compounds described herein may be synthesized in the form of mixtures of tautomers and stereoisomeric forms that can be separated from one another following art-known resolution procedures.


General Synthetic Schemes

All abbreviations used in the general schemes are as defined below or as in the Table in the part Examples. Variables are as defined in the scope or as specifically defined in the general Schemes. Where compounds/intermediates in the schemes below contain a double bond, the substituents may be in the E or the Z configuration or be mixtures thereof.


In general, compounds of Formula (I-aa), can be prepared according to the following reaction Scheme 1. In Scheme 1, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl, 9-fluorenylmethoxycarbonyl, or benzyl, and LG is a leaving group such as for example chloro, bromo, iodo or tosylate or mesylate or triflate; all other variables are defined according to the scope of the present invention.




embedded image


In Scheme 1, the following reaction conditions apply:


Step 1: when PG=Boc, at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable acid, for example a protic acid such as trifluoroacetic acid (TFA) or hydrochloric acid, in a suitable solvent such as dichloromethane (DCM) or 1,4-dioxane;


Alternatively, when PG=9-fluorenylmethoxycarbonyl, at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable base such as piperidine, in a suitable solvent such as dichloromethane (DCM);


Alternatively, when PG=benzyl, at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, TIF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


Step 2:

In the case of a reductive amination reaction employing an aldehyde or a ketone: at a suitable temperature in a range between room temperature and 70° C., in the presence of a suitable reducing agent such as for example sodium triacetoxyborohydride or sodium cyanoborohydride, in a suitable solvent such as for example methanol, dichloromethane or 1,2-dichloroethane, optionally in the presence of zinc chloride or sodium acetate or acetic acid;


In the case of an alkylation reaction employing LG-Y—R3: at a suitable temperature such as for example room temperature, in the presence of a suitable deprotonating agent such as for example sodium hydride or potassium carbonate, or an amine base such as triethylamine in a suitable aprotic solvent such as for example dimethylformamide or dimethylsulfoxide or acetonitrile.


In general, compounds of Formula (I) wherein Q is limited to —O—, —NRq—, can be prepared via intermediates of Formula (VIc). Intermediates of Formula (VIc) can be prepared according to the following reaction Scheme 2. In Scheme 2, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl; all other variables are defined according to the scope of the present invention.




embedded image


In Scheme 2, the following reaction conditions apply:


Step 1: at a suitable temperature in a range between 100° C. and 140° C., in the presence of a suitable base such as for example potassium tert-butoxide or potassium phosphate, in the presence of a suitable catalyst such as palladium acetate (Pd(OAc)2) or tris(dibenzylideneacetone)dipalladium(0) (Pd2dba3) or [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (Pd(dppf)Cl2), in the presence of a suitable ligand such as 9,9-dimethyl-4,5-bis(diphenylphosphino)xanthene (Xantphos), in a suitable solvent such as for example dioxane or dimethylformamide.


In general, intermediates of Formula (V) can be prepared according to the following reaction Scheme 2B. In Scheme 2B, W1 represents fluoro, chloro, bromo or iodo, BPin represents 4,4,5,5-tetramethyl-1,3,2-dioxaborolane, and all other variables are defined according to the scope of the present invention.




embedded image


In Scheme 2B, the following reaction conditions apply:


Step 1: at a suitable temperature in a range between room temperature and 100° C., in the presence of a suitable base such as for example potassium carbonate, in the presence of a suitable catalyst such as [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water; Alternatively, when R2=Me, a boron containing reagent such as trimethyl boroxine can be used in the presence of a suitable catalyst such as (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water in the presence of an inorganic base such as potassium carbonate at a reaction temperature between 80° C. and 120° C.;


Additional step to achieve the double bond reduction to obtain R2 is C3-6cycloalkyl, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents: at a suitable temperature such as room temperature, in the presence of a suitable catalyst such as palladium on charcoal (Pd/C), in a suitable solvent such as methanol, under H2 pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


Step 2: at a suitable temperature such as for example between 0° C. and room temperature, in the presence of a suitable bronination reagent such as for example N-Bromosuccinimide or CuBr2, in a suitable solvent such as for example dimethylformamide or acetonitrile;


Step 3: at a suitable temperature such as for example 80° C. and 130° C., in the presence of a suitable catalyst such as copper (Cu), in the presence of a base such as potassium carbonate, in a suitable solvent such as dimethylformamide; Alternatively a copper (I) source may be used, such as CuI in the presence of a suitable diamine ligand, such as trans-N,N′-dimethylcyclohexane-1,2-diamine in the presence of an inorganic base, such as potassium carbonate in an aprotic solvent such as dimethylformamide at a temperature between 80° C. and 150° C. In certain cases said conversion may also be effected by a nucleophilic aromatic substitution using an inorganic base such as potassium tert-butoxide or sodium hydride or the like, in an aprotic solvent such as dimethylformamide at a temperature between 0° C. and 80° C.;


Someone skilled in the art will appreciate that the steps 2 and 3 in Scheme 2B may also be reversed, i.e. first the cross coupling of intermediate (III) with the reagent (Va), followed by bromination of the aza indole moiety to provide the intermediate (V).


In general, intermediates of Formula (VIIa), can be prepared via intermediates of Formula (VIe). Intermediates of Formula (VIe) can also be prepared according to the following reaction Scheme 3. In Scheme 3, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl; all other variables are defined according to the scope of the present invention.




embedded image


In Scheme 3, the following reaction conditions apply:


Step 1: at a suitable temperature in a range between 70° C. and 100° C., in the presence of a suitable base such as for example potassium phosphate, in presence of a suitable catalyst such as palladium acetate (Pd(OAc)2), optionally in the presence of a suitable phosphine ligand such as 2-dicyclohexylphosphino-2′-(N,N-dimethylamino)biphenyl (Davephos), in a suitable solvent such as for example dioxane or dimethylformamide;


Step 2: at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, THF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


In general, intermediates of Formula (IXb) can be prepared according to the following reaction Scheme 4. In Scheme 4 PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl; all other variables are defined according to the scope of the present invention.




embedded image


Step 1: at a suitable temperature such as for example −78° C., in the presence of a suitable deprotonating agent such as for example n-Butyllithium, in presence of a suitable reagent such as 2,2,6,6-Tetramethylpiperidine (HTMP), in a suitable solvent such as tetrahydrofuran;


In general, compounds of Formula (I-a), can be prepared according to the following reaction Scheme 5. In Scheme 5, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl and LG is a leaving group such as for example chloro, bromo, iodo or tosylate or mesylate or triflate; all other variables are defined according to the scope of the present invention.




embedded image


embedded image


In Scheme 5, the following reaction conditions apply:


Step 1: at a suitable temperature such as for example 100° C., in the presence of a suitable catalyst such as copper (Cu), in the presence of a base such as potassium carbonate, in a suitable solvent such as dimethylformamide; Alternatively a copper (I) source may be used, such as CuI in the presence of a suitable diamine ligand, such as trans-N,N′-dimethylcyclohexane-1,2-diamine, in the presence of an inorganic base, such as potassium carbonate in an aprotic solvent such as dimethylformamide at a temperature between 80° C. and 150° C.;


Step 2: at a suitable temperature such as for example room temperature, in the presence of a suitable condensation reagent such as 2-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU), in the presence of a base such as N,N-Diisopropylethylamine (DIPEA), in a suitable solvent such as dimethylformamide; Alternatively the acid chloride may be prepared by reacting intermediate VIII with thionyl chloride optionally in a halogenated solvent such as dichloromethane at a temperature in a range between 0° C. and room temperature. The intermediate acid chloride may then be reacted with the amine HNRxaRxb optionally in an aprotic solvent such as dimethylformamide and optionally in the presence of a tertiary amine such as N,N-diisopropylethylamine;


Step 3: at a suitable temperature in a range between 60° C. and 120° C., such as for example 100° C., in the presence of a suitable base such as for example potassium carbonate, in presence of a suitable catalyst such as [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water;


Step 4: at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, THF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


Step 5: at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable acid, for example a protic acid such as trifluoroacetic acid (TFA) or hydrochloric acid, in a suitable solvent such as dichloromethane (DCM) or 1,4-dioxane; Step 6:


In the case of a reductive amination reaction employing an aldehyde or a ketone: at a suitable temperature in a range between room temperature and 70° C., in the presence of a suitable reducing agent such as for example sodium triacetoxyborohydride or sodium cyanoborohydride, in a suitable solvent such as for example methanol, dichloromethane or 1,2-dichloethane, optionally in the presence of zinc chloride or sodium acetate or acetic acid;


In the case of an alkylation reaction employing LG-Y—R3: at a suitable temperature such as for example room temperature, in the presence of a suitable deprotonating agent such as for example sodium hydride or potassium carbonate, or an amine base such as triethylamine in a suitable aprotic solvent such as for example dimethylformamide or dimethylsulfoxide or acetonitrile.


In general, compounds of Formula (I-b) wherein R1a is limited to —S(═O)2—R18,




embedded image


and Q represents —CHRy—, can be prepared according to the following reaction Scheme 6. In Scheme 6, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl, 9-fluorenylmethoxycarbonyl, or benzyl; LG is a leaving group such as for example chloro, bromo, iodo or tosylate or mesylate; LGI is a leaving group such as for example fluoro, chloro, bromo, iodo or tosylate or mesylate; all other variables are defined according to the scope of the present invention.




embedded image


embedded image


In Scheme 6, the following reaction conditions apply:


Step 1: at a suitable temperature in a range between 50° C. and 90° C., in the presence of a suitable base such as for example potassium hydroxide or sodium hydroxide, in a suitable solvent, preferably a protic solvent, such as methanol, ethanol or isopropanol.


Step 2: at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, THF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


Step 3: at a suitable temperature in a range between 50° C. and 100° C., in the presence of a suitable inorganic base such as for example potassium carbonate or potassium tert-butoxide, in a suitable aprotic solvent such as for example dioxane, dimethylformamide or acetonitrile or dimethylsulfoxide;


Step 4: when PG=Boc, at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable acid, for example a protic acid such as trifluoroacetic acid or hydrochloric acid, in a suitable solvent such as dichloromethane or 1,4-dioxane;


Alternatively, when PG=9-Fluorenylmethoxycarbonyl, at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable base such as piperidine, in a suitable solvent such as dichloromethane (DCM);


Alternatively, when PG=benzyl, at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, THF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


Step 5: In the case of a reductive amination reaction employing an aldehyde or a ketone: at a suitable temperature in a range between room temperature and 70° C., in the presence of a suitable reducing agent such as for example sodium triacetoxyborohydride or sodium cyanoborohydride, in a suitable solvent such as for example methanol, dichloromethane or 1,2-dichloroethane, optionally in the presence of zinc chloride or sodium acetate or acetic acid;


In the case of an alkylation reaction employing LG-Y—R3: at a suitable temperature such as for example room temperature, in the presence of a suitable deprotonating agent such as for example sodium hydride or potassium carbonate, or an amine base such as triethylamine in a suitable aprotic solvent such as for example dimethylformamide or dimethylsulfoxide or acetonitrile;


In general, compounds of Formula (I-c), can be prepared according to the following reaction Scheme 7. In Scheme 7, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl, 9-fluorenylmethoxycarbonyl, or benzyl, all other variables are defined according to the scope of the present invention.




embedded image


embedded image


In Scheme 7, the following reaction conditions apply:


Step 1: at a suitable temperature such as for example −78° C., in the presence of a suitable deprotonating agent such as for example lithium bis(trimethylsilyl)amide (LiHMDS) and sodium hydride, in a suitable solvent such as for example tetrahydrofuran;


Step 2: at a suitable temperature in a range between room temperature and 100° C., in the presence of a suitable catalyst such as for example rhodium acetate dimer (Rh2(OAc)4), in a suitable solvent such as for example dichloromethane;


Step 3: at a suitable temperature in a range between room temperature and 100° C., in the presence of a suitable catalyst such as for example tetrakis(triphenylphosphine)palladium (Pd(PPh3)4), in the presence of a suitable base such as for example morpholine, in a suitable solvent such as for example tetrahydrofuran;


Step 4: at a suitable temperature such as for example −78° C., in the presence of a suitable deprotonating agent such as for example n-Butyllithium, in presence of a suitable reagent such as 2,2,6,6-Tetramethylpiperidine (HTMP), in a suitable solvent such as tetrahydrofuran;


Step 5: at a suitable temperature such as for example 100° C., in the presence of a suitable base such as for example potassium carbonate, in presence of a suitable catalyst such as [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water;


Step 6: at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, THF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine;


Step 7: when PG=Boc, at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable acid, for example a protic acid such as trifluoroacetic acid or hydrochloric acid, in a suitable solvent such as dichloromethane or 1,4-dioxane;


Alternatively, when PG=9-fluorenylmethoxycarbonyl, at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable base such as piperidine, in a suitable solvent such as dichloromethane (DCM);


Alternatively, when PG=benzyl, at a suitable temperature such as room temperature, in the presence of a suitable heterogenous catalyst such as palladium on charcoal (Pd/C), in a common solvent such as methanol, ethanol, THF or the like under hydrogen pressure such as for example from 1 to 3 bar, optionally in the presence of a base such as triethylamine.


An example of steps 1 and 2 in Scheme 7 is the preparation of a 5-membered intermediate (XVIIcc), as shown in Scheme 7a which can be prepared according to the general procedures outlined in steps 1 and 2 in Scheme 7.




embedded image


In general, intermediates of Formula (XVIIId), can be prepared according to the following reaction Scheme 8. In Scheme 8, W2 represents chloro, bromo or iodo, all other variables are defined according to the scope of the present invention. A skilled person will realize that cyclobutyl in Scheme 8 can be C3-5cycloalkyl in general, and that an intermediate of Formula (XVIIId) can be further functionalized into a compound of Formula (I) by analogous reaction protocols as described in the general schemes herein, combined with standard synthetic processes commonly used by those skilled in the art of organic chemistry.




embedded image


In Scheme 8, the following reaction conditions apply:


Step 1: at a suitable temperature such as for example 0° C., in the presence of a suitable condensation reagent such as propylphosphonic anhydride (T3P), in the presence of a base such as N,N-Diisopropylethylamine (DIPEA), in a suitable solvent such as dimethylformamide or dichloromethane;


Step 2: at a suitable temperature such as for example 0° C., in a suitable solvent such as tetrahydrofuran;


Step 3: at a suitable temperature in a range between room temperature and 70° C., in the presence of a suitable reducing agent such as for example sodium triacetoxyborohydride or sodium cyanoborohydride, in a suitable solvent such as for example methanol, dichloromethane or 1,2-dichloroethane, optionally in the presence of zinc chloride or sodium acetate or acetic acid;


Step 4: at a suitable temperature in a range between 0° C. and 40° C., such as room temperature, in the presence of a suitable acid such as hydrochloric acid (HCl, 1N), in a suitable solvent such as acetonitrile.


In general, intermediates as described in Scheme 9, wherein Q represents —CHRy—, can be prepared according to the following reaction Scheme. In Scheme 9, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl, all other variables are defined according to the scope of the present invention.




embedded image


In Scheme 9, the following reaction conditions apply:


Step 1: at a suitable temperature, in a range between room temperature and 70° C., such as 60° C., in the presence of zinc, in the presence of suitable activating agents such as trimethylsilylchloride or 1-bromo, 2-chloroethane, in a suitable solvent such as tetrahydrofuran. Optionally, the procedure can also be performed with the use of a flow-apparatus;


Step 2: at a suitable temperature, in a range between room temperature and 70° C., such as 50° C., in the presence of a suitable catalyst such as 4th generation RuPhos Pd precatalyst (RuPhos Pd G4), in a suitable solvent such as tetrahydrofuran.


In general, intermediates as described in Scheme 10, wherein Q represents —CHRy—, can be prepared according to the following reaction Scheme. In Scheme 10, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl, all other variables are defined according to the scope of the present invention. BPin represents 4,4,5,5-tetramethyl-1,3,2-dioxaborolane. W1 and W3 represent fluoro, chloro, bromo or iodo.




embedded image


embedded image


Step 1: at a suitable temperature, such as −78° C., in the presence of a suitable deprotonating agent such as n-Butyllithium, in a suitable solvent such as tetrahydrofuran, in the presence of suitable electrophile, such as DMF;


Step 2: at a suitable temperature in a range between 80° C. and 120° C., in the presence of a diol protection reagent such as for example glycol, in the presence of a Bronsted acid such as for example para-toluenesulfonic acid in a suitable solvent such as for example toluene;


Step 3: at a suitable temperature in a range between room temperature and 100° C., in the presence of a suitable base such as for example potassium carbonate, in the presence of a suitable catalyst such as [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water; Alternatively, when R2=Me, a boron containing reagent such as trimethyl boroxine can be used in the presence of a suitable catalyst such as (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water in the presence of an inorganic base such as potassium carbonate at a reaction temperature between 80° C. and 120° C.; Step 4: in the presence of a suitable base, such as for example sodium t-butoxide, in the presence of a suitable palladium source such as palladium(II)acetate (Pd(OAc)2), in the presence of a suitable ligand, such as 1,1′-(9,9-Dimethyl-9H-xanthene-4,5-diyl)bis[1,1-diphenylphosphine], Xantphos, in the presence of a suitable solvent, such as 1,4-dioxane, at a suitable temperature range between 50° C. and 120° C.;


Step 5: in the presence of a suitable Bronsted acid, such as hydrochloric acid, in the presence of a suitable solvent such as 1,4-dioxane or tetrahydrofuran, and water, at a suitable temperature range such a room temperature and 60° C.;


Step 6: in the presence of a suitable deprotonating agent, such as n-butyllithium, in a suitable solvent such as tetrahydrofuran, at a suitable temperature range such as −78° C. and room temperature.


Step 7: in the presence of a suitable Bronsted acid, such as hydrochloric acid, in the presence of a suitable solvent such as 1,4-dioxane or tetrahydrofuran, and water, at a suitable temperature range such a room temperature and 100° C.;


Step 8: at a suitable temperature such as for example between 0° C. and room temperature, in the presence of a suitable bromination reagent such as for example N-bromosuccinimide or CuBr2, in a suitable solvent such as for example dimethylformamide or acetonitrile;


Step 9: at a suitable temperature such as for example 80° C. and 130° C., in the presence of a suitable catalyst such as copper (Cu), in the presence of a base such as potassium carbonate, in a suitable solvent such as dimethylformamide. Alternatively a copper (I) source may be used, such as CuI in the presence of a suitable diamine ligand, such as trans-N,N′-dimethylcyclohexane-1,2-diamine in the presence of an inorganic base, such as potassium carbonate in an aprotic solvent such as dimethylformamide at a temperature between 80° C. and 150° C. In certain cases said conversion may also be effected by a nucleophilic aromatic substitution using an inorganic base such as potassium tert-butoxide or sodium hydride or the like, in an aprotic solvent such as dimethylformamide at a temperature between 0° C. and 80° C.;


Step 10: at a suitable temperature such as for example 100° C., in the presence of a suitable base such as for example potassium carbonate, in presence of a suitable catalyst such as [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (Pd(dppf)Cl2) in a suitable solvent such as for example dioxane or dimethylformamide and water.


As can be appreciated by a person skilled in the art, the intermediate obtained in scheme 10, can be further elaborated to obtain compounds of Formula (A) by means of using the procedures outlined in the general schemes mentioned above, in particular in scheme 1 and scheme 3.


In general, intermediates as described in Scheme 11, can be prepared according to the following reaction Scheme. In Scheme 11, PG represents a suitable protecting group, such as for example tert-butyloxycarbonyl, all other variables are defined according to the scope of the present invention. BPin represents 4,4,5,5-tetramethyl-1,3,2-dioxaborolane.




embedded image


Step 1: when PG″=a silyl containing protecting group, such as tert-butyldimethylsilyl, at a suitable temperature in a range between room temperature and 80° C., such as room temperature, in the presence of a base, such as imidazole, in the presence of a suitable reagent, such as tert-butyldimethylsilylchloride, in a suitable solvent, such as DMF. When, PG″ is a different protecting group as defined herein, general protection conditions may be used, known to those skilled in the art.


Step 2: at a suitable temperature, between room temperature and 60° C., such as room temperature, for example in the presence of a suitable alkyl halide, in the presence of a suitable base, such as K2CO3, in the presence of a suitable photocatalyst, such as [4,4′-Bis(1,1-dimethylethyl)-2,2′-bipyridine-N1,N1′]bis[3,5-difluoro-2-[5-(trifluoromethyl)-2-pyridinyl-N]phenyl-C]Iridium(III) hexafluorophosphate, [Ir{dF(CF3)ppy}2(dtbpy)]PF6, in the presence of suitable nickel salt, such as NiCl2-glyme, in the presence of a suitable ligand, such as 4-4′-dimethoxy-2-2′-bipyridine, in a suitable solvent, such as acetonitrile and in the presence of water as an additive, employing blue LED irradiation (Johnston, C., Smith, R., Allmendinger, S. et al. Metallaphotoredox-catalysed sp3-sp3 cross-coupling of carboxylic acids with alkyl halides. Nature 536, 322-325 (2016)).


Step 3: at a suitable temperature, such as room temperature, in the presence of a suitable fluoride source, such a tetrabutylammonium fluoride, in a suitable solvent, such as tetrahydrofuran. When, PG is a different protecting group as defined herein, general protection conditions may be used, known to those skilled in the art.


Step 4: at a suitable temperature, such as between −78° C. and 40° C., in the presence of Dess-Martin periodinane, in a suitable solvent such as dichloromethane. Other oxidation methods, known to those skilled in the art may also be employed.


Step 5: at a suitable temperature such as for example −78° C., in the presence of a suitable deprotonating agent such as for example n-Butyllithium, in presence of a suitable reagent such as 2,2,6,6-Tetramethylpiperidine (HTMP), in a suitable solvent such as tetrahydrofuran.


It will be appreciated that where appropriate functional groups exist, compounds of various formulae or any intermediates used in their preparation may be further derivatized by one or more standard synthetic methods employing condensation, substitution, oxidation, reduction, or cleavage reactions. Particular substitution approaches include conventional alkylation, arylation, heteroarylation, acylation, sulfonylation, halogenation, nitration, formylation and coupling procedures.


The compounds of Formula (I) may be synthesized in the form of racemic mixtures of enantiomers which can be separated from one another following art-known resolution procedures. The racemic compounds of Formula (I) containing a basic nitrogen atom may be converted into the corresponding diastereomeric salt forms by reaction with a suitable chiral acid. Said diastereomeric salt forms are subsequently separated, for example, by selective or fractional crystallization and the enantiomers are liberated therefrom by alkali. An alternative manner of separating the enantiomeric forms of the compounds of Formula (I) involves liquid chromatography using a chiral stationary phase. Said pure stereochemically isomeric forms may also be derived from the corresponding pure stereochemically isomeric forms of the appropriate starting materials, provided that the reaction occurs stereospecifically.


In the preparation of compounds of the present invention, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t-butoxycarbonyl (Boc), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 4th ed., Wiley, Hoboken, N.J., 2007.


Pharmacology

It has been found that the compounds of the present invention block the interaction of menin with MLL proteins and oncogenic MLL fusion proteins per se, or can undergo metabolism to a (more) active form in vivo (prodrugs). Therefore the compounds according to the present invention and the pharmaceutical compositions comprising such compounds may be useful for the treatment or prevention, in particular treatment, of diseases such as cancer, including but not limited to leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN); and diabetes.


In particular, the compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of cancer. According to one embodiment, cancers that may benefit from a treatment with menin/MLL inhibitors of the invention comprise leukemias, lymphomas, myelomas or solid tumor cancers (e.g. prostate cancer, lung cancer, breast cancer, pancreatic cancer, colon cancer, liver cancer, melanoma and glioblastoma, etc.). In some embodiments, the leukemias include acute leukemias, chronic leukemias, myeloid leukemias, myelogeneous leukemias, lymphoblastic leukemias, lymphocytic leukemias, Acute myelogeneous leukemias (AML), Chronic myelogenous leukemias (CML), Acute lymphoblastic leukemias (ALL), Chronic lymphocytic leukemias (CLL), T cell prolymphocytic leukemias (T-PLL), Large granular lymphocytic leukemia, Hairy cell leukemia (HCL), MLL-rearranged leukemias, MLL-PTD leukemias, MLL amplified leukemias, MLL-positive leukemias, leukemias exhibiting HOX/MEIS1 gene expression signatures etc.


In particular, the compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of myelodysplastic syndrome (MDS) or myeloproliferative neoplasms (MPN).


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of leukemias, in particular nucleophosmin (NPM1)-mutated leukemias, e.g. NPM1c.


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of AML, in particular nucleophosmin (NPMT)-mutated AML (i.e., NPM1mut AML), more in particular abstract NPM1-mutated AML.


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of MLL-rearranged leukemias, in particular MLL-rearranged AML or ALL.


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of leukemias with MLL gene alterations, in particular AML or ALL with MLL gene alterations.


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be suitable for Q.D. dosing (once daily).


In particular, compounds according to the present invention and the pharmaceutical compositions thereof may be useful in the treatment or prevention of hematological cancer in a subject exhibiting NPM1 gene mutations and/or mixed lineage leukemia gene (MLL; MLL1; KMT2A) alterations, mixed lineage leukemia (MLL), MLL-related leukemia, MLL-associated leukemia, MLL-positive leukemia, MLL-induced leukemia, rearranged mixed lineage leukemia, leukemia associated with a MLL, rearrangement/alteration or a rearrangement/alteration of the MLL gene, acute leukemia, chronic leukemia, myelodysplastic syndrome (MDS), myeloproliferative neoplasms (MPN), insulin resistance, pre-diabetes, diabetes, or risk of diabetes, hyperglycemia, chromosomal rearrangement on chromosome 11q23, type-1 diabetes, type-2 diabetes; promoting proliferation of a pancreatic cell, where pancreatic cell is an islet cell, beta cell, the beta cell proliferation is evidenced by an increase in beta cell production or insulin production; and for inhibiting a menin-MLL interaction, where the MLL fusion protein target gene is HOX or MEIS1 in human.


Hence, the invention relates to compounds of Formula (I), the tautomers and the stereoisomeric forms thereof, and the pharmaceutically acceptable salts, and the solvates thereof, for use as a medicament.


The invention also relates to the use of a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, or a pharmaceutical composition according to the invention, for the manufacture of a medicament.


The present invention also relates to a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, or a pharmaceutical composition according to the invention, for use in the treatment, prevention, amelioration, control or reduction of the risk of disorders associated with the interaction of menin with MLL proteins and oncogenic MLL fusion proteins in a mammal, including a human, the treatment or prevention of which is affected or facilitated by blocking the interaction of menin with MLL proteins and oncogenic MLL fusion proteins.


Also, the present invention relates to the use of a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, or a pharmaceutical composition according to the invention, for the manufacture of a medicament for treating, preventing, ameliorating, controlling or reducing the risk of disorders associated with the interaction of menin with MLL proteins and oncogenic MLL fusion proteins in a mammal, including a human, the treatment or prevention of which is affected or facilitated by blocking the interaction of menin with MLL proteins and oncogenic MLL fusion proteins.


The invention also relates to a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, for use in the treatment or prevention of any one of the diseases mentioned hereinbefore.


The invention also relates to a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, for use in treating or preventing any one of the diseases mentioned hereinbefore.


The invention also relates to the use of a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, for the manufacture of a medicament for the treatment or prevention of any one of the disease conditions mentioned hereinbefore.


The compounds of the present invention can be administered to mammals, preferably humans, for the treatment or prevention of any one of the diseases mentioned hereinbefore.


In view of the utility of the compounds of Formula (I), the tautomers and the stereoisomeric forms thereof, and the pharmaceutically acceptable salts, and the solvates thereof, there is provided a method of treating warm-blooded animals, including humans, suffering from any one of the diseases mentioned hereinbefore.


Said method comprises the administration, i.e. the systemic or topical administration, of a therapeutically effective amount of a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, to warm-blooded animals, including humans.


Therefore, the invention also relates to a method for the treatment or prevention of any one of the diseases mentioned hereinbefore comprising administering a therapeutically effective amount of compound according to the invention to a patient in need thereof.


One skilled in the art will recognize that a therapeutically effective amount of the compounds of the present invention is the amount sufficient to have therapeutic activity and that this amount varies inter alias, depending on the type of disease, the concentration of the compound in the therapeutic formulation, and the condition of the patient. An effective therapeutic daily amount would be from about 0.005 mg/kg to 100 mg/kg. The amount of a compound according to the present invention, also referred to herein as the active ingredient, which is required to achieve a therapeutically effect may vary on case-by-case basis, for example with the particular compound, the route of administration, the age and condition of the recipient, and the particular disorder or disease being treated. A method of treatment may also include administering the active ingredient on a regimen of between one and four intakes per day. In these methods of treatment the compounds according to the invention are preferably formulated prior to administration.


The present invention also provides compositions for preventing or treating the disorders referred to herein. Said compositions comprising a therapeutically effective amount of a compound of Formula (I), a tautomer or a stereoisomeric form thereof, or a pharmaceutically acceptable salt, or a solvate thereof, and a pharmaceutically acceptable carrier or diluent.


While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical composition. Accordingly, the present invention further provides a pharmaceutical composition comprising a compound according to the present invention, together with a pharmaceutically acceptable carrier or diluent. The carrier or diluent must be “acceptable” in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.


The pharmaceutical compositions may be prepared by any methods well known in the art of pharmacy, for example, using methods such as those described in Gennaro et al. Remington's Pharmaceutical Sciences (18thed., Mack Publishing Company, 1990, see especially Part 8 Pharmaceutical preparations and their Manufacture).


The compounds of the present invention may be administered alone or in combination with one or more additional therapeutic agents. Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound according to the present invention and one or more additional therapeutic agents, as well as administration of the compound according to the present invention and each additional therapeutic agent in its own separate pharmaceutical dosage formulation.


Therefore, an embodiment of the present invention relates to a product containing as first active ingredient a compound according to the invention and as further active ingredient one or more anticancer agent, as a combined preparation for simultaneous, separate or sequential use in the treatment of patients suffering from cancer.


The one or more other medicinal agents and the compound according to the present invention may be administered simultaneously (e.g. in separate or unitary compositions) or sequentially in either order. In the latter case, the two or more compounds will be administered within a period and in an amount and manner that is sufficient to ensure that an advantageous or synergistic effect is achieved. It will be appreciated that the preferred method and order of administration and the respective dosage amounts and regimes for each component of the combination will depend on the particular other medicinal agent and compound of the present invention being administered, their route of administration, the particular condition, in particular tumour, being treated and the particular host being treated.


The following examples further illustrate the present invention.


Examples

Several methods for preparing the compounds of this invention are illustrated in the following examples. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification, or alternatively can be synthesized by a skilled person by using well-known methods.













Abbreviation
Meaning







CH3COONH4
ammonium acetate


1,2-DCE
1,2-dichloroethane


sat. or Sat.
saturated


° C.
degree Celsius


AcOH or CH3COOH
acetic acid


Ac2O
acetic anhydride


aq.
aqueous


atm
atmosphere


BH3•THF
Borane tetrahydrofuran complex


Boc or boc
tert-butyloxycarbonyl


BOC-anhydride
di-tert-butyl dicarbonate


BPin or PinB
4,4,5,5-tetramethyl-1,3,2-dioxaborolane


Celite
diatomaceous earth


CO2
carbon dioxide


Cs2CO3
cesium carbonate


CPME
cyclopentyl methylether


DCM or CH2Cl2
dichloromethane


DEA
diethanolamine


DEE
diethyl ether


DIEA or DIPEA
N-ethyl-N-(propan-2-yl)propan-2-amine


DMA
N,N-dimethylacetamide


DME
1,2-dimethoxyethane


DMF
N,N-dimethylformamide


DMSO
(methanesulfinyl)methane


DSC
differential scanning calorimetry


EDCI or EDCI•HCl
3-{[(ethylimino)methylidene]amino}-N,N-



dimethylpropan-1-amine


ee
enantiomeric excess


ESI
electrospray ionization


EtOAc or EA
ethyl acetate


EtOH
ethanol


FA
formic acid


FCC
flash column chromatography


h or hr
hour(s)


HATU
1-[bis(dimethylamino)methylene]-1H-



1,2,3-triazolo[4,5-b]pyridinium 3-oxide



hexafluorophosphate


HCl
hydrochloric acid


Hex
hexane


HOBt
1-hydroxybenzotriazole


IBX
2-iodoxybenzoic acid


Insolute ® SCX-3
ethylbenzene sulfonic acid cation



exchange resin


Ir[dF(CF3)ppy]2(dtbpy))PF6
[4,4′-Bis(1,1-dimethylethyl)-2,2′-



bipyridine-N1,N1′]bis[3,5-difluoro-



2-[5-(trifluoromethyl)-2-



pyridinyl-N]phenyl-C]Iridium(III)



hexafluorophosphate


i-PrNH2 or iPrNH
isopropyl amine


i-PrOH, iPrOH or IPA
isopropyl alcohol


IPAC
isopropyl acetate


K2CO3
potassium carbonate


KOAc
potassium acetate


LCMS
liquid chromatography-mass spectrometry


LiAlH4
lithium aluminium hydride


Li-HMDS or LiHMDS
lithium bis(trimethylsilyl)amide


M or N
mol/L


MeCN or CH3CN
acetonitrile


MeI
iodomethane


MeOH
methanol


mg
milligram


min
minute(s)


mL
milliliter


mmol
millimole


m-CPBA
meta-chloroperoxybenzoic acid


MS
mass spectrometry


N2
nitrogen


Na2CO3
sodium carbonate


Na2SO4
sodium sulfate


NaBH(OAc)3
sodium triacetoxyborohydride


NaBH3CN
sodium cyanoborohydride


Na4EDTA
Ethylenediaminetetraacetic acid



tetrasodium salt


NaHCO3
sodium hydrogencarbonate


NaOAc
sodium acetate


NaOH
sodium hydroxide


NH3
ammonia


NH3•H2O or NH4OH
ammonium hydroxide


NH4HCO3
ammonium hydrogencarbonate


n-BuLi
n-butyllithium


NBS
N-Bromosuccinimide


NH4Cl
ammonium chloride


NMP
1-methyl-2-pyrrolidinone


NMR
nuclear magnetic resonance


Pd(dppf)Cl2
[1,1′-



bis(diphenylphosphino)ferrocene]



dichloropalladium(II)


Pd(PPh3)4
tetrakis(triphenylphosphine)palladium(0)


Pd/C
palladium on carbon


PE
petroleum ether


PIDA
Diacetoxy iodobenze or iodobenzen



diacetate


Prep. HPLC
preparative high-performance liquid



chromatography


Prep. SFC
preparative supercritical fluid



chromatography


Prep. TLC
preparative thin-layer chromatography


Rf
retention factor


Rh2(OAc)4
rhodium acetate


RP
Reverse(d) phase


rt, r.t. or RT
room temperature


RuPhos Pd G4/
Palladium, [[2′,6′-bis(1-methylethoxy)[1,1′-


4th generation RuPhos Pd
biphenyl]-2-yl]dicyclohexylphosphine-


precatalyst
κP](methanesulfonato-κO)[2′-(methyl-



amino-κN)[1,1′-biphenyl]-2-yl-κC]-,



(SP-4-3)-(ACI)



CAS 1599466-85-9


sat
saturated


SFC
supercritical fluid chromatography


SiliaBond ®
Propylsulfonic acid bound to silica


propylsulfonic
stationary phase support


acid resin



SiliaMetS ®
N1-propylethane-1,2-diamine bound to


Diamine
silica stationary phase support


t-butyl
tert-butyl


t-BuOK
potassium tert-butoxide


T3P
propylphosphonic anhydride


TEA or Et3N
triethylamine


Temp
temperature


TFA
trifluo acetic acid


THF
tetrahydrofuran


TLC
thin-layer chromatography


Tonset
Temperature at which melting onset occurs



(measure by DSC)


Ts
tosyl


UV
ultraviolet


v/v
volume to volume


w/v
weight to volume


w/w
weight to weight


ZnCl2
zinc chloride


Xantphos
4,5-bis(diphenylphosphino)-9,9-



dimethylxanthene









As understood by a person skilled in the art, compounds synthesized using the protocols as indicated may exist as a solvate e.g. hydrate, and/or contain residual solvent or minor impurities. Compounds or intermediates isolated as a salt form, may be integer stoichiomnetric i.e. mono- or di-salts, or of intermediate stoichiometry. When an intermediate or compound in the experimental part below is indicated as ‘HCI salt’ without indication of the number of equivalents of HCl, this means that the number of equivalents of HCl was not determined. The same principle will also apply to all other salt forms referred to in the experimental part, such as e.g. ‘oxalate salt’, ‘HCOOH salt’ (‘formate salt’), or




embedded image


The stereochemical configuration for centers in some compounds may be designated “R” or “S” when the mixture(s) was separated and absolute stereochemistry was known, or when only one enantiomer was obtained and absolute stereochemistry was known; for some compounds, the stereochemical configuration at indicated centers has been designated as “*R” or “*S” when the absolute stereochemistry is undetermined (even if the bonds are drawn stereo specifically) although the compound itself has been isolated as a single stereoisomer and is enantiomerically pure. In case a compound designated as “*R” is converted into another compound, the “*R” indication of the resulting compound is derived from its starting material.


For example, it will be clear that Compound 135




embedded image


For compounds wherein the stereochemical configuration of two stereocentres is indicated by * (e.g. *R or *S), the absolute stereochemistry of the stereocentres is undetermined (even if the bonds are drawn stereospecifically), although the compound itself has been isolated as a single stereoisomer and is enantiomerically pure. In this case, the configuration of the first stereocentre indicated by * is independent of the configuration of the second stereocentre indicated by * in the same compound. “*R” or “*S” is assigned randomly for such molecules. Similar for compounds wherein the stereochemical configuration of three stereocentres is indicated by * (e.g. *R or *S), the absolute stereochemistry of the stereocentres is undetermined (even if the bonds are drawn stereospecifically), although the compound itself has been isolated as a single stereoisomer and is enantiomerically pure. In this case, the configuration of the stereocentres indicated by * are independent of the configuration of the other stereocentres indicated by * in the same compound. “*R” or “*S” is assigned randomly for such molecules.


For example, for Compound 9b




embedded image


this means that the compound is




embedded image


A skilled person will realize that the paragraphs above about stereochemical configurations, also apply to intermediates.


A skilled person will realize that, even where not mentioned explicitly in the experimental protocols below, typically after a column chromatography purification, the desired fractions were collected and the solvent was evaporated.


In case no stereochemistry is indicated, this means it is a mixture of stereoisomers or undetermined stereochemistry, unless otherwise is indicated or is clear from the context.


When a stereocenter is indicated with ‘RS’ this means that a racemic mixture was obtained at the indicated centre, unless otherwise indicated.


A double bond indicated with EZ means the compound/intermediate was obtained as a mixture of E and Z isomers.


Preparation of Intermediates and Compounds

For intermediates that were used in a next reaction step as a crude or as a partially purified intermediate, in some cases no mol amounts are mentioned for such intermediate in the next reaction step or alternatively estimated mol amounts or theoretical mol amounts for such intermediate in the next reaction step are indicated in the reaction protocols described below.


Preparation of Intermediate 1:



embedded image


To a solution of 4-bromo-TH-pyrrolo[2,3-c]pyridine (2 g, 95% purity, 9.64 mmol) in 1,4-dioxane (30 mL) and water (4 mL) was added 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (7.26 g, 50% in THF, 28.9 mmol) and potassium carbonate (4.0 g, 28.9 mmol). The suspension was degassed and exchanged with N2 twice. [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (706 mg, 0.964 mmol) was added into the reaction mixture. The reaction mixture was heated up to 100° C. and stirred at this temperature overnight. After cooled down to r.t., the reaction mixture was filtered and the filtrate was concentrated. The resulting residue was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 80% to give intermediate 1 (1.01 g, 95% purity, 75.3% yield).


Alternatively, intermediate 1 can also be prepared with the following procedure:


Into a 20 L 4-necked round-bottom flask were added 4-bromo-1H-pyrrolo[2,3-c]pyridine (1330 g, 6750 mmol, 1.00 equiv), Pd(dppf)Cl2 (493.9 g, 675 mmol, 0.10 equiv), K2CO3 (2798.69 g, 20250.21 mmol, 3.00 equiv), 1,4-dioxane (13 L), H2O (2 L) and 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (2542.01 g, 20250.21 mmol, 3.00 equiv) at room temperature. The resulting mixture was stirred for overnight at 100° C. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The resulting mixture was diluted with water (15 L). The aqueous layer was extracted with EtOAc (3×10 L) and the organic layer was washed with water (2×5 L). The resulting liquid was dried with Na2SO4, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with with 10% methanol in dichloromethane to afford intermediate 1 (640 g, yield: 72%) as a grey solid.


Preparation of Intermediate 2:



embedded image


At 0° C., to a solution of intermediate 1 (918 mg, 95% purity, 6.6 mmol) in DMF (60 mL) was added a solution of N-bromosuccinimide (1.17 g, 6.6 mmol) in DMF (10 mL) dropwise. The reaction mixture was stirred at this temperature for 30 minutes. The reaction mixture was quenched with water and extracted with ethyl acetate (50 mL) twice. The organic layer was washed with brine (25 mL), dried over sodium sulfate, filtered and concentrated to afford the crude product, which was purified by silica gel column chromatography eluting with ethyl acetate in petroleum from 0% to 60% to give intermediate 2 (1.14 g, 97.1% purity, 79.5% yield) as a white solid.


Alternatively, intermediate 2 can also be prepared with the following procedure:


Into a 10 L 4-necked round-bottom flask were added intermediate 1 (640 g, 4842.39 mmol, 1.00 equiv) and DMF (5.00 L) at room temperature. To the above mixture was added NBS (861.87 g, 4842.40 mmol, 1.00 equiv) in portions over 1 h at room temperature. The resulting mixture was stirred for additional 30 min at room temperature. The reaction was quenched by the addition of aqueous solution of Na2S2O3 (10 L, 10% (w/v)) at room temperature. The aqueous layer was extracted with EtOAc (3×5 L) and the organic layer was washed with brine (1×5 L). The resulting liquid was dried with Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 20% ethyl acetate in petroleum ether to afford intermediate 2 (800 g, yield: 78%) as a grey solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 2














Int. No.
Structure
Starting Materials







Intermediate 3


embedded image


4-chloro-1H- pyrrolo[2,3-c]pyridine





Intermediate 350


embedded image


4-ethyl-1H- pyrrolo[2,3-c]pyridine









Preparation of Intermediate 4:



embedded image


To a solution of intermediate 2 (1.14 g, 97.1% purity, 5.24 mmol) in DMF (80 mL) were added 5-fluoro-2-iodobenzoic acid (1.40 mg, 5.24 mmol), copper powder (333 mg, 5.24 mmol) and potassium carbonate (2.18 g, 15.7 mmol). The reaction mixture was heated up to 100° C. and stirred at this temperature overnight. After the mixture was cooled down to r.t., the reaction mixture was concentrated and the resulting residue was acidified with HCl (1 N) to pH=˜3. The resulting mixture was filtered and the filter cake was washed with water twice. The filter cake was dried under vacuum to give crude intermediate 4 (1.8 g, 91% purity, 89.4% yield) as a yellow solid.


Alternatively, intermediate 4 can also be prepared with the following procedure:


Into a 10 L 4-necked round-bottom flask were added intermediate 2 (560 g, 2653.24 mmol, 1.00 equiv), Cu (252.91 g, 3979.87 mmol, 1.50 equiv), K2CO3 (1100.08 g, 7959.74 mmol, 3.00 equiv) and 5-fluoro-2-iodobenzoic acid (705.79 g, 2653.24 mmol, 1.00 equiv) in DMF (6.00 L) at room temperature. The resulting mixture was stirred for additional 2 h at 100° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with DMF (1×5 L) and the filtrate was concentrated under reduced pressure. The resulting mixture was diluted with water (8 L). The mixture was acidified to pH 3 with aqueous HCl (conc.). The precipitated solids were collected by filtration and washed with water (3×3 L). The resulting solid was dried under vacuum to afford intermediate 4 (1300 g, crude) as a grey solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 4














Int.

Starting


No.
Structure
Materials







Intermediate 5


embedded image


Intermediate 3





Intermediate 110


embedded image


Intermediate 1





Intermediate 351


embedded image


Intermediate 350









Preparation of Intermediate 6:



embedded image


At 0° C., to a solution of intermediate 4 (1.8 g, 91% purity, 4.69 mmol) in DMF (50 mL) was added HATU (4.46 g, 11.7 mmol), N,N-diisopropylethylamine (3.03 g, 23.5 mmol) and N-methylpropan-2-amine (858 mg, 11.7 mmol). After addition, the mixture was stirred at room temperature overnight. The reaction mixture was concentrated and the resulting residue was purified by silica gel column chromatography eluted with methanol in dichloromethane from 0% to 5% to give intermediate 6 (2.0 g, 93% purity, 98.1% yield) as a yellow oil.


Alternatively, intermediate 6 can also be prepared with the following procedure:


Into a 20 L 4-necked round-bottom flask were added intermediate 4 (920 g, 2634.90 mmol, 1.00 equiv, same as 1300 g crude), DMF (7.5 L), HATU (1102.06 g, 2898.39 mmol, 1.10 equiv) and DIEA (1021.63 g, 7904.70 mmol, 3.00 equiv) at room temperature. The resulting mixture was stirred for additional 30 min at room temperature. To the above mixture was added N-methylpropan-2-amine (211.99 g, 2898.39 mmol, 1.10 equiv) dropwise over 10 min at 0° C. The resulting mixture was stirred overnight at room temperature. The reaction was quenched by the addition of water (20 L) at room temperature. The aqueous layer was extracted with EtOAc (3×7 L) and the organic layer was washed with water (3×5 L). The resulting liquid was dried with Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with 50% ethyl acetate in petroleum ether (1:1) to afford intermediate 6 (700 g, yield: 66%) as a light yellow solid.


Alternative Approach for the Preparation of Intermediate 6

Intermediate 111 (1.3 g, 4.0 mmol) was dissolved in MeCN (40 mL). Next, CuBr2 (2.7 g, 12 mmol) was added, and the mixture was stirred at room temperature for 5 h. Next, 7N NH3/MeOH (20 mL) was added. The reaction mixture was stirred vigorously for −30 min. Then, water (40 mL) and isopropyl acetate were added. The layers were separated, and the water layer was extracted twice with isopropyl acetate. The organic layers were combined, washed with brine, dried over Na2SO4, filtered and evaporated to dryness. The residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 3% to provide intermediate 6 (1.2 g, yield 72%) as an orange oil.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 6














Int. No.
Structure
Starting Materials







Intermediate 7


embedded image


Intermediate 4 & N-ethylpropan-2-amine





Intermediate 8


embedded image


Intermediate 5 & N-methylpropan-2-amine





Intermediate 111


embedded image


Intermediate 110 & N-methylpropan-2-amine





Intermediate 317


embedded image


Intermediate 4 & propan-2-amin





Intermediate 345


embedded image


Intermediate 4 & diisopropylamine





Intermediate 352


embedded image


Intermediate 351 & N- methylpropan-2-amine









Preparation of Intermediate 9:



embedded image


To a mixture of intermediate 6 (4 g, 4.312 mmol), tert-butyl 3-((4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)methylene)azetidine-1-carboxylate (2.92 g, 9.9 mmol) and potassium carbonate (2.7 g, 19.7 mmol) in 1,4-dioxane (70 mL) and water (23 mL) was added Pd(dppf)Cl2 (724 mg, 0.99 mmol). The mixture was degassed under nitrogen atmosphere three times and the reaction was stirred at 100° C. under nitrogen atmosphere for 16 h. After the mixture was cooled down to RT, the reaction mixture was diluted with H2O and extracted with EtOAc. The combined organic phase was washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography eluting with 90% ethyl acetate in petroleum ether to give intermediate 9 (1.8 g, 45.7% purity, 38.7% yield) as a yellow solid.


Preparation of Intermediate 10:



embedded image


A mixture intermediate 6 (12.0 g, 29.8 mmol), tert-butyl 3-((4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)methylene)pyrrolidine-1-carboxylate (9.2 g, 29.8 mmol) and potassium carbonate (12.3 g, 89.1 mmol) in 1,4-dioxane (120 mL) and water (20 mL) was degassed and exchanged with N2 twice. Pd(dppf)Cl2 (2.16 g, 2.95 mmol) was added and the reaction mixture was heated up to 100° C. and stirred at this temperature overnight. After the reaction mixture was cooled down to r.t., the resulting mixture was concentrated and the residue was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 80% to give intermediate 10 (12.0 g, 79.4% yield) as a yellow oil.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 10














Int. No.
Structure
Starting Materials







Intermediate 11


embedded image


Intermediate 6 & tert-butyl 4- ((4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)methylene)piperidine-1- carboxylate





Intermediate 12


embedded image


Intermediate 7 & tert-butyl 3- ((4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)methylene)azetidine-1- carboxylate





Intermediate 13


embedded image


Intermediate 8 & tert-butyl 3- ((4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)methylene)azetidine-1- carboxylate





Intermediate 14


embedded image


Intermediate 8 & tert-butyl-3- ((4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)methylene)pyrrolidine-1- carboxylate (mixture of E and Z isomers)





Intermediate 362


embedded image


Intermediate 361 & tert-butyl 3- ((4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)methylene)azetidine-1- carboxylate









Preparation of Intermediate 15:



embedded image


A mixture of intermediate 9 (6.0 g, 12.2 mmol) in methanol (100 mL) was degassed under nitrogen atmosphere three times. 10 w/w % palladium on charcoal (3 g) was added and the mixture was degassed under hydrogen atmosphere three times. The mixture was stirred at r.t. under hydrogen atmosphere (balloon) for 16 h. The mixture was filtered and the filtrate was concentrated and purified by silica gel column chromatography eluting with 50% ethyl acetate in petroleum ether to give intermediate 15 (5.2 g, 97% purity, 83.7% yield) as a yellow solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 15














Int. No.
Structure
Starting Materials







Intermediate 19


embedded image


Intermediate 11





Intermediate 20


embedded image


Intermediate 12





Intermediate 21


embedded image


Intermediate 13





Intermediate 22


embedded image


Intermediate 14





Intermediate 363


embedded image


Intermediate 362









Preparation of Intermediate 16, 17 & 18:



embedded image


To a solution of intermediate 10 (2.5 g, 93% purity, 4.59 mmol) in methanol (40 mL) was added w/w % palladium on charcoal (1 g) under N2. The suspension was degassed under vacuum and purged with H2 several times. The reaction mixture was heated up to 30° C. and stirred at this temperature overnight. After the reaction was cooled down to r.t., the reaction mixture was filtered and the filtrate was concentrated and purified by silica gel column chromatography eluted with methanol in dichloromethane from 0% to 5% to give intermediate 16 (2.5 g, 93% purity, 99.6% yield) as a yellow oil.


Intermediate 16 (8 g, 95% purity, 14.9 mmol) was separated by chiral IG-SFC (separation condition: Column: IG; Mobile Phase: CO2-IPA: 65:35, at 60 mL/min; Temp: 40° C.; Wavelength: 214 nm) to afford intermediate 17 (first fraction, 3.29 g, 98% purity, 42.4% yield) as a yellow oil and intermediate 18 (second fraction, 3.36 g, 98% purity, 43.3% yield) as a yellow solid.


Chiral SFC method 2 was employed to match the stereochemistry of intermediate 18 and intermediate 201, retention time=5.97-6.10 min.


Preparation of Intermediate 23 & 24:



embedded image


Intermediate 22 (1.40 g, 2.65 mmol) was separated by SFC (DAICEL CHIRALPAK IG (250 mm*50 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA; Isocratic: A:B=55:45; Flow rate: 200 mL/min) to afford two fractions. The first fraction was collected as intermediate 23 (620 mg, 98.6% purity, 44% yield) as yellow solid. The second fraction was collected as intermediate 24 (650 mg, 99.9% purity, 46% yield) as a yellow solid.


Preparation of Intermediate 25:



embedded image


To a cooled (ice bath) solution of intermediate 15 (1.1 g, 2.2 mmol) in dichloromethane (14 mL) was added dropwise TFA (7 mL). Then, the mixture was stirred at r.t. for 2 h. The solvent was removed by evaporation and the residue was dissolved in DCM, the pH was adjusted to 8-9 with saturated sodium carbonate aqueous solution, and extracted with DCM. The organic phase was dried over Na2SO4 and concentrated under vacuum to give intermediate 25 (680 mg, 72% yield) as a white solid.


The Following Intermediates and Compounds were Synthesized by an Analogous Method as Described for Intermediate 25
















Starting Materials &


Int./Co No.
Structure
Methods







Intermediate 26


embedded image


Intermediate 17





Intermediate 27


embedded image


Intermediate 18





Intermediate 28


embedded image


Intermediate 19 The reaction mixture was concentrated to afford TFA salt





Intermediate 29


embedded image


Intermediate 20





Intermediate 30


embedded image


Intermediate 21





Intermediate 31


embedded image


Intermediate 23





Intermediate 32


embedded image


Intermediate 24





Compound 503


embedded image


Intermediate 184





Compound 504


embedded image


Intermediate 185





Compound 505


embedded image


Intermediate 186





Compound 522


embedded image


Intermediate 187





Compound 506


embedded image


Intermediate 203





Intermediate 213


embedded image


Intermediate 212





Intermediate 215


embedded image


Intermediate 214





Intermediate 231


embedded image


Intermediate 230





Intermediate 236


embedded image


Intermediate 235





Compound 507


embedded image


Intermediate 303a





Compound 508


embedded image


Intermediate 303b





Compound 509


embedded image


Intermediate 304a





Compound 510


embedded image


Intermediate 304b





Intermediate 319


embedded image


Intermediate 318





Compound 511


embedded image


Intermediate 322





Intermediate 338


embedded image


Intermediate 337





Intermediate 342


embedded image


Intermediate 341





Intermediate 347


embedded image


Intermediate 346





Intermediate 354


embedded image


Intermediate 353





Intermediate 358


embedded image


Intermediate 357





Intermediate 364


embedded image


Intermediate 363





Intermediate 399


embedded image


Intermediate 10









Preparation of Intermediate 33:



embedded image


To a solution of cis-3-[[(1,1-dimethylethoxy)carbonyl]amino]-cyclobutanecarboxylic acid (10.0 g, 46.5 mmol) in DMF (100 mL) was added HOBt (8.15 g, 60.3 mmol), EDCI (11.6 g, 60.5 mmol) and DIEA (30.0 mL, 182 mmol) at 0° C. Then N,O-dimethylhydroxylamine hydrochloride (5.90 g, 60.5 mmol) was added at 0° C. The mixture was stirred at room temperature for 16 hours. The mixture was diluted with ethyl acetate (500 mL), washed with 1 M aq. HCl solution (150 mL), saturated aq. NaHCO3 solution (100 mL×2) and brine (300 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure to give intermediate 33 (11.0 g, crude) as a white solid, which was used in the next step without further purification.


Preparation of intermediate 34:




embedded image


To a solution of intermediate 33 (11.0 g, 6.97 mmol) in THF (100 mL) was added isopropylmagnesium chloride (64.0 mL, 128 mmol, 2M in THF) dropwise at 0° C. under N2 atmosphere. The mixture was stirred at room temperature for 12 hours under N2 atmosphere. The mixture was quenched with saturated aq. NH4Cl solution (100 mL). The mixture was filtered through a pad of Celite® and the filtrate was extracted with ethyl acetate (200 mL×2). The combined organic layers were washed with brine (200 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 15% to yield intermediate 34 (6.30 g) as a white solid.


Preparation of Intermediate 35:



embedded image


To a solution of 3,3-dimethoxycyclobutanecarboxylic acid (12.0 g, 75 mmol) in DCM (145 mL) was added T3P (100 mL, 168 mmol. 50% in EtOAc) and DIEA (64 mL, 372 mmol) at 0° C. Then N,O-dimethylhydroxylamine hydrochloride (8.8 g, 89.5 mmol) was added at 0° C. The mixture was stirred at room temperature for 16 hours. The mixture was poured into a saturated solution of NaHCO3 and EtOAc was added. The organic layer was separated, washed with brine, dried over MgSO4, filtered and concentrated under reduced pressure to give intermediate 35 (16.0 g, crude) which was used in the next step without further purification.


Preparation of Intermediate 36:



embedded image


To a solution of intermediate 35 (15.7 g, 77.7 mmol) in THF (420 mL) was added isopropylmagnesium chloride (178.5 mL, 232 mmol, 2M in THF) dropwise at 0° C. under N2 atmosphere. The reaction mixtures were stirred at room temperature for 12 hours under N2 atmosphere. The reaction was performed twice on 15.7 g of intermediate 35 and respective reaction media were mixed for the work-up and purification. The combined reaction mixture was poured into ice-water and a 10% aqueous solution of NH4Cl and extracted with EtOAc. The organic layer was washed with brine, dried over MgSO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography eluting with 10% ethyl acetate in heptane. The pure fractions were collected and evaporated to dryness yielding 22 g (76% yield) of intermediate 36 as a colourless oil.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 36














Int. No.
Structure
Starting Materials







Intermediate 37


embedded image


3,3- dimethoxycyclobutanecarboxylic acid & ethyl magnesium bromide





Intermediate 38


embedded image


3,3- dimethoxycyclobutanecarboxylic acid & methyl magnesium bromide









Preparation of Intermediate 39:



embedded image


To a solution of N,3,3-trimethoxy-N-methylcyclobutanecarboxamide (1.5 g) in THF (50 mL) was added 1 M lithium aluminum hydride in THF (14 mL, 13.8 mmol) dropwise at −78° C. under nitrogen atmosphere. The resulting mixture was stirred at −78° C. for 3 hours. The mixture was quenched by sodium sulfate decahydrate, and then filtered and concentrated to give intermediate 39 (crude, 1.2 g) as colorless oil, which was used directly in the next step.


Preparation of Intermediate 40:



embedded image


A mixture of magnesium (6.0 g, 247 mmol) and diiodine (100 mg, 0.394 mmol) in THF (100 mL) was stirred at 25° C. Then, 2-(2-bromoethyl)-1,3-dioxolane (20.0 g, 110 mmol) in THF (50 mL) was slowly added to the mixture while maintaining the inner temperature between 20˜30° C. The mixture was stirred at 25° C. for 1 hr and slowly introduced to a solution of N-methoxy-N,2-dimethylpropanamide (10.0 g, 76.24 mmol) in THF (100 mL). The resulting mixture was stirred at 25° C. for 8 hours. The mixture was quenched with 300 mL of saturated solution of ammonium chloride and extracted with ethyl acetate (100 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 15% to afford intermediate 40 (12.8 g, 67% yield) as colorless oil.


The Following Intermediate was Synthesized by an Analogous Method as Described for Intermediate 40














Int. No.
Structure
Starting Materials







Intermediate 41


embedded image


N-methoxy-N- methylpropionamide & 2-(2- bromoethyl)-1,3-dioxolane









Preparation of Intermediate 42, 42a & 42b:



embedded image


To a solution of intermediate 25 (2.7 g, 6.844 mmol) in methanol (60 mL) was added intermediate 36 (5.0 g, 26.8 mmol), sodium cyanoborohydride (2.149 g, 34.197 mmol) and zinc dichloride (932 mg, 6.837 mmol). The mixture was stirred at 60° C. in a sealed tube for 16 h. After the reaction mixture was cooled down to r.t., the reaction mixture was concentrated and purified by silica gel column chromatography eluting with 10% methanol in dichloromethane to give intermediate 42 (3.8 g) as a white solid, which was separated by chiral Prep. HPLC into the individual enantiomers (separation condition: Column: Chiralpak IA 5 μm 20*250 mm; Mobile Phase: Hex:IPA:DEA=85:15:0.3 at 25 mL/min; Temp: 30° C.; Wavelength: 230 nm) to give the first fraction as intermediate 42a (1.03 g, 26.6% yield) as a white solid and the second fraction as intermediate 42b (1.16 g, 30.0% yield) as a white solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 42














Int. No.
Structure
Starting Materials







Intermediate 43


embedded image


Intermediate 29 & 36





Intermediate 44


embedded image


Intermediate 26 & 36





Intermediate 45


embedded image


Intermediate 27 & 36





Intermediate 46


embedded image


Intermediate 26 & 37





Intermediate 47


embedded image


Intermediate 27 & 37





Intermediate 48


embedded image


Intermediate 26 & 38





Intermediate 49


embedded image


Intermediate 27 & 38





Intermediate 50


embedded image


Intermediate 26 & 39





Intermediate 51


embedded image


Intermediate 27 & 39





Compound 512


embedded image


Intermediate 26 & 40





Compound 513


embedded image


Intermediate 27 & 40





Compound 514


embedded image


Intermediate 26 & 41





Compound 515


embedded image


Intermediate 27 & 41





Intermediate 56


embedded image


Intermediate 30 & 36





Intermediate 57


embedded image


Intermediate 31 & 37





Intermediate 58


embedded image


Intermediate 32 & 37





Compound 370


embedded image


Intermediate 31 & 40





Compound 373


embedded image


Intermediate 32 & 40





Compound 525


embedded image


Intermediate 30 & 34









Preparation of Compound 366 & 367:



embedded image


Compound 512 (1.0 g, 1.77 mmol) was separated by SFC (separation condition: Column DAICEL CHIRALCEL OD (250 mm*50 mm, 10 um); Mobile phase: A: 0.1% NH3H2O, B: MeOH, A:B=80:20 at 200 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The pure fractions were collected, and the volatiles were removed under vacuum. The first fraction was collected as Compound 366 (220 mg) and the second fraction was collected as Compound 367 (200 mg) as white solid.


Preparation of Compound 368 & 369:



embedded image


Compound 513 (1.0 g, 1.77 mmol) was separated by SFC (separation condition: DAICEL CHIRALPAK AD (250 mm*50 mm, 10 um); Mobile phase: A: 0.1% NH3H2O, B: IPA, A:B=75:25 at 200 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The pure fractions were collected, and the volatiles were removed under vacuum. The first fraction was collected as Compound 368 (380 mg, 94.8% purity, 36% yield) and the second fraction was collected as Compound 369 (280 mg, 83.5% purity, 23% yield) as a white solid.


Preparation of Intermediate 56a & 56b:



embedded image


Intermediate 56 (700 mg, 1.20 mmol) was separated by SFC (separation condition: DAICEL CHIRALPAK AD (250 mm*30 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA, A:B=65:35 at 70 mL/min). The pure fractions were collected and the solvent was evaporated under vacuum to give the products. The first fraction was collected as intermediate 56a (280 mg, 100% purity, 40.0% yield) as a colorless oil and the second fraction was collected as intermediate 56b (300 mg, 99.8% purity, 42.8% yield) as a colorless oil.


Preparation of Intermediate 58a & 58b:



embedded image


Intermediate 58 (210 mg, 0.36 mmol) was separated by SFC (separation condition: DAICEL CHIRALPAK AD (250 mm*30 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA, A:B=75:25 at 60 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The pure fraction was collected, and the solvent was evaporated under vacuum. The first fraction was collected as intermediate 58a (92.0 mg, 98.8% purity, 24.7% yield) as a yellow oil and the second fraction was collected as intermediate 58b (70.0 mg, 98.7% purity, 18.8% yield) as a yellow oil.


Preparation of Compound 371 & 372:



embedded image


Compound 370 (250 mg, 0.427 mmol) was separated by SFC (DAICEL CHIRALCEL OD-H (250 mm*30 mm, 5 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O MeOH; Isocratic: A:B=75:25; Flow rate: 70 mL/min). The pure fraction was collected, and the solvent was evaporated under vacuum. The first fraction was collected as Compound 371 (80 mg, 92.1% purity, 29% yield) as a yellow oil and the second fraction was collected as Compound 372 (90 mg, 89.6% purity, 32% yield) as a yellow oil.


Preparation of Compound 374 & 375:



embedded image


Compound 373 (250 mg, 0.43 mmol) was separated by SFC (DAICEL CHIRALPAK AD (250 mm*30 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA; Isocratic: A:B=70:30; Flow rate: 70 mL/min). The pure fraction was collected, and the solvent was evaporated under vacuum. The first fraction was collected as Compound 374 (90 mg, 97.0% purity, 35% yield) as a yellow oil and the second fraction was collected as Compound 75 (80 mg, 97.7% purity, 31% yield) as yellow oil as a yellow oil.


Preparation of Intermediate 67a:



embedded image


To a solution of intermediate 42a (110 mg, 95% purity from LCMS, 0.185 mmol) in acetonitrile (2.5 mL) was added aqueous hydrochloric acid solution (1 N, 0.8 mL) at room temperature. The reaction mixture was heated up to 35° C. and stirred at this temperature for 40 minutes. After the reaction mixture was cooled down to r.t., the reaction mixture was basified with saturated NaHCO3 aqueous solution until the pH=−8 and extracted with DCM (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated to afford intermediate 62, which was used in the next step without further purification.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 62a














Int. No.
Structure
Starting Materials







Intermediate 62b


embedded image


Intermediate 42b





Intermediate 63


embedded image


Intermediate 43





Intermediate 64


embedded image


Intermediate 44





Intermediate 65


embedded image


Intermediate 45





Intermediate 66


embedded image


Intermediate 46





Intermediate 67


embedded image


Intermediate 47





Intermediate 68


embedded image


Intermediate 48





Intermediate 69


embedded image


Intermediate 49





Intermediate 70


embedded image


Intermediate 50





Intermediate 71


embedded image


Intermediate 51





Intermediate 72


embedded image


Compound 512





Intermediate 72a


embedded image


Compound 366





Intermediate 72b


embedded image


Compound 367





Intermediate 73


embedded image


Compound 513





Intermediate 73a


embedded image


Compound 368





Intermediate 73b


embedded image


Compound 369





Intermediate 74


embedded image


Compound 514





Intermediate 75


embedded image


Compound 515





Intermediate 76a


embedded image


Intermediate 56a





Intermediate 76b


embedded image


Intermediate 56b





Intermediate 77


embedded image


Intermediate 57





Intermediate 78a


embedded image


Intermediate 58a





Intermediate 78b


embedded image


Intermediate 58b





Intermediate 79a


embedded image


Compound 371





Intermediate 79b


embedded image


Compound 372





Intermediate 80a


embedded image


Compound 374





Intermediate 80b


embedded image


Compound 375









Preparation of Compound 376:



embedded image


A 4M solution of HCl in dioxane (1.90 mL, 7.60 mmol) was added to a solution of compound 525 (310 mg, 0.48 mmol) in dioxane (3 mL) at 0° C. After stirring at r.t. for 1 hr, the reaction mixture was concentrated under reduced pressure to give Compound 376 (420 mg, crude), which was used in next step without further purification.


Preparation of Intermediate 82:



embedded image


EDCI·HCl (35.0 g, 183 mmol) was added to a solution of 4-((tert-butoxycarbonyl) (methyl)amino)butanoic acid (28.0 g, 129 mmol), N,O-dimethylhydroxylamine hydrochloride (16.0 g, 164 mmol), HOBt (17.5 g, 130 mmol) and 4-methylmorpholine (78.0 g, 771 mmol) in CHCl3 (500 mL). After stirring at r.t. for 16 hours, the reaction mixture was subsequently washed with water (250 mL×2), 0.1N aq. HCl solution (250 mL×2), sat. aq. NaHCO3 solution (250 mL×2) and brine (250 mL×2). The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated in vacuo to give the crude product which was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 50% to afford intermediate 82 (27 g, 80% yield) as a colorless oil.


Preparation of Intermediate 83:



embedded image


At 0° C., to a solution of intermediate 82 (27.0 g, 104 mmol) in THF (800 mL) was added prop-1-en-2-ylmagnesium bromide (260 mL, 260 mmol, 1 M) under N2. The mixture was stirred at 0° C. under N2 for 1 hour, slowly warmed up to room temperature and stirred at room temperature for 16 hours. The mixture was quenched with sat. aq. NH4Cl solution (400 mL) and extracted with EtOAc (500 mL×3). The combined organic layers were washed with H2O (300 mL×2) and brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give intermediate 83 (25 g, crude) as light yellow oil, which was used in the next step without further purification.


_Preparation of Intermediate 84:



embedded image


To a solution of intermediate 83 (11.0 g, crude) in MeOH (100 mL) was added 10 w/w % Pd/C (1 g) under N2 atmosphere. The mixture was degassed under vacuum and purged with H2 three times. The mixture was stirred at rt for 2 hours under H2 (15 psi) atmosphere. The reaction mixture was filtered through a pad of Celite®, and the filter cake was washed with MeOH (30×2 mL). The filtrate was concentrated under reduced pressure to give the crude product which was purified by silica gel column chromatography eluting with 20% ethyl acetate in petroleum ether to give intermediate 84 (9.5 g, 86% yield) as a colorless oil.


Preparation of Intermediate 85:



embedded image


To a solution of intermediate 84 (20.0 g, 82.2 mmol) in DCM (200 mL) was added 4M HCl in dioxane solution (120 mL, 480 mmol). After stirring at r.t for 1 hour, the reaction mixture was concentrated under reduce pressure to give intermediate 85 (17.8 g, crude) as a white solid, which was used in next step without further purification.


Preparation of Intermediate 86:



embedded image


To a solution of intermediate 85 (70 g, crude), K2CO3 (224 g, 1621 mmol) and NaI (146 g, 974 mmol) in DMF (700 ml) was added 1-bromo-2-methoxyethane (54 g, 389 mmol). The mixture was stirred at 50° C. for 5 hours. The insoluble residues were removed via filtration, and the filtrate was concentrated under reduced pressure to give the crude product, which was poured into water (500 mL) and extracted with ethyl acetate (500 mL×3). The combined organic layers were washed with brine (100 mL×3), 5% aq. LiCl solution (100 mL×3) and water (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give intermediate 86 (25 g, 38% yield) as a brown oil.


Preparation of Intermediate 87:



embedded image


To a solution of tert-butyl (trans)-rel-octahydropyrrolo[3,4-c]pyrrole-2-carboxylate hemioxalate (1.00 g, 3.89 mmol) in anhydrous dichloromethane (20.0 mL) was added triethylamine (2.00 g, 19.8 mmol). Then acetic anhydride (600 mg, 5.88 mmol) was added dropwise, and the reaction mixture was stirred at 25° C. for 70 minutes. The reaction mixture was diluted with dichloromethane (30 mL) and washed with water (20 mL×1), brine (20 mL×1) and saturated aqueous sodium bicarbonate solution (20 mL×1). The organic phase was dried over Na2SO4, filtered, and concentrated under reduced pressure to give intermediate 87 (990 mg, 95.0% purity, 95.2% yield) as a white solid.


Preparation of Intermediate 88:



embedded image


Intermediate 87 was separated by SFC (separation condition: DAICEL CHIRALPAK AS (250 mm*50 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O EtOH, A:B=85:15 at 200 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The second fraction was collected as intermediate 88 (3.36 g, 97.0% purity, 43.2% yield) as a white solid.


Preparation of Intermediate 89:



embedded image


To a solution of intermediate 88 (300 mg, 1.18 mmol) in anhydrous dichloromethane (2 mL) was added trifluoroacetic acid (2 mL). After stirring at 25° C. for 1 h, the reaction mixture was concentrated under reduced pressure to give intermediate 89 (300 mg, crude) as yellow oil, which was used in the next step without further purification.


Preparation of Compound 377:



embedded image


To a solution of intermediate 26 (100 mg, 95% purity, 0.223 mmol) in methanol (3 mL) were added tert-butyl 4-formylpiperidine-1-carboxylate (104 mg, 0.465 mmol) and sodium triacetoxyborohydride (98.1 mg, 0.465 mmol). After stirring at r.t. for 6 hours, the reaction mixture was concentrated, and the residue was purified by preparative TLC (10% MeOH in DCM) to give Compound 377 (130 mg, 95% purity, 87.7% yield) as a white oil.


Preparation of Compound 378:



embedded image


To a solution of intermediate 27 (3.5 g, 95%, 8.14 mmol) in DCM (80 mL) was added tert-butyl 4-formylpiperidine-1-carboxylate (3.66 g, 16.3 mmol) and sodium triacetoxyborohydride (2.58 g, 12.2 mmol). After stirring at room temperature for 6 hours, the reaction mixture was poured into saturated aqueous sodium bicarbonate solution and extracted with dichloromethane (80 mL) twice. The combined organic layers were washed with brine (80 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to afford the crude product, which was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 6% to give Compound 378 (4.66 g, 95% purity, 89.8% yield) as a white oil.


The Following Intermediates and Compounds were Synthesized by an Analogous Method as Described for Compound 378


In case reactions were performed with a ketone starting material, a typical procedure makes use of either 2 eq. acetic acid or 2 eq. of zinc(II)chloride (ZnC2), in the presence of 2 eq. sodium cyanoborohydride (NaCNBH3), in methanol at 50° C. or 70° C. overnight.














Int./Co No.
Structure
Starting Materials







Compound 62


embedded image


Intermediate 25 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 382


embedded image


Intermediate 25 & tert-butyl 4- formyl-4-methylpiperidine-1- carboxylate





Compound 383


embedded image


Intermediate 25 & tert-butyl 3- formylpiperidine-1-carboxylate





Compound 384


embedded image


Intermediate 25 & tert-butyl 3- formylpyrrolidine-1-carboxylate





Compound 380


embedded image


Intermediate 28 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 385


embedded image


Intermediate 28 & tert-butyl 3- formylazetidine-1-carboxylate





Intermediate 157


embedded image


Intermediate 27 & tert-butyl (1- oxopropan-2-yl)carbamate





Compound 386


embedded image


Intermediate 27 & tert-butyl 6- formyl-2-azaspiro[3.3]heptane- 2-carboxylate





Compound 387 & Compound 388


embedded image


Intermediate 27 & tert-butyl 4- acetylpiperidine-1-carboxylate The product was separated by supercritical fluid chromatography (separation condition: DAICEL CHIRALPAK AD(250 mm*30 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA, A:B = 75:25 at 70 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 387 and the second fraction as Compound 388








embedded image








Intermediate 171


embedded image


Intermediate 27 & tert-butyl (1- oxopropan-2-yl)carbamate





Compound 389


embedded image


Intermediate 27 & tert-butyl 4- formyl-4-methylpiperidine-1- carboxylate





Compound 501


embedded image


Intermediate 28 & 177





Compound 391


embedded image


Intermediate 27 & 3- methylpiperidin-4-one





Compound 392


embedded image


Intermediate 202 & tert-butyl 4- fluoro-4-formylpiperidine-1- carboxylate





Compound 394


embedded image


Intermediate 202 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 395 & Compound 396


embedded image


Intermediate 25 & tert-butyl 4- acetylpiperidine-1-carboxylate The product was separated by SFC (separation condition: DAICEL CHIRALPAK AD(250 mm*30 mm, 10 um); Mobile phase: A: 0.1% NH3H2O, B: EtOH, A:B = 75:25 at 70 mL/min; Column Temp: 38; Nozzle Pressure: 100Bar; Nozzle Temp: 60; Evaporator Temp: 20; Trimmer Temp: 25; Wavelength: 220 nm). The first fraction was collected as Compound 395 & the second fraction as Compound 396.








embedded image








Compound 397


embedded image


Intermediate 225 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 526a & Compound 526b


embedded image


Intermediate 25 & (trans)- methyl 4- acetylcyclohexanecarboxylate. The product was separated by SFC (separation condition: DAICEL CHIRALPAK IC(250 mm*30 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O MeOH, A:B = 50:50 at 80 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 526a & the second fraction as Compound 526b.








embedded image








Compound 398 & Compound 399


embedded image


Intermediate 202 & tert-butyl 3- acetylazetidine-1-carboxylate The product was separated by SFC (separation condition: DAICEL CHIRALPAK IC(250 mm*30 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O MeOH, A:B = 50:50 at 80 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 398 & the second fraction as Compound 399.








embedded image








Compound 400


embedded image


Intermediate 202 & tert-butyl 4- (2-oxoethyl)piperidine-1- carboxylate





Compound 401


embedded image


Intermediate 25 & tert-butyl 4- (2-oxoethyl)piperidine-1- carboxylate





Compound 402 & Compound 403


embedded image


Intermediate 202 & tert-butyl 4- acetylpiperidine-1-carboxylate The product was separated by SFC (separation condition: DAICEL CHIRALPAK AD(250 mm*50 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA, A:B = 65:35 at 200 mL/min; Column Temp: 38; Nozzle Pressure: 100Bar; Nozzle Temp: 60; Evaporator Temp: 20; Trimmer Temp: 25; Wavelength: 220 nm). The first fraction was collected as Compound 402 & the second as Compound 403.








embedded image








Compound 404 & Compound 405


embedded image


Intermediate 202 & tert-butyl (4- oxocyclohexyl)carbamate








embedded image








Compound 407


embedded image


Intermediate 202 & 1,4- dioxaspiro[4.5]decane-8- carbaldehyde





Compound 408


embedded image


Intermediate 225 & 3-Boc-6- oxo-3-aza-bicyclo[3.1.1]heptane





Compound 409


embedded image


Intermediate 202 & 3-Boc-6- oxo-3-aza-bicyclo[3.1.1]heptane





Compound 410 & Compound 411


embedded image


Intermediate 225 & tert-butyl (4- oxocyclohexyl)carbamate








embedded image








Compound 412


embedded image


Intermediate 202 & tert-butyl 3- oxopiperidine-1-carboxylate





Compound 413


embedded image


Intermediate 202 & tert-butyl 4- oxoazepane-1-carboxylate





Compound 414


embedded image


Intermediate 202 & tert-butyl 4- formyl-4-methylpiperidine-1- carboxylate





Compound 415


embedded image


Intermediate 225 & tert-butyl 4- formyl-4-methylpiperidine-1- carboxylate





Compound 416


embedded image


Intermediate 225 & tert-butyl 4- oxoazepane-1-carboxylate





Compound 417


embedded image


Intermediate 202 & tert-butyl 5- oxo-2-azabicyclo[2.2.1]heptane- 2-carboxylate





Compound 418


embedded image


Intermediate 202 & tert-butyl 3,3-dimethyl-4-oxopiperidine-1- carboxylate





Compound 419


embedded image


Intermediate 319 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 420 & Compound 421


embedded image

embedded image

Intermediate 202 & tert-butyl 4- methyl-3-oxopiperidine-1- carboxylate The product was separated by SFC (separation condition: DAICEL CHIRALPAK AD(250 mm*30 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O EtOH, A:B = 75:25 at 70 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 420 & the second fraction as Compound 421.





Compound 422 & Compound 423


embedded image


Intermediate 202 & 327 The product was separated by SFC (separation condition: DAICEL CHIRALPAK IG (250 mm*30 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O IPA, A:B = 70:30 at 75 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 422 & the second fraction as Compound 423.








embedded image








Intermediate 330


embedded image


Intermediate 202 & tert-butyl 3-(((tert- butyldimethylsilyl)oxy)methyl)- 4-oxopiperidine-1-carboxylate





Compound 424


embedded image


Intermediate 338 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 425


embedded image


Intermediate 342 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 426


embedded image


Intermediate 347 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 427


embedded image


Intermediate 354 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 428


embedded image


Intermediate 358 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 527


embedded image


Intermediate 364 & tetrahydro- 2H-pyran-4-carbaldehyde





Compound 528


embedded image


Intermediate 368 & 1- acetylpiperidine-4-carbaldehyde





Compound 529


embedded image


Intermediate 368 & tert-butyl 4- formylpiperidine-1-carboxylate





Compound 531


embedded image


Intermediate 368 & tert-butyl 4- formyl-4-methylpiperidine-1- carboxylate





Compound 532


embedded image


Intermediate 368 & intermediate 398





Compound 533


embedded image


Intermediate 368 & 7- oxoazepane-4-carbaldehyde





Compound 534


embedded image


Intermediate 368 & tetrahydro- 2H-pyran-4-carbaldehyde









Preparation of Compound 381:



embedded image


Compound 531 (70 mg, 0.104 mmol) was dissolved in DCM (3 mL, 46.837 mmol). TFA (1 mL, 13.067 mmol) was added. The mixture was stirred at RT for 2 hours. The solvent was removed to give Compound 381, which was used in the next step without further purification.


The Following Intermediates and Compounds were Synthesized by an Analogous Method as Described for Compound 381














Int./Co No.
Structure
Starting Materials







Compound 429


embedded image


Compound 62





Compound 430


embedded image


Compound 382





Compound 431


embedded image


Compound 383





Compound 432


embedded image


Compound 384





Compound 433


embedded image


Compound 380





Compound 434


embedded image


Compound 491





Compound 435


embedded image


Compound 492





Compound 436


embedded image


Compound 385





Compound 437


embedded image


Intermediate 157





Compound 438


embedded image


Compound 386





Compound 439


embedded image


Compound 493





Compound 502


embedded image


Compound 440





Compound 441


embedded image


Compound 487





Compound 442


embedded image


Compound 387





Compound 443


embedded image


Compound 388





Compound 444


embedded image


Intermediate 171





Compound 445


embedded image


Compound 389





Compound 446


embedded image


Compound 91





Compound 447


embedded image


Compound 392





Compound 448


embedded image


Compound 394





Compound 449


embedded image


Compound 395





Compound 450


embedded image


Compound 396





Compound 451


embedded image


Compound 397





Compound 452


embedded image


Compound 398





Compound 453


embedded image


Compound 399





Compound 454


embedded image


Compound 500





Compound 455


embedded image


Compound 495





Compound 456


embedded image


Compound 400





Compound 457


embedded image


Compound 401





Compound 458


embedded image


Compound 402





Compound 459


embedded image


Compound 403





Compound 406a


embedded image


Compound 404





Compound 406b


embedded image


Compound 405





Compound 462


embedded image


Compound 408





Compound 463


embedded image


Compound 409





Compound 464


embedded image


Compound 410





Compound 465


embedded image


Compound 411





Compound 466


embedded image


Compound 412





Compound 467


embedded image


Compound 413





Compound 468


embedded image


Compound 414





Compound 469


embedded image


Compound 415





Compound 470


embedded image


Compound 496





Compound 471


embedded image


Compound 416





Compound 472


embedded image


Compound 417





Compound 473


embedded image


Compound 418





Compound 474


embedded image


Compound 419





Compound 475


embedded image


Compound 420





Compound 476


embedded image


Compound 421





Compound 477


embedded image


Compound 422





Compound 478


embedded image


Compound 423





Compound 479


embedded image


Intermediate 330





Compound 480


embedded image


Compound 424





Compound 481


embedded image


Compound 425





Compound 482


embedded image


Compound 426





Compound 483


embedded image


Compound 427





Compound 484


embedded image


Compound 428





Compound 530


embedded image


Compound 529





Intermediate 378


embedded image


Compound 531





Intermediate 386


embedded image


Compound 532









Preparation of Compound 485:



embedded image


At 0° C., to a solution of Compound 378 (650 mg, 95% purity, 1.02 mmol) in DCM (8 mL) was added hydrogen chloride in ethyl acetate (2.2 mL, 7 M). After stirring at room temperature for 2 hours, the reaction mixture was concentrated and the residue was basified with aqueous sodium hydroxide solution (1M) and extracted with DCM (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated to afford Compound 485, which was used in the next step without purification.


Preparation of Compound 486:



embedded image


To a solution of intermediate 26 (300 mg, 0.734 mmol) and tert-butyl ((trans)-4-formylcyclohexyl)carbamate (334 mg, 1.47 mmol) in anhydrous methanol (8 mL) was added acetic acid (88.2 mg, 1.47 mmol). The reaction mixture was heated and stirred at 45° C. for 30 minutes before sodium cyanotrihydroborate (92.3 mg, 1.47 mmol) was added. After stirring at 45° C. for another 12 h, the reaction mixture was cooled down to room temperature, diluted with dichloromethane (50 mL), basified to pH=8 with saturated aq. sodium bicarbonate solution (40 mL) and extracted with dichloromethane (30 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by preparative-HPLC (Column: Boston Green ODS 150*30 mm*5 um, Mobile Phase A: water (0.225% FA), Mobile Phase B: acetonitrile, Flow rate: 35 mL/min, gradient condition from 1% B to 30% B). The pure fractions were collected and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give Compound 486 (400 mg, 92.5% purity, 81.3% yield) as a white powder.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 486














Co No.
Structure
Starting Materials







Compound 487


embedded image


Intermediate 25 & tert-butyl ((trans)-4- formylcyclohexyl)carbamate









Preparation of Compound 488:



embedded image


To a solution of Compound 486 (380 mg, 0.613 mmol) in anhydrous dichloromethane (4 mL) was added trifluoroacetic acid (4 mL). After stirring at 25° C. for 1 hour, the reaction mixture was concentrated under reduced pressure to give the Compound 488 (380 mg, crude, TFA salt) as a yellow oil.


Preparation of Intermediate 114:



embedded image


To a mixture of (trans)-4-(methoxycarbonyl)cyclohexanecarboxylic acid (5 g, 26.8 mmol), methanamine hydrochloride (2.72 g, 40.3 mmol), EDCI (6.2 g, 32.3 mmol), HOBt (6.0 g, 32.5 mmol) in DCM (80 mL) was added DIPEA (22.5 mL, 136 mmol). After stirring at room temperature overnight, the reaction mixture was diluted with DCM (50 mL), washed with 1 mol/L aq. HCl (100 mL), NaHCO3 aq. (100 mL) and brine (100 mL) and dried over sodium sulfate. The solution was filtered and concentrated in vacuo to give intermediate 114 (4.4 g, 90% purity, 74.0% yield) as a white solid.


Preparation of Intermediate 115:



embedded image


At 0° C., to a solution of LiAlH4 (570 mg, 15.0 mmol) in dry THF (10 mL) under N2, was added a solution of intermediate 114 (2.5 g, 12.5 mmol) in dry THF (20 mL) dropwise over 10 min. After addition, the reaction mixture was stirred at 0° C. for 2 h. The reaction was quenched with H2O (0.5 mL), 10% aq. NaOH (0.5 mL), THF (10 mL), H2O (1.5 mL), stirred for 10 min, and dried over Na2SO4. The suspension was filtered through Celite and the filtrate was concentrated to give crude intermediate 115 (1 g, 90% purity, 41.9% yield) as a white solid.


Preparation of Intermediate 116:



embedded image


At 0° C., to a solution of intermediate 115 (500 mg, crude) and triethylamine (1 ml, 7.20 mmol) in DCM (5 ml) was added a solution 4-methylbenzene-1-sulfonyl chloride (557 mg, 2.92 mmol) in DCM (5 ml) and N,N-dimethylpyridin-4-amine (71.5 mg, 0.585 mmol). After stirring at room temperature overnight, the reaction mixture was concentrated and the residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 9% to afford intermediate 116 (300 mg, 99.1% purity, 31.1% yield) as white solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 116














Int. No.
Structure
Starting Materials







Intermediate 207


embedded image


(trans)-methyl 4- (hydroxymethyl) cyclohexanecarboxylate





Intermediate 290


embedded image


(cis)-methyl 4- (hydroxymethyl) cyclohexanecarboxylate










Preparation of intermediate 117:




embedded image


To a solution of 6-chloro-N-methylpyrazine-2-carboxamide (0.55 g, 3.2 mmol) in DMA (20 mL) was added 1,4-dioxa-8-azaspiro[4.5]decane (0.46 g, 3.2 mmol), followed by DIPEA (1.7 mL, 9.6 mmol). The mixture was heated up to 130° C. and stirred at that temperature overnight. After the reaction mixture was cooled down to ambient temperature, water and EtOAc were added. The layers were separated, and the water layer was extracted 3×more with EtOAc. The organic layers were combined, dried over Na2SO4, filtered, and evaporated to dryness. The residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 5% to give intermediate 117 (0.83 g, 3.0 mmol, yield: 93%) as an orange oil.


Preparation of Intermediate 118:



embedded image


Intermediate 117 (830 mg, 3.0 mmol) was dissolved in THF (33 mL). To this solution was added 1M aq. HCl (33 mL) and the mixture was stirred at 50° C. until full consumption of the starting material (˜3 h). The mixture was cooled down to ambient temperature and sat. aq. NaHCO3 solution and EtOAc were added. After separation of the layers, the water layer was extracted twice with EtOAc. The organic layers were combined, dried over Na2SO4, filtered, and evaporated to dryness. Purification by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 4% gave intermediate 118 (0.40 g, 1.7 mmol, yield: 57%) as a yellow solid.


Preparation of Intermediate 119:



embedded image


Intermediate 6 (0.40 g, 0.99 mmol), Cs2CO3 (1.09 g, 3.4 mmol), Pd(dppf)Cl2 (0.072 g, 0.10 mmol) and t-butyl 3-methyleneazetidine-1-carboxylate (0.31 g, 1.8 mmol) were added to a flame dried vial, equipped with a stir bar. Next, the vial was evacuated and refilled with N2, which was repeated three times. Then, anhydrous DMF (8.0 mL) was added, and the mixture stirred at 100° C. overnight. MeOH was added to dissolve the mixture and evaporated to dryness. The residue was partitioned between DCM/water. The layers were separated, and the water layer was extracted twice more with DCM. Organic layers were combined, dried over Na2SO4, filtered, and evaporated to dryness. Purification by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 2.5% gave intermediate 119 (313 mg, 86% purity, 54% yield).


Preparation of Intermediate 120:



embedded image


To a mixture of intermediate 119 (0.31 g, 0.55 mmol) in MeOH (30 mL) was added a catalytic amount of Pd/C (10% w/w), and the solution was stirred under H2 atmosphere with balloon for 2.5 h. Then, the mixture was filtered, washed with MeOH, and evaporated to dryness. The residue was purified by silica gel column chromatography eluting with ethyl acetate in heptane from 40% to 80% to give intermediate 120 (0.14 g, 0.29 mmol, 52% yield).


Preparation of Intermediate 121:



embedded image


Dissolve intermediate 120 (0.14 g, 0.28 mmol) in DCM (3 mL). Next, TFA (3 mL) was added. The mixture was subsequently stirred at ambient temperature for ˜3 h. Then, the mixture was evaporated to dryness, and applied to a SiliaBond@ propylsulfonic acid resin as a solution in MeOH. The resin was eluted with MeOH (7 fractions), followed by 3.5N NH3 in MeOH (7 fractions). Product containing fractions were pooled and evaporated to dryness to give intermediate 121 (0.11 g, 0.26 mmol), which was used in the next step without further purification.


Preparation of Intermediate 122:



embedded image


2-Iodopropane (1.64 mL, 16.4 mmol) was added at r.t. to a solution of 2,5-difluorothiophenol (2.00 g, 13.7 mmol) and potassium carbonate (2.65 g, 19.2 mmol) in acetone (46 mL) and the reaction mixture was stirred at 75° C. for 2 h. The reaction mixture was cooled back to r.t., quenched with water (20 mL) and concentrated under reduced pressure to remove the acetone. The aqueous layer was extracted with DCM (4×40 mL) and the combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give intermediate 122 (2.35 g, 91.2% yield) as a pale yellow oil which was used without further purification in the following step.


Preparation of Intermediate 123:



embedded image


Iodobenzene diacetate (PIDA) (8.16 g, 25.3 mmol) was added at rt to a stirred solution of intermediate 122 (2.34 g, 12.1 mmol) and ammonium carbamate (1.41 g, 18.1 mmol) in MeOH (24 mL) and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated under reduced pressure to afford a yellow mixture. The crude product was purified by silica gel column chromatography eluting with ethyl acetate in heptane from 10% to 100% to give intermediate 123 (2.45 g, yield 93%) as a colorless oil.


Preparation of Intermediate 124:



embedded image


Mel (1.04 mL, 16.8 mmol) was added under nitrogen to a mixture of intermediate 123 (2.45 g, 11.2 mmol) and KOH (1.25 g, 22.4 mmol) in DMSO (61 mL) and the reaction mixture was stirred at r.t. for 90 min. The mixture was diluted with water (600 mL), extracted with ethyl acetate (×4), and the combined organic phases were dried over sodium sulfate, filtered and evaporated to dryness. The crude product was purified by silica gel column chromatography eluting with ethyl acetate in heptane from 30% to 100% to give intermediate 124 (2.41 g, 92.4% yield) as a pale yellow oil.


Preparation of Intermediate 125:



embedded image


t-BuOK 1.0 M in THF (5.7 mL, 5.70 mmol) was added under nitrogen at 0° C. to a stirred solution of intermediate 123 (1.00 g, 4.56 mmol) in anhydrous THF (15 mL). After 15 min, BOC-anhydride (1.99 g, 9.12 mmol) in anhydrous THF (30 mL) was added and the reaction was left under stirring at rt for 60 h. The reaction was evaporated to dryness, and the residue was dissolved in EtOAc, and the organic phase was washed with water (×2), dried over sodium sulfate, filtered, and evaporated to dryness. The crude product was purified by silica gel column chromatography eluting with ethyl acetate in heptane from 10% to 80% to give intermediate 125 (1.5 g, 99.9% yield) as a white solid.


Preparation of Intermediate 126:



embedded image


t-Butyl 3-formylpyrrolidine-1-carboxylate (4.15 g, 20.8 mmol) was added to a stirred mixture of intermediate 1 (2.50 g, 18.9 mmol) and NaOH (2.27 g, 56.7 mmol) in MeOH (77 mL), and the solution was refluxed for 26 h. The reaction was evaporated to dryness and purified by silica gel column chromatography eluting with methanol in dichloromethane from 2% to 20% to give intermediate 126 as an E & Z mixture (6.11 g, yield 62.9%, 61% purity) which was used in the following step without any further purification.


The Following Intermediate was Synthesized by an Analogous Method as Described for Intermediate 126














Int. No.
Structure
Starting Materials







Intermediate 237


embedded image


Intermediate 1 & t-butyl 3- formylazetidine- 1-carboxylate









Preparation of Intermediate 127:



embedded image


To a solution of intermediate 126 (6.10 g, 19.5 mmol) in MeOH (79 mL) was added Pd/C (10% w/w) (1.90 g, 1.78 mmol) under nitrogen. The suspension was hydrogenated at 1 bar Hydrogen at rt for 16 h. The reaction was filtered over Celite®, and the filtrate was evaporated to dryness. The residue was purified by reversed-phase prep. HPLC purification (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give intermediate 127 (1.71 g, yield 45.7%) as a white solid.


The Following Intermediate was Synthesized by an Analogous Method as Described for Intermediate 127














Int. No.
Structure
Starting Materials







Intermediate 238


embedded image


Intermediate 237









Preparation of Intermediate 128:



embedded image


Potassium tert-butoxide (0.42 mL, 1 M in THF, 0.418 mmol) was added under nitrogen to a solution of intermediate 127 (110 mg, 0.350 mmol) in anhydrous dioxane (1.7 mL). After 10 min, this solution was added to a solution of intermediate 124 (203 mg, 0.872 mmol) in anhydrous dioxane (1.5 mL), and the mixture was stirred at 80 SC overnight. The reaction was cooled down to rt, evaporated to dryness, and the crude was purified by silica gel column chromatography eluting with methanol in dichloromethane from 000 to 15% to give intermediate 128 (96 mg, yield 35.4%) as a yellow glassy solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 128














Int. No.
Structure
Starting Materials







Intermediate 129


embedded image


Intermediate 125 & 127





Intermediate 239


embedded image


Intermediate 238 & 124





Intermediate 241


embedded image


Intermediate 238 & 125





Intermediate 277


embedded image


Intermediate 140a & 276





Intermediate 297


embedded image


Intermediate 140b & 276





Intermediate 309


embedded image


intermediate 125 & 140a





Intermediate 310


embedded image


intermediate 125 & 140b









Preparation of Intermediate 130:



embedded image


TFA (1.01 mL, 13.2 mmol) was added to a stirred solution of intermediate 128 (97.0 mg, 0.183 mmol) in DCM (1.0 mL). After 30 min, the reaction was evaporated to dryness, and the crude product was dissolved in MeOH and transferred to a column loaded with SiliaBond® propylsulfonic acid resin resin. The column was first eluted with MeOH (20 mL), followed by NH3 in methanol (7N, 12 mL). The tubes containing the product were concentrated under reduced pressure to give intermediate 130 (78 mg, yield 66.5%) as a pale yellow glassy solid.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 130














Int. No.
Structure
Starting Materials







Intermediate 131


embedded image


Intermediate 129





Intermediate 131a & Intermediate 131b & Intermediate 131c & Intermediate 131d


embedded image

embedded image

Intermediate 131 was separated via Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as intermediate 131a, the second fraction as intermediate 131b and the third fraction which was a mixture of intermediate 131c & 131d was further separated by Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as intermediate 131c and the second fraction as intermediate 131d








embedded image











embedded image








Intermediate 240


embedded image


Intermediate 239





Intermediate 242


embedded image


Intermediate 241





Intermediate 278


embedded image


Intermediate 277





Intermediate 298


embedded image


Intermediate 297





Intermediate 311a & Intermediate 311b


embedded image


Intermediate 309 The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as intermediate 311a & the second fraction as intermediate 311b.








embedded image








Intermediate 312a & Intermediate 312b


embedded image


Intermediate 310 The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as intermediate 312a & the second fraction as intermediate 312b.








embedded image











Preparation of Intermediate 134:



embedded image


To a solution of 5-(hydroxymethyl)piperidin-2-one (300 mg, 2.32 mmol) in DMF (5 mL), was added sodium hydride (60% in mineral oil) (140 mg, 3.484 mmol) at 0° C. After 10 min, 4-methylbenzene-1-sulfonyl chloride (532 mg, 2.787 mmol) was added and the mixture was stirred at 0° C. for 3 hr. The mixture was quenched by water (30 mL) and extracted with EtOAc (20 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to get the crude, which was purified by silica gel column chromatography eluting with MeOH in DCM from 0% to 10% to give intermediate 134 (217 mg, 89.15% purity from LCMS, 29.4% yield) as a white solid.


Preparation of Intermediate 135:



embedded image


At 0° C., to a solution of 6-(hydroxymethyl)piperidin-2-one (180 mg, 1.39 mmol), DIEA (0.48 mL, 2.8 mmol) and 4-dimethylaminopyridine (17.0 mg, 0.14 mmol) in DCM (10.8 mL) was added 4-methylbenzenesulfonyl chloride (433.2 mg, 2.27 mmol). After stirring at r.t. for 16 hours, the resulting mixture was washed with brine, drived over Na2SO4. The organic solvent was removed and the residue was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 50% to 100% to afford intermediate 135 (300 mg, 90% purity, 68% yield).


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 135














Int. No.
Structure
Starting Materials







Intermediate 149


embedded image


tert-butyl (S)-3- (hydroxymethyl) pyrrolidine-1- carboxylate





Intermediate 150


embedded image


tert-butyl (R)-3- (hydroxymethyl) pyrrolidine-1- carboxylate





Intermediate 159


embedded image


(S)-5- (hydroxymethyl)- 205-yrrolidine- 2-one





Intermediate 162


embedded image


(R)-5- (hydroxymethyl)- 205-yrrolidine- 2-one





Intermediate 177


embedded image


methyl 3- (hydroxymethyl) cyclobutanecarboxylate





Intermediate 249


embedded image


4-(hydroxymethyl)- pyrrolidine- 2-one





Intermediate 250


embedded image


(1R,5S,6r)-tert-butyl 6- (hydroxymethyl)-3- azabicyclo[3.1.0]hexane- 3- carboxylate





Intermediate 273


embedded image


4-(hydroxymethyl) piperidin- 2- one





Intermediate 287


embedded image


tert-butyl (1R,5S,6s)-6- (hydroxymethyl)-3- azabicyclo[3.1.0]hexane-3- carboxylate





Intermediate 336


embedded image


5-(hydroxymethyl)azepan- 2-one









Preparation of Intermediate 138:



embedded image


HCl in water (0.376 mL, 0.1 M, 0.038 mmol) was added to a stirred solution of intermediate 123 (550 mg, 2.51 mmol) and m-CPBA (1.237 g, 5.02 mmol) in THF (9.8 mL). The mixture was heated at reflux for 24 h. The mixture was cooled down to rt, diluted with EtOAc and washed with NaOH 1N (×3), water (×1), sat Na2S2O3 (×3), dried over sodium sulfate, filtered and evaporated to dryness. The crude was purified by by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 10% to 70% to give intermediate 138 (300 mg, yield 54.3%) as a colorless oil.


Preparation of Intermediate 127, 140a & 140b:



embedded image


Intermediate 126 (6.10 g, 11.9 mmol) was dissolved in MeOH (48 mL) and added under nitrogen to Pd/C (10% w/w) (1.90 g, 1.78 mmol), and the mixture was hydrogenated at 1 bar hydrogen at r.t. during 16 h. The reaction was filtered over Celite®, evaporated to dryness to afford 5.3 g of crude product. 400 mg of crude product was purified by prep HPLC and the remaining was purified by reversed-phase prep HPLC purification (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give intermediate 127 (1.58 g, yield 42.3%) as white solid. A further purification was performed via Prep SFC (Stationary phase: Chiralpak Diacel AD 20×250 mm, Mobile phase: CO2, iPrOH+0.4 iPrNH2). The first fraction was collected as intermediate 140a and the second fraction as intermediate 140b.


Preparation of Intermediate 141:



embedded image


Potassium tert-butoxide (0.68 mL, 1 M in THF, 0.681 mmol) was added under nitrogen to a solution of intermediate 127 (179 mg, 0.568 mmol) in anhydrous dioxane (2.6 mL) at rt. After 10 min, this solution was added to a solution of intermediate 138 (300 mg, 1.36 mmol) in dioxane (2.6 mL). The mixture was stirred at 80° C. overnight. The mixture was evaporated to dryness and purified by by silica gel column chromatography eluting with methanol in dichloromethane from 2% to 15% to give intermediate 141 (145 mg, yield 49.5%) as a pale yellow solid.


Preparation of Intermediate 142:



embedded image


TFA (1.5 mL, 20.3 mmol) was added to a stirred solution of intermediate 142 (145 mg, 0.281 mmol) in DCM (1.5 mL). After 30 min the reaction was evaporated to dryness, and the crude product was dissolved in MeOH and added to a column loaded with SiliaBond® propylsulfonic acid resin. The column was first eluted with MeOH (10 mL), followed by NH3 in MeOH (7 N, 5 mL). The tubes containing the product were concentrated under reduced pressure to give intermediate 142 (99 mg, yield 84.7%) as a yellow solid.


Preparation of Compound 489:



embedded image


A mixture of intermediate 142 (50 mg, 0.12 mmol) and tert-butyl 4-formylpiperidine-1-carboxylate (51.3 mg, 0.241 mmol) in MeOH (1.20 mL) was stirred for 30 min after which sodium cyanoborohydride (15.1 mg, 0.241 mmol) was added. The reaction mixture was stirred at rt for 2 h, after which it was quenched with water. The mixture was purified on a column loaded with SiliaBond® propylsulfonic acid resin. The column was first eluted with MeOH (10 mL), followed by NH3 in MeOH (7 N, 4 mL). The tubes containing the product were concentrated under reduced pressure to give Compound 489 (64 mg, yield 79%) as a yellow solid.


Preparation of Compound 490:



embedded image


TFA (0.57 mL, 7.40 mmol) was added to a stirred solution of Compound 489 (63 mg, 0.10 mmol) in DCM (0.58 mL). After 30 min the reaction was evaporated to dryness, and the crude product was dissolved in MeOH (2 mL), stirred for 30 min and added to a column loaded with SiliaMetS® Diamine resin, filtered and evaporated to dryness to give Compound 490 (55 mg, quantitative yield) as a yellow solid.


Preparation of Intermediate 147:



embedded image


3,3-difluoropyrrolidine. HCl (0.30 g, 2.1 mmol) was suspended in DCM (10 mL). Next, the mixture was cooled to 0° C. in an ice bath. Then, triethylamine (0.73 mL, 5.2 mmol) was added and the mixture stirred at 0° C. for −5 min. Next, chloroacetylchloride (0.18 mL, 2.2 mmol) was added dropwise. The resulting mixture was stirred at 0° C. for ˜1 h, after which water was added. Then, the mixture was stirred for an additional 5 min, after which it was transferred to a separatory funnel. Next, 1M aq. HCl solution was added and the layers were separated. The organic layer was dried over Na2SO4, filtered and evaporated to dryness to give intermediate 147 as a dark coloured oil (0.29 g, yield 76%).


The Following Intermediate was Synthesized by an Analogous Method as Described for Intermediate 147














Int. No.
Structure
Starting Materials







Intermediate 148


embedded image


3-azabicyclo [3.1.0]hexane.HCl









Preparation of Compound 491:



embedded image


To a mixture of intermediate 27 (70 mg, 0.171 mmol) in NMP (3 mL) was added intermediate 149 (182.7 mg, 0.51 mmol), DIEA (0.088 mL, 0.51 mmol) and potassium iodide (28.4 mg, 0.17 mmol) at rt. Then the mixture continued to stir for 6 h at 80° C. The mixture was diluted by water and extracted with DCM three times. The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by RP silica gel column chromatography eluting with MeCN in water with 0.05% formic acid from 5% to 95% to afford Compound 491 (50 mg, 49% yield) as a yellow oil.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 491














Co No.
Structure
Starting Materials







Compound 492


embedded image


Intermediate 27 & 150





Compound 493


embedded image


Intermediate 28 & 150





Compound 440


embedded image


Intermediate 28 & 149





Compound 495


embedded image


Intermediate 202 & 250





Compound 496


embedded image


Intermediate 202 & 287





Compound 497


embedded image


Intermediate 222 & 225









Preparation of Intermediate 179:



embedded image


To a solution of Compound 501 (70 mg, 0.055 mmol) in methanol (3 mL) and tetrahydrofuran (3 mL) was added 2 M aqueous lithium hydroxide hydrate solution (0.14 mL, 0.274 mmol). The mixture was stirred at room temperature for 1 hour. The mixture was concentrated under reduced pressure. The residue was dissolved in water (10 mL) and washed with ethyl acetate (10 mL) for three times. The combined aqueous phase was acidified with 1 M aq. hydrogen chloride to pH=1 and the precipitate was filtered and dried in vacuo to give intermediate 179, which used directly in the next step.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 179














Int. No.
Structure
Starting Materials







Intermediate 209


embedded image


Compound 498





Intermediate 247


embedded image


Compound 153





Intermediate 248


embedded image


Compound 154





Intermediate 292


embedded image


Compound 499





Intermediate 316


embedded image


Compound 497









Preparation of Intermediate 180:



embedded image


n-Butyllithium (2.5 M in hexane, 2.41 mL, 6.02 mmol) was added dropwise to a solution of 2,2,6,6-tetramethylpiperidine (0.88 g, 6.02 mmol) in tetrahydrofuran (11 mL) under N2 at −40° C., after which the mixture was stirred at −40° C. for an extra 30 min. A solution of bis(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)methane (1.35 g, 5.02 mmol) in tetrahydrofuran (11 mL) was next added dropwise at −78° C. The resulting mixture was stirred at −78° C. for 30 min after which a solution of I-tert-butyl-2-methyl-4-oxopyrrolidine-1-carboxylate (1.0 g, 5.02 mmol) in tetrahydrofuran (11 mL) was added dropwise at −78° C. The mixture was allowed to warm to room temperature and stirred overnight. The mixture was quenched with saturated aq. ammonium chloride solution at 0° C. and stirred for an extra hour at 0° C. The precipitate was removed by filtration and the filtrate diluted with water and ethyl acetate. Phases were separated and the water layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The crude product was purified by silica column chromatography eluting with ethyl acetate in heptane from 0% to 10% to give intermediate 180 (958 mg, yield 59%).


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 180














Int. No.
Structure
Starting Materials







Intermediate 181


embedded image


(S)-tert-butyl-2-methyl-4- oxopyrrolidine-1-carboxylate





Intermediate 197


embedded image


Intermediate 196





Intermediate 228


embedded image


t-butyl 4-oxoazepnae-1- carboxylate





Intermediate 301


embedded image


t-butyl 2-methyl-3- oxopyrrolidine-1- carboxylate









Preparation of Intermediate 182



embedded image


A reaction flask was consecutively charged with intermediate 6 (921 mg, 2.28 mmol), dioxane (7.1 mL), water (0.9 mL), intermediate 180 (958 mg, 2.96 mmol), cesium carbonate (1.49 g, 4.56 mmol) and 1,1′-bis(diphenylphosphino)ferrocene dichloropalladium(II) dichloromethane complex (186 mg, 0.23 mmol), degassed and refilled with nitrogen. The resulting mixture was stirred at 100° C. for 5 h. The reaction mixture was diluted with water and ethyl acetate. Phases were separated and the water layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 4% to give intermediate 182 (1.08 g, yield 87%) as a foam.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 182














Int. No.
Structure
Starting Materials







Intermediate 183


embedded image


Intermediate 6 & 181





Intermediate 198


embedded image


Intermediate 6 & 197





Intermediate 229


embedded image


Intermediate 6 & 228





Intermediate 234


embedded image


Intermediate 6 & 233





Intermediate 302 Intermediate 302a & Intermediate 302b


embedded image


Intermediate 6 & 301 Intermediate 302 was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as intermediate 302a & the second fraction as intermediate 302b.








embedded image











embedded image











Preparation of Intermediate 184 & 185:



embedded image


To a mixture of intermediate 182 (1.08 g, 2.07 mmol) in MeOH (100 mL) was added a catalytic amount of Pd/C (10% w:w) (221 mg, 0.207 mmol) and the solution was stirred under H2 atmosphere overnight. Then, the mixture was filtered over Celite®, the Celite® washed with MeOH. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 5% to give the mixture of diastereomers (1030 mg, yield 91%) as a white foam, which were separated by chiral prep SFC (Stationary phase: Chiralpak Diacel AD 20×250 mm, Mobile phase: CO2, iPrOH+0.4 iPrNH2) to give intermediate 184 (192 mg, yield 18%) and intermediate 185 (551 mg, yield 51%). The absolute configuration was determined by NMR.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 184 & 185














Int. No.
Structure
Starting Materials







Intermediate 186


embedded image


Intermediate 183





Intermediate 187


embedded image


Intermediate 183





Intermediate 199


embedded image


Intermediate 198





Intermediate 230


embedded image


Intermediate 229





Intermediate 235


embedded image


Intermediate 234





Intermediate 303a & Intermediate 303b


embedded image


Intermediate 302a The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IC 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as intermediate 303a & the second fraction as intermediate 303b.








embedded image








Intermediate 304a & Intermediate 304b


embedded image


Intermediate 302b The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IC 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as intermediate 304a & the second fraction as intermediate 304b.








embedded image











Preparation of Intermediate 192:



embedded image


A reaction flask was consecutively charged with tert-butyl carbamate (4.6 g, 39.0 mmol), sodium benzenesulfinate (9.6 g, 58.5 mmol), THF (16 mL), water (39 mL), 2-(tetrahydro-2H-pyran-4-yl)acetaldehyde (5.0 g, 39.0 mmol) and formic acid (10.3 mL, 273.1 mmol). The reaction mixture was stirred for 4 days at r.t. The precipitate was isolated by filtration, washed with water and dried in a vacuum oven at 50° C. to give intermediate 192 (10.9 g, yield 76%) as a white fluffy solid.


Preparation of Intermediate 193:



embedded image


To a solution of allyl acetoacetate (5.0 g, 35.2 mmol), 4-acetamidobenzenesulfonyl azide (9.3 g, 38.7 mmol) in MeCN (176 mL) at 0° C. was added dropwise Et3N (9.8 mmol, 70.3 mmol). The reaction mixture was allowed to warm to room temperature and stirred for 3 h. The solvent was removed under reduced pressure. The residue was suspended in diethyl ether and the solid (4-acetamidobenzenesulfonamide) was removed by filtration. The filtrate was concentrated under reduced pressure and the crude product purified by silica gel column chromatography eluting with ethyl acetate in heptane from 0% to 20% to give intermediate 193 (4.9 g, yield 83%) as a yellow oil.


Preparation of Intermediate 194:



embedded image


NaH (758 mg (60% dispersion in mineral oil), 18.9 mmol) was added portionwise to a solution of intermediate 192 (3185 mg, 18.9 mmol) in THF (80 mL) at r.t., after which stirring was continued for 20 min. Simultaneously Li-HMDS (18.9 mL, 1M in THF, 18.9 mmol) was added to a solution of intermediate 193 (3186 mg, 18.9 mmol) in THF (80 mL) at −78° C. The reaction mixture was stirred for 10 min at −78° C. after which the above reaction solution was added. The resulting reaction mixture was stirred for an additional 60 min after which it was quenched with 10 M acetic acid in THF. The mixture was warmed to room temperature and partitioned between EtOAc and water. The organic layer was separated, washed with water and brine, dried (MgSO4), filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 2% to give intermediate 194 (3.44 g, yield 46%).


Preparation of Intermediate 195:



embedded image


A mixture of intermediate 194 (500 mg, 1.26 mmol) and Rh2(OAc)4 (14 mg, 0.03 mmol) in DCM (30 mL) was stirred under a nitrogen atmosphere at rt for 2 hr. The reaction mixture was transferred as such to be purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 2% to give intermediate 195 (262 mg, yield 57%) as a yellow oil.


Preparation of Intermediate 196:



embedded image


Pd(PPh3)4 (8 mg, 0.007 mmol) and morpholine (750 mg, 8.61 mmol) were added to a solution of intermediate 195 (2110 mg, 5.74 mmol) in THF (136 mL) and stirred at room temperature overnight. The reaction mixture was concentrated to give a crude product which was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 2% to give intermediate 196 (1.2 g, yield 74%).


Preparation of Intermediate 201—Method A:



embedded image


Into a 2 L 4-necked round-bottom flask were added THF (345 mL) and Zn (120.87 g, 1847.90 mmol, 5.00 equiv) at 30° C. under a nitrogen atmosphere. A solution of TMSCl (8.03 g, 73.91 mmol, 0.2 equiv) and 1-bromo-2-chloroethane (10.60 g, 73.91 mmol, 0.20 equiv) in THF (230 mL) were added into above round-bottom flask with a Lead Fluid-BT100F peristaltic pump (rate: 10 mL/min) under a nitrogen atmosphere. The resulting mixture was stirred for additional 40 min at 30° C. Next, a Lead Fluid-BT100F peristaltic pump was used to remove the solvent in above RBF quickly, and then fresh THF (575 mL) was re-charged under a nitrogen atmosphere. The mixture was heated to 60° C. Next, a solution of tert-butyl (3R)-3-(iodomethyl)pyrrolidine-1-carboxylate (115 g, 369.58 mmol, 1.00 equiv) in THF (575 mL) was added into above RBF with a Lead Fluid-BT100F peristaltic pump (rate: 15.0 mL/min) under a nitrogen atmosphere (temperature rises to 60-65° C.). The solution was stirred at 60° C. for an additional 1 h. The mixture was then cooled to 30° C. and allowed to stand for 1 h. The solution of {[(3R)-1-(tert-butoxycarbonyl)pyrrolidin-3-yl]methyl}(iodo)zinc was used directly in the next step. The concentration of the product was about 0.37 moL/L in THE Into a 2 L 4-necked round-bottom flask were added intermediate 6 (105 g, 259.71 mmol, 1.00 equiv) and THF (500 mL) at 30° C. under nitrogen atmosphere. To the stirred solution was added the 4th Generation RuPhos Pd precatalyst (5.65 g, 6.49 mmol, 0.025 equiv) under nitrogen atmosphere. Next, the solution of {[(3R)-1-(tert-butoxycarbonyl)pyrrolidin-3-yl]methyl}(iodo)zinc was added with a Lead Fluid-BT100F peristaltic pump into the 2 L 4-need RBF quickly under a nitrogen atmosphere (the excess zinc dust was not transferred). The resulting mixture was stirred for an additional 16 h at 50° C. The reaction was repeated 6 times in parallel. The reaction was quenched by the addition of aqueous sat. NH4Cl solution (12 L). The aqueous layer was extracted with EtOAc (3×6 L), the organic layer was washed with water (2×3 L) and brine (1×3 L). The resulting mixture was dried with Na2SO4 and concentrated under reduced pressure. The crude product as a black oil (1100 g, crude) was used directly into the next step (preparation of intermediate 202)


Alternatively, the Procedure Described Below can be Employed for the Preparation of Intermediate 201—Method B

A column (1.5 cm×15 cm) was stoppered with cotton wool and filled with granular zinc (20-mesh), 22 g. The column volume of the filled column was determined by measuring the time for THF to fill the column at 1 m/min flow rate. Column volume=4.3 mL. The zinc was activated by flowing a strong activating solution through the column at 0.5 mL/min for 10 mins. The strong activating solution consists of 1 mL TMSCl (0.67 M) & 0.75 mL chlorobromoethane (0.71 M) in 10 mL THF. After activation, the column was washed with dry THF: 10 mL, 1 ml/min. tert-butyl (R)-3-(iodomethyl)pyrrolidine-1-carboxylate (10 g, 37 mmol) was dissolved in THF (60 mL). The iodide solution was flowed through the activated zinc column at 50° C., flow rate 0.45 mL/min. After reaction: titration with iodine shows a concentration of 0.30 M.


Intermediate 6 (1.2 g, 2.4 mmol) was added with RuPhos Pd G4 (0.051 g, 0.06 mmol) in a sealed vial with a stirring bar in a glove box. Then, a solution of freshly made R-((1-(tert-butoxycarbonyl)-3-yl)methyl)zinc(II) iodide (12 mL, 0.3 M, 3.6 mmol) which was prepared by the above procedure was added. Next, the solution was heated to 50° C. under nitrogen atmosphere during 16 h. The solution was concentrated in vacuo and the residue redissolved in DCM. Next, water was added, followed by aq. Na4EDTA solution (pH>10). The layers were separated and the water layer was extracted once more with DCM. Organic layers were combined, dried over Na2SO4, filtered and evaporated to dryness. The residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 000 to 10% to give intermediate 201 (1.4 g, 1.5 mmol (55% purity), 63& yield).


The Following Intermediates were Synthesized by an Analogous Method (Method B) as Described for Intermediate 201














Int. No.
Structure
Starting Materials







Intermediate 212


embedded image


Intermediate 6 & tert-butyl (S)- 3-(iodomethyl)piperidine-1- carboxylate





Intermediate 214


embedded image


Intermediate 6 & tert-butyl (R)- 3-(iodomethyl)piperidine-1- carboxylate





Intermediate 224


embedded image


Intermediate 6 & tert-butyl (S)- 3-(iodomethyl)pyrrolidine-1- carboxylate





Intermediate 318


embedded image


Intermediate 317 & tert-butyl (R)-3-(bromomethyl)pyrrolidine- 1-carboxylate





Intermediate 322


embedded image


intermediate 6 & tert-butyl 3- (bromomethyl)-3- methylpyrrolidine-1-carboxylate





Intermediate 337


embedded image


Intermediate 7 & tert-butyl (R)- 3-(bromomethyl)pyrrolidine-1- carboxylate





Intermediate 341


embedded image


Intermediate 7 & tert-butyl (S)- 3-(bromomethyl)pyrrolidine-1- carboxylate





Intermediate 346


embedded image


Intermediate 345 & tert-butyl (R)-3-(bromomethyl)pyrrolidine- 1-carboxylate





Intermediate 353


embedded image


Intermediate 352 & tert-butyl (R)-3-(bromomethyl)pyrrolidine- 1-carboxylate





Intermediate 357


embedded image


Intermediate 352 & tert-butyl (S)-3-(bromomethyl)pyrrolidine- 1-carboxylate





Intermediate 367


embedded image


Intermediate 361 & tert-butyl (R)-3-(iodomethyl)pyrrolidine- 1- carboxylate









Preparation of Intermediate 202:



embedded image


The mixture of intermediate 201 (17 g, 33.09 mmol) in dichloromethane (50 mL), was added the solution 24 mL of chlorine hydride (7 M in ethyl acetate). After stirring at r.t. for 5 h, the reaction mixture was concentrated, and the residue was diluted with DCM and basified with sodium hydroxide aqueous solution (1M) to pH˜ 10. The layers were separated and the aqueous layer was extracted with DCM three times and the combined organic layer was washed with brine (30 mL), dried over sodium sulfate, filtered and concentrated to afford intermediate 202 (13 g, 31.1 mmol, 94.2% yield) as a yellow solid, which was used in the next step without purification.


Alternatively, intermediate 202 can also be prepared as a 0.2TFA salt by using the following procedure:


Intermediate 201 (5.2 g, 6.95 mmol, 68% pure) is dissolved in DCM (44.5 mL) and TFA (5.3 mL) was added and stirred for 4 h at rt. The solution was concentrated in vacuo and coevaporated with toluene. Next, the mixture was washed with 1M NaOH and extracted four times with 10 DCM and EtOAc and Me-THF to obtain the combined organics which were then dried with anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified via by silica gel column chromatography eluting with methanol (containing 7N NH3) in dichloromethane from 0% to 10% to give intermediate 202 as a 0.2TFA salt.


Alternatively, intermediate 202 can also be prepared with the following procedure:


Into a 10 L 4-necked round-bottom flask were added 4N HCl in 1,4-dioxane (1.8 L). Then, crude intermediate 201 in THF (3 L) was added dropwise (calculated by 735 g intermediate 201, 1.82 mol, 1.0 equiv) at 0° C. The resulting mixture was stirred for an additional 2 h at 0° C. The resulting mixture was diluted with ethyl acetate (3 L) and water (3 L). The aqueous layer was washed with DCM (10×1 L). The pH of the aqueous layer was adjusted to pH 8 with saturated aqueous Na2CO3 solution and extracted with CH2Cl2 (4×2 L). The organic layers were dried with Na2SO4 and concentrated under vacuum to afford intermediate 202 (389 g, yield 53% over 2 steps) as a light yellow solid.


The Following Intermediate were Synthesized by an Analogous Method as Described for Intermediate 707














Int. No.
Structure
Starting Materials







Intermediate 225


embedded image


Intermediate 224





Intermediate 368


embedded image


Intermediate 367









Preparation of Intermediate 203:



embedded image


A stir bar, 4,4′-di-tert-butyl-2,2′-bipyridine (69.6 mg, 0.259 mmol), DME (40 mL), nickel(II) chloride ethylene glycol dimethyl ether complex (65.2 mg, 0.297 mmol) were added to 40 mL glass bottle, the mixture was purged with argon for 15 min, then intermediate 6 (1 g, 2.474 mmol), tert-butyl 3-(bromomethyl)-3-methylazetidine-1-carboxylate (1.3 g, 4.921 mmol), Ir[dF(CF3)ppy]2(dtbpy))PF6 (282.6 mg, 0.252 mmol), sodium carbonate (782.6 mg, 7.384 mmol) and tris(trimethylsilyl)silane (1.3 mL, 4.214 mmol, 0.806 g/mL) were added to the mixture, the mixture was purged with argon for 15 min. The vial was sealed with parafilm and irradiated with blue light for 12 hours. The reaction mixture was diluted with dichloromethane (50 mL) and the saturated solution of sodium bicarbonate (50 mL) was added, the mixture was extracted with dichloromethane (40 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue which was purified by preparative-HPLC (Column: Boston Uni C18 40*150*5 um, Mobile Phase A: water, Mobile Phase B: acetonitrile, Flow rate: 60 mL/min, gradient condition from 30% B to 60% B). The pure fractions were collected and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give intermediate 203 (380 mg, 92.2% purity, 21.9% yield).


Preparation of Compound 498:



embedded image


A stir bar, intermediate 25 (300 mg, 0.760 mmol), MeCN (3 mL), intermediate 207 (230 mg, 0.919 mmol), potassium carbonate (318 mg, 2.30 mmol) and potassium iodide (252 mg, 1.52 mmol) were added into a 8 mL glass. The reaction mixture was heated and stirred at 100° C. for 2 h under microwave irradiation. The reaction mixture was filtered through a pad of Celite®, the filter cake was washed with MeCN (5 mL×5). The combined filtrates were concentrated under reduced pressure to give the crude product which was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 9% to give Compound 498 (270 mg, 43.5% purity, 28.1% yield) as yellow solid.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 498














Co No.
Structure
Starting Materials







Compound 499


embedded image


Intermediate 25 & 290









Preparation of Intermediate 211:



embedded image


A mixture of Compound 92 (53 mg, 0.097 mmol) and iodine (2.5 mg, 0.01 mmol) in acetone (1.2 mL) was stirred at refluxing temperature (56° C.) for 10 min. The mixture was evaporated to dryness, and the crude was purified by silica gel column chromatography eluting with methanol (+1% NH3 (7N) in methanol) in dichloromethane from 1% to 10% to give intermediate 211 (35 mg, yield 58.9%) as a white solid.


The Following Intermediate was Synthesized by an Analogous Method as Described for Intermediate 211














Int. No.
Structure
Starting Materials







Intermediate 264


embedded image


Compound 407









Preparation of Intermediate 216:



embedded image


To a solution of (S)-tert-butyl 2-(hydroxymethyl)piperidine-1-carboxylate (400 mg, 1.86 mmol) in dichloromethane (8 mL) was added triethylamine (376 mg, 3.72 mmol) and methanesulfonyl chloride (277 mg, 2.42 mmol) at 0° C. The mixture was stirred at 0° C. for 60 minutes. The reaction was quenched with water and the mixture was diluted with dichloromethane, washed with 0.5M HCl (aq.), dried over Na2SO4, and concentrated to give intermediate 216 (385 mg, 17.5% purity from LCMS, 12.3% yield) as yellow oil which was used directly in the next step without further purification.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 216














Int. No.
Structure
Starting Materials







Intermediate 222


embedded image


cis-methyl 4- (hydroxymethyl)cyclo- hexanecarboxylate





Intermediate 223


embedded image


trans-methyl 4- (hydroxymethyl)cyclo- hexanecarboxylate





Intermediate 260


embedded image


(R)-tert-butyl 2- (hydroxymethyl)piperi- dine-1-carboxylate









Preparation of Intermediate 221:



embedded image


To a solution of cyclopropylamine (2 g, 33.3 mmol) in dichloromethane (25 mL) in an ice water bath was added triethylamine (10.1 g, 99.8 mmol) and phenyl chloroformate (5.2 g, 33.3 mmol) in five portions. The reaction mixture was stirred at room temperature for 2 hours. It was poured into water and extracted with dichloromethane (30 mL) twice. The organic layer was washed with brine (30 mL), dried over sodium sulfate, filtered and concentrated to afford the crude product, which was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 20% to give intermediate 221 (4.22 g, 98% purity, 70.1% yield) as a white solid.


Preparation of Intermediate 233:



embedded image


2,2,6,6-tetramethylpiperidine (3.90 g, 27.6 mmol) was dissolved in THF (50 mL) and cooled to −30° C. under N2 atmosphere. n-BuLi (12.0 mL, 30.0 mmol, 2.5 M in n-Hexane) was added dropwise, and the reaction mixture was stirred at the same temperature for 30 minutes. Next, the reaction mixture was cooled to −78° C., and a solution of 2,2′-(ethane-1,1-diyl)bis(4,4,5,5-tetramethyl-1,3,2-dioxaborolane) (6.00 g, 21.3 mmol) in THF (30.0 mL) was added dropwise at −78° C. After stirring for 30 min, a solution of 1-boc-3-azetidinone (4.40 g, 25.7 mmol) in THF (40 mL) was added dropwise at −78° C. The reaction mixture was warmed to 25° C. slowly and stirred at 25° C. for 12 hours. The reaction mixture was cooled to 0° C. and quenched with aq. NH4Cl solution (30 mL). After additional stirring for 10 minutes, the resulting mixture was concentrated under reduced pressure to remove THF, the residue was extracted with ethyl acetate (40 mL×2), and the organic layers were washed with brine (50 mL×1), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The crude was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 0% to 6% to give intermediate 233 (4.00 g, 70% purity, 42.56% yield) as a colorless liquid.


Preparation of Compound 500:



embedded image


To a mixture of intermediate 202 (200 mg, 0.49 mmol) in EtOH (4 mL) and H2O (0.4 mL) was added tert-butyl 1-oxa-6-azaspiro[2.5]octane-6-carboxylate (114.9 mg, 0.539 mmol) and TEA (49.6 mg, 0.49 mmol). The mixture was stirred at 25° C. for 12 hours. The mixture was neutralized with aqueous Na2CO3 (10 mL), poured into H2O (20 mL) and extracted with DCM (3×20 mL). The combined organic layer was dried over anhydrous Na2SO4 which was purified by preparative-HPLC (Column: Welch Xtimate C18 150*30 mm*5 um, Mobile Phase A: water (NH3H2O+NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 35 mL/min, gradient condition from 47% B to 77% B). The pure fractions were collected and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give Compound 500 (100 mg, 30.94% yield) as a white powder.


Preparation of Intermediate 258:



embedded image


1-(tetrahydro-2H-pyran-4-yl)ethan-1-one (2.5 g, 19.5 mmol) was dissolved in MeOH (39.5 mL) and NBS (3471.6 mg, 19.5 mmol) was added and the solution was stirred for 3 hours at 50° C. The reaction mixture was concentrated in vacuo and redissolved in DCM and washed with water three times. The combined organics were dried and purified by silica gel column chromatography eluting with 30% ethyl acetate in heptane to give intermediate 258 (2.4 g, 59% yield).


The Following Intermediate was Synthesized by an Analogous Method as Described for Intermediate 258














Int. No.
Structure
Starting Materials







Intermediate 265


embedded image


1-(4-acetylpiperidino)ethan- 1-one









Preparation of Intermediate 259:



embedded image


Intermediate 258 (100 mg, 0.483 mmol) was dissolved in DMF (3.7 mL) and KOAc (142.19 mg, 1.449 mmol) was added and stirred for 4 hr at rt. The solution was extracted with EtOAc and washed with brine, and the combined organic layers were dried over Na2SO4 anhydrous, concentrated in vacuo and purified by silica gel column chromatography eluting with ethyl acetate in heptane from 0% to 100% to give intermediate 259 (65 mg, 72% yield) as an oil.


_Preparation of Intermediate 266:



embedded image


Intermediate 265 (0.50 g, 1.0 mmol, 50% purity) was dissolved in MeOH (12 mL), after which sodium formate (0.41 g, 6.0 mmol) was added. The resulting solution was heated at 55° C. overnight, after which it was evaporated to dryness. The residue was suspended in DCM and purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 9% to give intermediate 266 (0.16 g, 0.78 mmol, 77% yield).


Preparation of Intermediate 274:



embedded image


1-bromo-3-chloropropane (0.37 mL, 3.76 mmol) was added to a stirred suspension of 2,5-difluorobenzenethiol (0.50 g, 3.42 mmol) and K2CO3 (0.61 g, 4.4 mmol) in anhydrous DMF (6.6 mL) and the mixture was left under stirring for overnight at rt. The mixture was diluted with water and extracted with EtOAc (×3). Reunited organic phases were washed with water (×2), brine (×1), dried over anhydrous sodium sulfate, filtered and evaporated to dryness to give intermediate 274 (951 mg, yield 93.6%) as a colorless oil. The desired product was used in the next step without further purification.


Preparation of Intermediate 275:



embedded image


Iodobenzene diacetate (1.14 g, 3.54 mmol) was added to a solution of intermediate 274 (0.5 g, 1.684 mmol) and ammonium carbamate (0.276 g, 3.54 mmol) in MeOH (3.4 mL) at r.t. and the reaction mixture was stirred at r.t. overnight. The reaction mixture was diluted with water and extracted with DCM (×3). Reunited organic phases were dried over sodium sulfate, concentrated under reduced pressure and purified by silica gel column chromatography eluting with ethyl acetate in heptane from 10% to 100% to give intermediate 275 (281 mg, yield 65.7%) as a pale yellow oil.


Preparation of Intermediate 276:



embedded image


NH3 (0.1% in H2O, 4.3 mL) was added to intermediate 275 (288 mg, 1.13 mmol) in MeOH (0.5 mL) into a microwave vial, which was sealed and heated at 80° C. for 5 h. The solvent reaction was cooled down at rt, quenched with NaOH 1N, and extracted with EtOAc (×3). Reunited organic phases were washed with water, brine, dried over anhydrous sodium sulfate, filtered, and evaporated to dryness to afford intermediate 276 (213 mg, yield 86.4%) as a colorless oil.


Preparation of Intermediate 299:



embedded image


Intermediate 265 (0.50 g, 0.79 mmol) was dissolved in acetone (15 mL), after which, NaN3 was added (0.16 g, 2.4 mmol). The mixture was stirred 50° C. for 1 h, after which the mixture was cooled to ambient temperature. Then, the mixture was filtered and evaporated to dryness. The residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 10% to give intermediate 299 (510 mg, 2.4 mmol).


Preparation of Intermediate 300:



embedded image


Intermediate 299 (0.40 g, 1.95 mmol) was dissolved in THF (20 mL), after Ac2O (0.18 mL, 2.0 mmol) and trimethylphosphine in THF (1M solution, 3.9 mL, 3.9 mmol) were added. The mixture was stirred at ambient temperature for 3 h. Then, MeOH was added and the mixture stirred at ambient temperature for ˜5 min. Next, the mixture was evaporated to dryness and the residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 8% to give intermediate 300 (0.24 g, yield: 54%).


Preparation of Intermediate 324:



embedded image


Intermediate 258 (320 mg, 1.54 mmol) in MeCN (3.2 mL) was treated sequentially with K2CO3 (640.7 mg, 4.6 mmol) and dimethylamine (2.3 mL, 2 M, 4.6 mmol). After stirring overnight at room temperature, the mixture was charged with aqueous 1N NaOH (2 mL), and the layers were separated. The aqueous layer was extracted with EtOAc (2×5 mL). The combined organic layers were dried over Na2SO4 anhydrous and concentrated under reduced pressure. The crude oil was further purified by silica gel column chromatography eluting with ethyl acetate (containing 25% EtOH) in heptane from 0% to 100% to give intermediate 324 as an oil (199 mg, 75% yield).


Preparation of Intermediate 327:



embedded image


To a solution of tert-butyl 4-(methoxy(methyl)carbamoyl)piperidine-1-carboxylate (2.5 g, 9.18 mmol) in tetrahydrofuran (30 ml) at 0° C. was added ethylmagnesium chloride (3.26 g, 36.7 mmol) and the resulting suspension was allowed to stir at room temperature for 4 hrs. After stirring at room temperature for 12 hours, the mixture was diluted with EtOAc, washed with sat. NH4Cl and concentrated. The residue was purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 20% to 100% to give intermediate 327 (3.1 g, yield: 83%).


Preparation of Intermediate 332:



embedded image


A solution of tetrahydro-2H-pyran-4-carbaldehyde (4 g, 33.29 mmol) in THF (20 mL) was dropwise vinylmagnesium bromide (67 mL) for 30 min at 0° C. The mixture was stirred at room temperature overnight. The mixture were quenched with 20 mL of NH4Cl (aq) at 0° C. and extracted with EtOAc (20 mL*3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give the crude product, which was purified by silica gel column chromatography eluting with EtOAc in petroleum ether from 0% to 50% to give intermediate 332 (3 g, 57.0% yield) as a colourless oil.


Preparation of Intermediate 333:



embedded image


To a solution of intermediate 332 (3 g, 20.04 mmol) in dichloromethane (50 mL) was added Dess-Martin Periodinane (13.28 g, 30.06 mmol) at 0° C. After stirring at 20° C. for 5 h, the mixture was basified to pH 7-8 with saturated sodium bicarbonate aqueous solution and extracted with DCM (30 mL) for three times. The combined organic layers were washed with brine (30 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and purified by silica gel column chromatography eluting with 20% ethyl acetate in petroleum ether to give the intermediate 333 (1.5 g, 48.05% yield) as a yellow oil.


Preparation of Intermediate 334:



embedded image


To a solution of intermediate 333 (500 mg, 3.21 mmol) in methanol (10 mL), was added sodium carbonate (aq.) (6.4 mL, 6.4 mmol) at rt. After stirring at rt for 18 h. The reaction mixture was quenched with H2O (10 mL) and extracted with DCM. The combined organic phase was washed with brine, dried by Na2SO4, filtered and concentrated and purified by silica gel column chromatography eluting with 10% ethyl acetate in petroleum ether to give intermediate 334 (400 mg, 57.8% yield) as a yellow oil.


Preparation of Intermediate 335:



embedded image


To a solution of intermediate 333 (1 g, 6.42 mmol) in H2O/MeCN (20 mL/5 mL), was added chromium(II) chloride (204 mg, 1.28 mmol) at rt. After stirring at 80° C. for 18 h, the reaction mixture was quenched with H2O (10 mL) and extracted with DCM. The combined organic phase was washed with brine, dried by Na2SO4, filtered and concentrated and purified by silica gel column chromatography eluting with ethyl acetate in petroleum ether from 50% to 100% to give intermediate 335 (800 mg, 63% yield) as a yellow oil.


Preparation of Intermediate 361:



embedded image


Intermediate 4 (3.3 g, 9.451 mmol) was dissolved in MeOH (38.2 mL) and cooled to 0° C. before thionyl chloride (13.7 mL, 189.0 mmol) was added dropwise. The solution was then heated to 70° C. for 2 hours. After cooling to ambient temperature, the solution was concentrated in vacuo and directly purified by silica gel column chromatography eluting with methanol (containing 7N NH3) in dichloromethane from 0% to 10% to give intermediate 361 (3.7 g, 100% yield) as an oil.


Preparation of Intermediate 366:



embedded image


Compound 527 (1.8 g, 3.66 mmol, 92% pure) was dissolved in THF (29.8 mL) and water (6.62 mL) and LiOH (175.6 mg, 7.3 mmol) was added. The solution was stirred at r.t. for 16 hours until full conversion. The solution was concentrated till dryness, then co-evaporated with tolunene till dryness to obtain intermediate 366 as lithium salt with 1 eq LiOH as excess as a solid (1.7 g, 90% yield).


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 366














Int. No.
Structure
Starting Materials







Intermediate 370


embedded image


Compound 528





Intermediate 374


embedded image


Compound 516





Intermediate 376


embedded image


Compound 517





Intermediate 380


embedded image


Compound 518





Intermediate 382


embedded image


Intermediate 381





Intermediate 384


embedded image


Compound 521





Intermediate 388


embedded image


Compound 519





Intermediate 390


embedded image


Compound 533





Intermediate 392


embedded image


Compound 534









Preparation of Compound 516:



embedded image


Compound 530 (641 mg, 1.2 mmol) was dissolved in MeCN (2.4 mL) and DIPEA (3.3 mL, 19.15 mmol) and isobutyryl chloride (1279 mg, 12 mmol) was added. The resulting mixture was stirred at rt for 16 h. Afterwards, the crude mixture was diluted with DCM and washed with water. The organic phase was dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude mixture was purified using silica gel column chromatography eluting with ethyl acetate in heptane from 0% to 100% to afford Compound 516 (454 mg, 71% yield).


The Following Intermediates were Synthesized by an Analogous Method as Described for Compound 516














Int. No.
Structure
Starting Materials







Compound 517


embedded image


Compound 350 & methyl chloroformate





Compound 518


embedded image


Intermediate 378 & acetyl chloride





Compound 519


embedded image


Intermediate 386 & acetyl chloride









Preparation of Compound 520:



embedded image


Compound 530 (870 mg, 0.945 mmol) is dissolved in DMF (7.3 mL) and DIPEA (0.97 mL, 5.67 mmol) and 2-hydroxy-2-methyl-propanoic acid (118.0 mg, 1.13 mmol) then HATU (538.9 mg, 1.4 mmol) are added and stirred for 2 hours at rt. The solution is extracted with EtOAc and washed three times with water (50 mL) and the combined organics are dried with MgSO4 anhydrous, filtered and concentrated in vacuo. The crude was further purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 10% to give Compound 520 (450 mg, 85% yield).


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 520














Co




No.
Structure
Starting Materials







Compound 521


embedded image


Compound 530 & 2-cyano-2- methylpropanoic acid









Preparation of Intermediate 1:



embedded image


To a solution of intermediate 26 (300 mg, 98% purity, 0.72 mmol) in methanol (0.7 mL) were added intermediate 86 (457 mg, 95% purity, 2.16 mmol), sodium cyanoborohydride (136 mg, 2.16 mmol) and zinc chloride (294 mg, 2.16 mmol). The reaction mixture was heated up to 68° C. and stirred at this temperature overnight. After cooled down to r.t., the reaction mixture was concentrated and the residue was purified by prep. HPLC (column: SunFire C18 150*19 mm*5 um, Mobile Phase A: water (0.1% TFA), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 13% B to 20% B)). The collected fraction was lyophilized and the residue was basified with sodium hydroxide aqueous solution (1M), extracted with dichloromethane (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated to afford the free base of Compound 1 (120 mg, 99% purity, 25% yield) as a yellow solid. A solution of the free base (38 mg) and fumaric acid (12.6 mg) in water (5 mL) was freeze dried to give Compound 1 (50 mg, fumarate, 99.4% purity) as a yellow solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 1














Compound No.
Structure
Starting Materials







Compound 2


embedded image


Intermediate 27 & 86





Compound 2a


embedded image


Compound 2 (free base) was separated by chiral Prep. HPLC (separation condition: Column: Chiralpak AD-H, Column size: 0.46 cm I.D. × 15 cm L; Mobile Phase: Hexane:EtOH:DEA = 95:5:0.1, at 1 mL/min; Temp: 35° C.; Wavelength: 254 nm) and the first fraction was collected





Compound 3


embedded image


Intermediate 31 & 86





Compound 4


embedded image


Intermediate 32 & 86





Compound 4a


embedded image


Compound 4 (free base) was separated by SFC (column: DAICEL CHIRALPAK AD (2.5 cm I.D. × 25 cm L, 5 μm), eluent: supercritical CO2 in Hexane/EtOH/DEA = 90/10/0.1 (V/V/V))) and the first fraction was collected





Compound 4b


embedded image


Compound 4 (free base) was separated by SFC (column: DAICEL CHIRALPAK AD (2.5 cm I.D. × 25 cm L, 5 μm), eluent: supercritical CO2 in Hexane/EtOH/DEA = 90/10/0.1 (V/V/V))) and the second fraction was collected





Compound 6


embedded image


4-fluoro-1H-pyrrolo[2,3- c]pyridine





Compound 7


embedded image


4-(trifluoromethyl)-1H- pyrrolo[2,3-c]pyridine









Preparation of Compound 8:



embedded image


To a solution of intermediate 72 (100 mg, 98% purity, 0.188 mmol) in methanol (2 mL) were added 1-(piperazin-1-yl)ethanone (48.2 mg, 0.376 mmol) and acetic acid (0.05 mL). The reaction mixture was stirred at room temperature for 30 minutes. Then sodium cyanoborohydride (23.6 mg, 0.376 mmol) was added into the mixture. After stirring at r.t. for 2 hours, the reaction mixture was basified with saturated NaHCO3 aqueous solution and extracted with dichloromethane (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated to afford the crude product, which was purified by Prep. HPLC (Column: SunFire C18 150*19 mm*5 um, Mobile Phase A: water (0.1% NH40Ac), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 10% B to 50% B) to give Compound 8 (100 mg, 99% purity, 83.1% yield) as a yellow gum.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 8


Alternatively, purification can also be performed using the following method: Prep. HPLC method (Column Welch Xtimate C18 150*25 mm*5 um, Mobile Phrase A: water (0.225% formic acid), mobile phase B: acetonitrile, Flow rate 25 mL/min, gradient condition from 1% B to 31% B).
















Compound No.
Structure
Starting Materials








Compound 8a


embedded image


Intermediate 72a & 1-(piperazin-1-yl)ethanone






Compound 8b


embedded image


Intermediate 72b & 1-(piperazin-1-yl)ethanone






Compound 9


embedded image


Intermediate 73 & 1-(piperazin-1-yl)ethanone






Compound 9b


embedded image


Intermediate 73b & 1-(piperazin-1-yl)thanone






Compound 10


embedded image


Intermediate 72 & 4-(methylsulfonyl)piperidine






Compound 11


embedded image


Intermediate 73 & 4-(methylsulfonyl)piperidine






Compound 12


embedded image


Intermediate 72 & 89






Compound 13


embedded image


Intermediate 73 & 89






Compound 14


embedded image


Intermediate 72 & 2-methoxyethanamine






Compound 15


embedded image


Intermediate 73 & 2-methoxyethanamine






Compound 16


embedded image


Intermediate 74 & 89






Compound 17


embedded image


Intermediate 75 & 89






Compound 18a


embedded image


Compound 371 & intermediate 89






Compound 18b


embedded image


Compound 372 & intermediate 89






Compound 19a


embedded image


Compound 374 & intermediate 89






Compound 19b


embedded image


Compound 75 & intermediate 89









Preparation of Intermediate 9a:



embedded image


To a solution of intermediate 73a (300 mg, 0.576 mmol) and 1-(piperazin-1-yl)ethanone (148 mg, 1.16 mmol) in anhydrous methanol (5 mL) was added acetic acid (69.2 mg, 1.15 mmol). The reaction mixture was heated up to 45° C. and stirred at this temperature for 30 minutes before the addition of sodium cyanotrihydroborate (72.4 mg, 1.15 mmol). After stirring at 45° C. for another 12 hours, the reaction mixture was cooled down to room temperature, diluted with dichloromethane (40 mL), basified to pH=8 with the saturated solution of sodium bicarbonate (30 mL) and extracted with dichloromethane (20 mL×3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by preparative-HPLC (Column: Boston Green ODS 150*30 mm*5 um, Mobile Phase A: water (0.225% FA), Mobile Phase B: acetonitrile, Flow rate: 35 mL/min, gradient condition from 1% B to 30% B). The pure fractions were collected, and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give Compound 9a (200 mg, 98.7% purity, 47.3% yield) as a yellow solid.


Preparation of Compound 20:



embedded image


At 0° C., to a solution of intermediate 89 (56.2 mg, 90% purity, 0.19 mmol) in methanol (2 mL) was added sodium hydroxide aqueous solution (0.07 mL, 1M) until the pH to 9. Then, intermediate 64 (67 mg, 0.094 mmol) and sodium cyanoborohydride (11.8 mg, 0.189 mmol) were added into the mixture. After stirring at r.t. for 4 hours, the reaction mixture was concentrated and the residue was purified with Prep. HPLC (Column: Xbridge C18 150*19 mm*5 um, Mobile Phase A: water (0.1% TFA), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 2% B to 30% B). The collected fraction was lyophilized and the residue was basified with sodium hydroxide aqueous solution (1M) and extracted with dichloromethane (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated to afford the product which was lyophilized to give Compound 20 (22.1 mg, 97.3% purity, 34% yield) as a white solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 20














Compound No.
Structure
Starting Materials







Compound 21


embedded image


Intermediate 65 & 89





Compound 22


embedded image


Intermediate 66 & 89





Compound 23


embedded image


Intermediate 67 & 89





Compound 24


embedded image


Intermediate 66 & 1-(piperazin-1-yl)ethanone





Compound 25


embedded image


Intermediate 67 & 1-(piperazin-1-yl)ethanone





Compound 28


embedded image


Intermediate 68 & 89





Compound 29


embedded image


Intermediate 69 & 89





Compound 30


embedded image


Intermediate 70 & 89





Compound 31


embedded image


Intermediate 71 & 89









Preparation of Compound 26a & 26b:



embedded image


Triethylamine (113 mg, 1.12 mmol) was added to a solution of intermediate 89 (90 mg, 0.336 mmol) in dry dichloromethane (5 mL). Then intermediate 77 (120 mg, 0.223 mmol) was added. The reaction mixture was stirred at 25° C. for 30 minutes before the addition of sodium triacetoxyborohydride (95 mg, 0.448 mmol). After stirring at 25° C. for another 12 h, the reaction mixture was diluted with dichloromethane (50 mL) and saturated solution of sodium bicarbonate (50 mL). The mixture was extracted with dichloromethane (40 mL×3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by preparative-HPLC (Column: Welch Xtimate C18 150*25 mm*5 um, Mobile Phase A: water (0.225% FA), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 1% B to 28% B). The pure fractions were collected, and the solvent was evaporated under vacuum to give the mixture Compound 26a & 26b, which was further purified by preparative-HPLC (Column: Phenomenex Gemini-NX C18 75*30 mm*3 um, Mobile Phase A: water (0.04% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 35% B to 65% B). The pure fractions were collected, and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The first faction was lyophilized to dryness to give Compound 26a (25 mg, 96.7% purity, 16.0% yield) as a white powder and the second fraction was lyophilized to dryness to give Compound 26b (20.0 mg, 95.3% purity, 12.6% yield) as a white powder.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 26a & 26b














Compound No.
Structure
Starting Materials







Compound 27a


embedded image


Intermediate 78a & 89





Compound 27b


embedded image








Compound 27c


embedded image


Intermediate 78b & 89





Compound 27d


embedded image











Preparation of Compound 32a:



embedded image


To a solution of intermediate 62a (240 mg, 70% purity, 0.324 mmol) in methanol (5 mL) were added 1-(piperazin-1-yl)ethanone (83 mg, 0.648 mmol) and acetic acid (0.05 mL). The reaction mixture was stirred at room temperature for 30 minutes. Then sodium cyanoborohydride (40.7 mg, 0.648 mmol) was added into the mixture. After stirring at r.t. for 1 hr, the reaction mixture was basified with saturated NaHCO3 aqueous solution and extracted with dichloromethane (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered, and concentrated to afford the crude product, which was purified by prep. HPLC (Column: SunFire C18 150*19 mm*5 um, Mobile Phase A: water (0.1% TFA), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 2% B to 40% B). The collected fraction was lyophilized and the residue was basified with sodium hydroxide aqueous solution (1 M) and extracted with dichloromethane (20 mL) twice. The combined organic layer was washed with brine (20 mL), dried over Na2SO4, filtered and lyophilized to afford Compound 32a (90 mg, 97.8% purity, 43% yield) as a white solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 32














Compound No.
Structure
Starting Materials







Compound 32b


embedded image


Intermediate 62b & 1- (piperazin-1-yl)ethanone





Compound 33c


embedded image


Intermediate 62b & 4- (methylsulfonyl)piperidine









Preparation of Compound 33a & 33b:



embedded image


To a solution of intermediate 62a (82 mg, 80% purity, 0.126 mmol) in methanol (5 mL) was added 4-(methylsulfonyl)piperidine (41.3 mg, 0.25 mmol) and acetic acid (0.05 mL). The reaction mixture was heated to 25° C. and stirred at this temperature for 30 minutes. Then sodium triacetoxyborohydride (15.9 mg, 0.25 mmol) was added and the reaction mixture was stirred at this temperature overnight. The reaction mixture was concentrated, and the residue was purified by Prep. HPLC (Column: SunFire C18 150*19 mm*5 um, Mobile Phase A: water (0.1% TFA), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 10% B to 30% B). The collected fraction was lyophilized and the residue was basified with sodium hydroxide aqueous solution (1 M) and extracted with dichloromethane (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and lyophilized to afford Compound 33a (40 mg, 99.8% purity, 47.4% yield) and Compound 33b (9 mg, 99.8% purity, 10.7% yield) as a white solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 33a & 33b


Alternatively, (additional) purification can also be performed using the following method Prep. HPLC method (Boston Green ODS 150*30 mm*5 um, Mobile Phase A: water (0.225% formic acid), Mobile Phase B: acetonitrile, Flow rate: 35 mE/min, gradient condition from 5% B to 35%).














Compound No.
Structure
Starting Materials







Compound 34a


embedded image


Intermediate 62a & 1- (methylsulfonyl)piperazine





Compound 34b


embedded image








Compound 35a


embedded image


Intermediate 63 & 1-(piper- azin-1-yl)ethanone





Compound 35b


embedded image








Compound 36a


embedded image


Intermediate 63 & 4- (methylsulfonyl)piperidine





Compound 36b


embedded image








Compound 37a


embedded image


Intermediate 63 & 1- (methylsulfonyl)piperazine





Compound 37b


embedded image








Compound 38a


embedded image


Intermediate 63 & 89





Compound 38b


embedded image








Compound 39a


embedded image


Intermediate 76a & 89





Compound 39b


embedded image








Compound 40a


embedded image


Intermediate 76b & 89





Compound 40b


embedded image











Preparation of Compound 41:



embedded image


To a mixture of formaldehyde (194 mg, 6.46 mmol, 37% in H2O), Compound 376 (420 mg, 0.647 mmol) in MeOH (4 mL) was added NaOAc (265 mg, 3.23 mmol). The mixture was stirred at 25° C. for 1 h. Then NaBH3CN (81.6 mg, 1.30 mmol) was added to the mixture and the resulting mixture was stirred at 25° C. for 18 hours. The mixture was concentrated under reduced pressure to remove the solvent and the residue was diluted with ethyl acetate (10 mL), washed with saturated NaHCO3 (10 mL), H2O (10 mL) and brine (5 mL). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to give crude product, which was purified by preparative HPLC (Column: Boston Green ODS 150*30 mm*5 um, Mobile Phase A: water (0.225% FA), Mobile Phase B: acetonitrile, Flow rate: 35 mL/min, gradient condition from 10% B to 40% B). The pure fractions were collected, and the volatile solvent was evaporated under vacuum to give the residue, which was adjusted to pH=12 by NaOH (2 mol/L), then the mixture was extracted with ethyl acetate (20 mL). The organic phase was evaporated under vacuum to give the residue, which was lyophilized to afford the product (70 mg, purity 93.4%, yield 18%) as white solid.


Preparation of Compound 42:



embedded image


To a solution of intermediate 26 (80 mg, 0.192 mmol) in methanol (0.7 mL) was added tetrahydro-2H-pyran-4-carbaldehyde (69.2 mg, 0.576 mmol), NaBH3CN (36.2 mg, 0.576 mmol) and acetic acid (0.05 mL). After stirring at r.t. overnight, the reaction mixture was concentrated and the residue was purified by prep. HPLC (Column: Xbridge C18 150*19 mm*5 um, Mobile Phase A: water (0.10% NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 30% B to 70% B). The collected fraction was lyophilized to give Compound 42 (40 mg, 99.5% purity, 40.9% yield) as a white solid.


Preparation of Compound 43:



embedded image


To a mixture of intermediate 27 (90 mg, 0.22 mmol) and tetrahydro-2H-pyran-4-carbaldehyde (74 mg, 0.65 mmol) in methanol (2 mL) was added sodium cyanoborohydride (40 mg, 0.65 mmol). The reaction mixture was stirred at 20° C. overnight. The mixture was concentrated and purified by Prep. HPLC (Column: GiLSON-2 Xbridge C18 (5 μm 19*150 mm), Mobile phase A: water (0.1% ammonium bicarbonate), Mobile phase B: acetonitrile, UV: 214 nm, Flow rate: 15 mL/min, Gradient: 20% B to 60% B). The collected fraction was lyophilized to give Compound 43 (48 mg, 95% purity, 41% yield) as a white solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 43


In case reactions were performed with a ketone starting material, a typical procedure makes use of either 2 eq. acetic acid or 2 eq. of zinc(II)chloride (ZnCl2), in the presence of 2 eq. sodium cyanoborohydride (NaCNBH3), in methanol at 50° C. or 70° C. overnight.














Compound No.
Structure
Starting Materials







Compound 44


embedded image


Intermediate 26 & isobutyraldehyde





Compound 45


embedded image


Intermediate 27 & isobutyraldehyde





Compound 46


embedded image


Intermediate 26 & oxetane-3- carbaldehyde





Compound 47


embedded image


Intermediate 27 & oxetane-3- carbaldehyde





Compound 48


embedded image


Intermediate 26 & 1-(tetrahydro- 2H-pyran-4-yl)ethan-1-one





Compound 49


embedded image


Intermediate 27 & 1-(tetrahydro- 2H-pyran-4-yl)ethan-1-one





Compound 49a & Compound 49b


embedded image


Compound 49 was separated by supercritical fluid chromatography (separation condition: Phenomenex- Cellulose-2 (250 mm*30mm, 10um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O EtOH, A:B = 60:40 at 80 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was colleted as Compound 49a and the second fraction was Compound 49b








embedded image








Compound 81


embedded image


Intermediate 27 & 1- acetylpiperidin-4-one





Compound 109


embedded image


Intermediate 28 & tetrahydro- 2H-pyran-4-carbaldehyde





Compound 124


embedded image


Intermediate 27 & tetrahydro- 2H-thiopyran-4-carbaldehyde 1,1-dioxide





Compound 128


embedded image


Intermediate 130 & 1- acetylpiperidine-4-carbaldehyde





Compound 128a


embedded image


Compound 128 was performed via Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2). The first fraction was collected at Compound 128a, the second fraction as Compound 128b, the third fraction as Compound 128c and the fourth fraction as Compound 128d.





Compound 128b


embedded image








Compound 128c


embedded image








Compound 128d


embedded image








Compound 130


embedded image


Compound 503 & 1- acetylpiperidine-4-carbaldehyde





Compound 131


embedded image


Compound 504 & 1- acetylpiperidine-4-carbaldehyde





Compound 132


embedded image


Compound 505 & 1- acetylpiperidine-4-carbaldehyde





Compound 133


embedded image


Compound 522 & 1- acetylpiperidine-4-carbaldehyde





Compound 138


embedded image


Intermediate 202 & tetrahydro- 2H-pyran-3-carbaldehyde





Compound 138a & Compound 138b


embedded image


Compound 138 was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 138a & the second fraction was collected as Compound 138b.





Compound 139


embedded image


Intermediate 202 & azepane-2,5- dione





Compound 139a & Compound 139b


embedded image


Compound 139 was separated by Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, MeOH + 0.4 iPrNH2). The first fraction was collected as Compound 139a & the second fraction was Compound 139b.








embedded image








Compound 145


embedded image


Intermediate 213 & 1- acetylpiperidine-4-carbaldehyde





Compound 146


embedded image


Intermediate 215 & 1- acetylpiperidine-4-carbaldehyde





Compound 147a & Compound 147b


embedded image


Intermediate 202 & tetrahydro- 2H-pyran-2-carbaldehyde The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IG 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2). The first fraction was collected as Compound 147a and the second fraction as Compound 147b.








embedded image








Compound 156


embedded image


Intermediate 225 & 1- acetylpiperidin-4-one





Compound 160


embedded image


Intermediate 231 & 1- acetylpiperidine-4-carbaldehyde





Compound 161


embedded image


Intermediate 20 & 1- (methylsulfonyl)piperidin-4-one





Compound 165a & Compound 165b


embedded image


Intermediate 202 & 2-methoxy- 1-(tetrahydro-2H-pyran-4- yl)ethan-1-one The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IG 20 × 250 mm, Mobile phase: CO2, MeOH + 0.4 iPrNH2). Thie first fraction was collected as Compound 165a and the second fraction as Compound 165b.








embedded image








Compound 166a & Compound 166b


embedded image


Intermediate 240 & tetrahydropyran-4-carbaldehyde The product was separated via Prep SFC (Stationary phase: Chiralpak Daicel IC 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 166a and the second fraction as Compound 166b.








embedded image








Compound 167a & Compound 167b


embedded image


Intermediate 242 & tetrahydropyran-4-carbaldehyde The product was separated via Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2). The first fraction was collected as Compound 167a and the second fraction as Compound 167b.








embedded image








Compound 169


embedded image


Intermediate 202 & 7- oxoazepane-4-carbaldehyde





Compound 169a & Compound 169b


embedded image


Compound 169 was separated by Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 169a & the second fraction as Compound 169b.








embedded image








Compound 171


embedded image


Intermediate 202 & 1-methyl-2- oxopiperidine-4-carbaldehyde





Compound 180a & Compound 180b


embedded image


Intermediate 131a & 1- (tetrahydro-2H-pyran-4- yl)ethenone The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 180a and the second fraction as Compound 180b.








embedded image








Compound 181a & Compound 181b


embedded image


Intermediate 131b & 1- (tetrahydro-2H-pyran-4- yl)ethenone The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 181a and the second fraction as Compound 181b.








embedded image








Compound 182a & Compound 182b


embedded image


Intermediate 131c & 1- (tetrahydro-2H-pyran-4- yl)ethenone The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 182a and the second fraction as Compound 182b.








embedded image








Compound 183a & Compound 183b


embedded image


Intermediate 131d & 1- (tetrahydro-2H-pyran-4- yl)ethenone The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2) The first fraction was collected as Compound 183a and the second fraction as Compound 183b.








embedded image








Compound 188a & Compound 188b


embedded image


Intermediate 225 & 1-acetyl-3- methylpiperidin-4-one The product was separated by Prep. HPLC (Column: SunFire C18 150*19 mm*5 um, Mobile PhaseA: water (0.1% NH4OAc), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 10% B to 50%). The first fraction was collected as Compound 188a and the second fraction as Compound 188b.








embedded image








Compound 190a & Compound 190b


embedded image


Intermediate 202 & tetrahydrofuran-3-carbaldehyde The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 190a and the second fraction as Compound 190b.








embedded image








Compound 191a & Compound 191b


embedded image


Intermediate 25 & tetrahydrofuran-3-carbaldehyde The product was separated by P Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2). The first fraction was collected as Compound 191a and the second fraction as Compound 191b.








embedded image








Compound 193


embedded image


Intermediate 20 & cyclohexanecarbaldehyde





Compound 194


embedded image


Intermediate 225 & cyclohexanecarbaldehyde





Compound 196


embedded image


Intermediate 25 & 2- oxopiperidine-4-carbaldehyde





Compound 198a & Compound 198b


embedded image


Intermediate 202 & 259 The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IG 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 198a and the second as Compound 198b.








embedded image








Compound 206a & Compound 206b


embedded image


Intermediate 202 & 266 The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 206a and the second as Compound 206b.








embedded image








Compound 213 & Compound 213b


embedded image


Intermediate 25 & 1-(1,1- dioxidotetrahydro-2H-thiopyran- 4-yl)ethan-1-one The product was separated by SFC (separation condition: DAICEL CHIRALPAK IG (250 mm*30 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O MeOH, A:B = 40:60 at 80 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar, Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 213a and the second fraction as Compound 213b.








embedded image








Compound 214


embedded image


Intermediate 278 & 1- acetylpiperidine-4-carbaldehyde Compound 214 was further separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 214a and the second fraction as Compound 214b.





Compound 214a & Compound 214b


embedded image











embedded image








Compound 224


embedded image


Intermediate 298 & 1- acetylpiperidine-4-carbaldehyde Compound 224 was further separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile Phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 224a and the second fraction as Compound 224b.





Compound 224a & Compound 224b


embedded image











embedded image








Compound 225


embedded image


Intermediate 202 & 300





Compound 226


embedded image


Compound 507 & 1- acetylpiperidine-4-carbaldehyde





Compound 227


embedded image


Compound 508 & 1- acetylpiperidine-4-carbaldehyde





Compound 228


embedded image


Compound 509 & 1- acetylpiperidine-4-carbaldehyde





Compound 229


embedded image


Compound 510 & 1- acetylpiperidine-4-carbaldehyde





Compound 230a & Compound 230b


embedded image


Intermediate 311a & 1-(4- acetylpiperidino)ethan-1-one The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 230a & the second fraction as Compound 230b.








embedded image








Compound 231a & Compound 231b


embedded image


Intermediate 311b & 1-(4- acetylpiperidino)ethan-1-one The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 231a & the second fraction as Compound 231b.








embedded image








Compound 232a & Compound 232b


embedded image


Intermediate 312a & 1-(4- acetylpiperidino)ethan-1-one The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 232a & the second fraction as Compound 232b.








embedded image








Compound 233a & Compound 233b


embedded image


Intermediate 312b & 1-(4- acetylpiperidino)ethan-1-one The product was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 233a & the second fraction as Compound 233b.








embedded image








Compoudn 239


embedded image


Compound 511 & tetrahydro- 2H-pyran-4-carbaldehyde





Compound 240a & Compound 240b


embedded image


Intermediate 20 & 324 The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IG 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 240a & the second fraction as Compound 240b.








embedded image








Compound 252a & Compound 252b


embedded image


Intermediate 20 & 334 The product was separated by Chiral Prep. HPLC (separation condition: Column: Chiralpak IA 5 um 30 * 250 mm, Mobile Phase: Hexane: iso-Propyl alcohol = 90:10 at 325 mL/min; Temp: 30° C.; Wavelength: 254 nm). The first fraction was collected as Compound 252a & the second fraction as Compound 252b.








embedded image








Compound 253a & Compound 253b


embedded image


Intermediate 202 & 335 The product was separated by chiral Prep. HPLC (separation condition: Column: Chiralpak IA 5 um 30 * 250 mm; Mobile Phase: Hexane: EtOH = 80:20 at 25 mL/min; Temp: 30° C.; Wavelength: 254 nm). The first fraction was collected as Compound 253a & the second fraction as Compound 253b.








embedded image








Compound 257


embedded image


Intermediate 338 & tetrahydro- 2H-pyran-4-carbaldehyde





Compound 523a (E or Z, not determined) & Compound 523b (Z or E, not determined)


embedded image


Intermediate 399 & tetrahydro- 2H-pyran-4-carbaldehyde








embedded image








Compound 524a (E or Z, not determined) & Compound 524b (Z or E, not determined)


embedded image


Intermediate 399 & 37% aq. Formaldehyde solution








embedded image











Preparation of Compound 50:



embedded image


To a solution of Compound 381 (70 mg, 0.102 mmol) and DIEA (79 mg, 0.61 mmol) in DCM (4 mL) was added acetic anhydride (52 mg, 0.51 mmol). After stirring at r.t. for 4 hours, the reaction mixture was concentrated, and the residue was purified by Prep-HPLC: Waters Xbridge C18 5 μm 19*150 mm. Mobile phase A:0.1% NH4OH+10 mM NH4HCO3 in water. B: CH3CN, gradient from 0% B to 100% B. The pure fraction was collected and lyophilized to afford Compound 50 (50 mg, 88% yield) as a white solid.


Preparation of Compound 51:



embedded image


At 0° C., to a solution of Compound 485 (1.04 g, 95% purity, 1.95 mmol) in DCM (10 mL) was added acetyl chloride (160 mg, 2.05 mmol) and triethylamine (592 mg, 5.85 mmol). After stirring at room temperature for 2 hours, the resulting mixture was poured into water and extracted with dichloromethane (20 mL) twice. The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated to afford the crude product, which was purified by prep HPLC (Column: Xbridge C18 150*19 mm*5 um, Mobile Phase A: water (0.1% NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 15% B to 60% B). The collected fraction was lyophilized to give Compound 51 (1.25 g, 99.8% purity, 74.9% yield) as a white solid.


Alternatively, compound 51 can also be prepared with the following procedure:


Intermediate 202 (as 0.2TFA salt) (0.20 g, 0.49 mmol) and 1-acetylpiperidine-4-carbaldehyde (0.097 g, 0.62 mmol) were dissolved in MeOH (5.5 mL). After stirring at ambient temperature for −5 min, solid NaCNBH3 (0.039 g, 0.62 mmol) was added. The resulting mixture was stirred at ambient temperature for ˜2 h, after which sat. aq. NaHCO3 solution was added. Then, most of the MeOH was evaporated to dryness, and DCM was added. The pH of the water layer was adjusted to pH>10 with 1M aq. NaOH solution. The layers were separated and the water layer was extracted three times more with DCM. The organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by silica gel column chromatography eluting with methanol (+1% 7N NH3 in MeOH) in dichloromethane from 0% to 10% to give compound 51 (0.060 g, 0.11 mmol, 35% yield).


Compound 51 (originating from route via intermediate 202; 0.051 g, purity 99.7%, LC/MS method 32) was dissolved in 2-3 drops of isopropylacetate (IPAC), after which the resulting solution was stirred at 45° C. for ˜5 h. Next, the mixture was allowed to stir at ambient temperature for 48 h, after which it was filtered to obtain a white solid material corresponding with Compound 51 in its crystalline free base Form. Melting point (via DSC): Tonset=121.6° C.


Compound 51 ((originating from route via intermediate 202; ˜1 g, 98.7% purity, LC/MS method 33) was dissolved in cyclopentylmethylether (CPME) (3 mL), after which heptane (2 mL) was slowly added, followed by the addition of −10 mg of seeding crystals (obtained via previous procedure). Next, 1 mL of heptane was added and the mixture stirred for 20 h, after which the suspension was filtered to give solid material which was dried at 40° C. under vacuum to yield Compound 51 in its crystalline free base Form (96% yield).


Chiral SFC method 1 was employed to match the stereochemistry of compound 51 obtained through the route employing Compound 485 or intermediate 202; retention time=4.73-4.77 min.


Preparation of Compound 51a:



embedded image


Compound 51 (0.50 g, 0.91 mmol, purity 95.2% (determined by LC/MS method 32)) was dissolved in acetone (0.50 mL) and stirred to give a clear solution. Next, a solution of 1M HCl in acetone was prepared as follows: 1 mL of concentrated aq. HCl solution was added to 11 mL of acetone. Then, a solution of 1M HCl in acetone (0.92 mL, 1 eq.) was added, keeping a solution. The solution was stirred at ambient temperature for ˜30-60 min, after which heptane (5.0 mL) was added. Next, acetone was added (3.0 mL). Vigorous stirring was initiated, and the mixture was stirred overnight. Then, a fine white suspension was obtained, and the suspension was filtered. The solid was rinsed with heptane and dried to give Compound 51a as a mono HCl trihydrate salt (when determined via dynamic vapor sorption analysis around 3 equivalents water) as a white solid (0.48 g, yield 78%). Melting point (via DSC): Tonset=139° C.


Compound 51a was obtained as a variable hydrate with equilibrated water content varying as function of humidity —mainly trihydrate at ambient % relative humidity.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 51


Alternatively, compounds can also be purified by the following method: prep. HPLC: (Column: Waters Sunfire C18 5 μm, 19*150 mm, Mobile Phase A: water (0.1% HCOOH), Mobile Phase B: acetonitrile, Flow rate: 17 mL/min, gradient condition from 0% B to 20% B).














Compound No.
Structure
Starting Materials







Compound 58


embedded image


Compound 433 & acetyl chloride





Compound 90a & Compound 90b


embedded image


Compound 431 & acetyl chloride A purification was performed via Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2) The first fraction was collected as Compound 90a and the second as Compound 90b








embedded image








Compound 102


embedded image


Compound 436 & acetyl chloride





Compound 121


embedded image


Compound 485 & isobutyryl chloride





Compound 122


embedded image


Compound 445 & acetyl chloride





Compound 135


embedded image


Compound 485 & cyclopropanecarbonyl chloride





Compound 136


embedded image


Compound 381 & 2- methoxyacetyl chloride





Compound 149


embedded image


Compound 448 & dimethylcarbamic chloride





Compound 170


embedded image


Compound 451 & dimethylcarbamic chloride





Compound 173


embedded image


Compound 448 & methoxy(methyl)carbamic chloride





Compound 174


embedded image


Compound 448 & morpholine-4- carbonyl chloride





Compound 187


embedded image


Compound 455 & acetyl chloride





Compound 200


embedded image


Compound 406b & acetyl chloride





Compound 201


embedded image


Compound 406a & acetyl chloride





Compound 203


embedded image


Compound 433 & methoxy(methyl)carbamic chloride





Compound 204


embedded image


Compound 433 & morpholine-4- carbonyl chloride





Compound 208


embedded image


Compound 462 & acetyl chloride





Compound 209


embedded image


Compound 463 & acetyl chloride





Compound 210


embedded image


Compound 465 & acetyl chloride





Compound 211


embedded image


Compound 464 & acetyl chloride





Compound 216


embedded image


Compound 467 & acetyl chloride





Compound 220


embedded image


Compound 470 & acetyl chloride





Compound 222


embedded image


Compound 471 & acetyl chloride





Compound 223


embedded image


Compound 472 & acetyl chloride





Compound 234


embedded image


Compound 473 & acetyl chloride





Compound 235


embedded image


Compound 433 & dimethylcarbamic chloride





Compound 238


embedded image


Compound 474 & acetyl chloride





Compound 255


embedded image


Compound 480 & acetyl chloride





Compound 256


embedded image


Compound 481 & acetyl chloride





Compound 258


embedded image


Compound 482 & acetyl chloride





Compound 259


embedded image


Compound 483 & acetyl chloride





Compound 260


embedded image


Compound 484 & acetyl chloride









Preparation of Compound 59:



embedded image


To a mixture of Compound 485 (70 mg, 0.138 mmol), methoxyacetic acid (18.7 mg, 0.208 mmol) and DIPEA (0.07 mL, 0.42 mmol) in DCM (4.2 mL) was added HATU (78.9 mg, 0.208 mmol). After stirring at rt for 16 hours, the reaction mixture was concentrated and the residue was purified by Prep. HPLC (Column: Waters Xbridge C18 5 μm, 19*150 mm, Mobile Phase A: water (0.1% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 17 mL/min, gradient condition from 30% B to 50% B). The pure fraction was collected and lyophilized to dryness to afford Compound 59 (65 mg, 79.6% yield).


Preparation of Compound 60:



embedded image


To a mixture of Compound 485, (70 mg, 0.138 mmol), cyanoacetic acid (17.7 mg, 0.208 mmol) and DIPEA (0.07 mL, 0.415 mmol) in DCM (5 mL) was added HATU (78.9 mg, 0.208 mmol). After stirring at RT for 16 hours, the reaction mixture was concentrated, and the residue was purified by Prep. HPLC (Column: Waters Xbridge C18 5 μm, 19*150 mm, Mobile Phase A: water (0.1% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 17 mL/min, gradient condition from 30% B to 50% B). The pure fraction was collected and lyophilized to dryness to afford Compound 60 (65 mg, 81% yield).


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 60


Alternatively, purification can also be performed using the following method: Prep. HPLC (Column: Xbrige C18 150*19 mm*5 um, mobile phase A: water (0.1% HCOOH), mobile phase B: acetonitrile, flow rate: 15 mL/min, gradient condition from 5% B to 60% B)














Compound No.
Structure
Starting Materials







Compound 82


embedded image


Compound 485 & 3- hydroxypropanoic acid





Compound 85


embedded image


Compound 485 & (R)-2- hydroxypropanoic acid





Compound 86


embedded image


Compound 485 & (S)-2- hydroxypropanoic acid





Compound 87


embedded image


Compound 485 & 2- hydroxyacetic acid





Compound 106


embedded image


Compound 485 & 2-(1,1- dioxidothietan-3-yl)acetic acid





Compound 108


embedded image


Compound 381 & 2-(1,1- dioxidothietan-3-yl)acetic acid





Compound 111


embedded image


Compound 436 & 2- methoxyacetic acid





Compound 117a


embedded image


Compound 442 & 2- methoxyacetic acid





Compound 117b


embedded image


Compound 443 & 2- methoxyacetic acid





Compound 123


embedded image


Compound 445 & 2- hydroxyacetic acid





Compound 125


embedded image


Compound 485 & 2-hydroxy-2- methylpropanoic acid





Compound 126


embedded image


Compound 485 & 2-cyano-2- methylpropanoic acid





Compound 177


embedded image


Compound 454 & acetic acid





Compound 237


embedded image


Compound 433 & 2-hydroxy-2- methylpropanoic acid





Compound 244


embedded image


Compound 451 & 2-hydroxy-2- methylpropanoic acid





Compound 245


embedded image


Compound 451 & 1- hydroxycyclopropane-1- carboxylic acid





Compound 248


embedded image


Compound 479 & acetic acid





Compound 249


embedded image


Compound 448 & 1- hydroxycyclopropane-1- carboxylic acid





Compound 250


embedded image


Compound 429 & 2-hydroxy-2- methylpropanoic acid





Compound 251


embedded image


Compound 429 & 1- hydroxycyclopropane-1- carboxylic acid









Preparation of Compound 61:



embedded image


To a solution of intermediate 25 (0.082 g, 0.21 mmol) in 1,2-DCE (2.0 mL) was add tetrahydropyran-4-carbaldehyde (0.028 g, 0.25 mmol), and followed by NaBH(OAc)3 (0.062 g, 0.29 mmol). After stirring at ambient temperature overnight, another portion of tetrahydropyran-4-carbaldehyde (0.028 g, 0.25 mmol) and NaBH(OAc)3 (0.062 g, 0.29 mmol) was added. After stirring for another 1.5 h, 1M aq. NaOH solution was added, followed by DCM. The layers were separated, and the aqueous layer was extracted 4× with DCM. The organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by RP-preparative HPLC (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give Compound 61 (0.058 g, 57% yield), after lyophilization, as a white fluffy powder.


Preparation of Compound 62:



embedded image


To a solution of intermediate 25 (0.18 g, 0.45 mmol) in 1,2-DCE (4.2 mL) was added N-Boc-piperidine-4-carboxaldehyde (0.11 g, 0.54 mmol), and followed by NaBH(OAc)3 (0.13 g, 0.63 mmol). After stirring at ambient temperature overnight, DCM and 1M aq. NaOH were added. The layers were separated, and the aqueous layer was extracted 4×more with DCM. The organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by silica gel column chromatography eluting with methanol (+1% 7N NH3 in MeOH) in dichloromethane from 0% to 10% to give Compound 62 (0.25 g, 94% yield) as a foam.


Preparation of Compound 63:



embedded image


To a solution of Compound 62 (0.25 g, 0.42 mmol) in DCM (5 mL) was added TFA (5 mL). After stirring at ambient temperature for 2 h, the reaction mixture was evaporated to dryness and the residue applied to SiliaBond® propylsulfonic acid resin as a solution in MeOH. The column was eluted with MeOH (8 fractions), followed by 3.5 N NH3 in MeOH (8 fractions). Product containing fractions were pooled and evaporated to give an intermediate, which was dissolved in DCM (3.6 mL). The solution was cooled to 0° C. in an ice bath and DIPEA (0.13 mL, 0.76 mmol) was added, followed by Ac2O (0.06 mL, 0.63 mmol). The resulting mixture was stirred at ambient temperature for 2 h, after which LC/MS showed full conversion of the starting material. Then, sat. aq. NaHCO3 solution was added. The resulting mixture was partitioned between 1M aq. NaOH solution and DCM. The water layer was extracted 5× with DCM and the organic layers were combined, dried over Na2SO4, filtered, and evaporated to dryness. The residue was purified by RP-preparative HPLC (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give Compound 63 (0.046 g, 68% yield) after lyophilization as a white fluffy powder.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 63














Compound No.
Structure
Starting Materials







Compound 64


embedded image


Compound 62 & methanesulfonyl chloride





Compound 65


embedded image


Compound 62 & methyl carbonochloridate









Preparation of Compound 66:



embedded image


To a solution of Compound 62 (450 mg, 0.760 mmol) in anhydrous dichloromethane (4 mL) was added trifluoroacetic acid (4 mL). After stirring at 25° C. for 30 minutes, the reaction mixture was concentrated under reduced pressure to give a residue, which was diluted with dichloromethane (80 mL) and then basified to pH=14 with 10% aqueous NaOH (50 mL). The mixture was extracted with dichloromethane (60 mL×3) and the combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product (260 mg, crude) as a white solid, which was used for next step without further purification. To a solution of crude product (100 mg, 0.203 mmol) and 2-methoxyacetic acid (18.3 mg, 0.203 mmol) in anhydrous dichloromethane (3 mL) was added N,N-diisopropylethylamine (31.5 mg, 0.244 mmol). HATU (77.3 mg, 0.203 mmol) was added to the mixture under stirring, then the reaction mixture was stirred at 25° C. for 1 h. The reaction mixture was diluted with dichloromethane (20 mL), water (30 mL) was added. The mixture was extracted with dichloromethane (20 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by preparative-HPLC (Column: Phenomenex Gemini-NX C18 75*30 mm*3 um, Mobile Phase A: water (0.05% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 28% B to 58% B). The pure fractions were collected and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give Compound 66 (50.0 mg, 99.4% purity, 43.4% yield) as a white powder.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 66














Compound No.
Structure
Starting Materials







Compound 67


embedded image


Compound 62 & cyanoacetic acid









Preparation of Compound 68:



embedded image


At 0° C., to a solution of Compound 488 (380 mg, 0.6 mmol) and triethylamine (500 mg, 4.94 mmol) in anhydrous dichloromethane (10 mL) was added methanesulfonyl chloride (500 mg, 4.37 mmol) dropwise. The reaction mixture was warmed up to r.t. and stirred for 2 hours. The reaction mixture was quenched with a saturated solution of sodium bicarbonate (20 mL) and H2O (20 mL) and extracted with dichloromethane (30 mL×3). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give the crude product, which was purified by preparative-HPLC (Column: Phenomenex Gemini-NX C18 75*30 mm*3 um, Mobile Phase A: water (0.04% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 33% B to 63% B). The pure fractions were collected and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give the desired compound (150 mg, 97.9% purity, 41% yield) as a white powder.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound_68














Compound No.
Structure
Starting Materials







Compound 69


embedded image


Intermediate 27 & methanesulfonyl chloride





Compound 115


embedded image


Compound 441 & methanesulfonyl chloride









Preparation of Compound 70:



embedded image


Intermediate 26 (75 mg, 0.184 mmol), DMF (4 mL), intermediate 116 (75 mg, 0.230 mmol), cesium carbonate (180 mg, 0.552 mmol) and potassium iodide (7 mg, 0.042 mmol) were added to a 50 mL round-bottomed flask. After degassing with N2, the reaction mixture was heated and stirred at 100° C. overnight. The reaction mixture was poured into water (10 mL) and extracted with DCM (10 mL×3). The combined organic extracts were dried over anhydrous Na2SO4, filtered, and concentrated to dryness under reduced pressure to give the crude product which was purified by prep. HPLC (Phenomenex Gemini-NX C18 75*30 mm*3 um, Mobile Phase A: water (0.05% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 32% B to 62% B). The pure fractions were collected, and the solvent was evaporated under vacuum to give a residue. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The solution was lyophilized to dryness to give Compound 70 (34.27 mg, 98.3% purity, 33% yield) as a yellow solid.


Preparation of Compound 71:



embedded image


A mixture of intermediate 27 (75 mg, 0.184 mmol), intermediate 116 (75 mg, 0.230 mmol), cesium carbonate (180 mg, 0.552 mmol) and potassium iodide (7 mg, 0.042 mmol) in DMF (4 mL) was degassed with N2 and the reaction mixture was heated and stirred at 100° C. overnight.


After cooled down to room temperature, the reaction mixture was poured into water (10 mL) and extracted with DCM (10 mL×3). The combined organic extracts were dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure to give the crude product which was purified by prep. HPLC (Welch Xtimate C18 150*30 mm*5 um, Mobile Phase A: water (0.05% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 35 mL/min, gradient condition from 25% B to 55% B). The pure fractions were collected, and the solvent was evaporated under vacuum to give a residue. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The solution was lyophilized to dryness to give Compound 71 (33 mg, 96.1% purity, 30.7% yield) as a yellow solid.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 71














Compound No.
Structure
Starting Materials







Compound 140


embedded image


Compound 506 & intermediate 116









Preparation of Compound 72:



embedded image


Triethylamine (80.0 mg, 0.791 mmol) was added to a solution of Compound 488 (80 mg, 0.126 mmol) in dichloromethane (3.0 mL). Then acetic anhydride (20.0 mg, 0.196 mmol) was added. After stirring at 25° C. for 30 minutes, the reaction mixture was suspended into aq. NaHCO3 solution (30 mL) and extracted with dichloromethane (20 mL×2). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by prep. HPLC (Column: Phenomenex Gemini-NX C18 75*30 mm*3 um, Mobile Phase A: water (0.05% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 46% B to 76% B). The pure fraction was collected and the solvent was evaporated under vacuum. The residue was re-suspended in water (10 mL) and the resulting mixtures were lyophilized to dryness to give Compound 72 (20.0 mg, 100% purity, 28.2% yield) as a white powder.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 72














Compound No.
Structure
Starting Materials







Compound 73


embedded image


Intermediate 27









Preparation of Compound 74:



embedded image


Intermediate 25 (185 mg, 0.469 mmol), DMF (5 mL), intermediate 116 (185 mg, 0.568 mmol), cesium carbonate (460 mg, 1.41 mmol) and potassium iodide (16 mg, 0.096 mmol) were combined into a 50 mL round-bottomed flask. After degassing with N2, the reaction mixture was heated and stirred at 100° C. for 6 hours. After cooled down to the room temperature, the reaction mixture was poured into water (30 mL) and extracted with EtOAc (30 mL×3). The combined organic extracts were dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure to give the crude product which was purified by prep. HPLC (Phenomenex Gemini-NX C18 75*30 mm*3 um, Mobile Phase A: water (0.05% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 33% B to 63% B). The pure fractions were collected, and the solvent was evaporated under vacuum to give a residue. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The solution was lyophilized to dryness to give Compound 74 (40.78 mg, 95.7% purity, 15% yield) as a brown powder.


Preparation of Compound 83:



embedded image


Intermediate 25 (0.060 g, 0.152 mmol) was dissolved in MeCN (1.6 mL). Then, 4-(2-chloroacetyl)morpholine (0.027 g, 0.17 g) and triethylamine (0.13 mL, 0.91 mmol) was added and the resulting mixture stirred at ambient temperature for 2 h. Next, MeOH was added and the mixture was evaporated to dryness. The residue was purified by RP-preparative HPLC (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give Compound 83 (32 mg, 0.059 mmol, 39% yield).


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 83














Compound No.
Structure
Starting Materials







Compound 96


embedded image


Intermediate 25 & 147





Compound 97


embedded image


Intermediate 25 & 148





Compound 98


embedded image


Intermediate 25 & 2-chloro-1- piperidin-1-yl-ethanone









Preparation of Compound 75:



embedded image


To a solution of intermediate 118 (0.060 g, 0.24 mmol) and intermediate 25 (0.11 g, 0.29 mmol) in MeOH (1 mL) was added AcOH (28 μL, 0.48 mmol), followed by NaBH3CN (0.030 g, 0.48 mmol). The mixture was stirred at ambient temperature overnight. Next, sat. aq. NaHCO3 solution was added. After stirring for −5 min, the mixture was evaporated to dryness. The residue was purified by preparative HPLC (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give Compound 75 (0.042 g, 49% yield).


Preparation of Compound 76:



embedded image


To a solution of intermediate 118 (0.058 g, 0.23 mmol) and intermediate 121 (0.14 g, 0.28 mmol) in MeOH (1 mL) was added AcOH (27 μL, 0.47 mmol), followed by NaBH3CN (0.029 g, 0.47 mmol). The mixture was stirred at ambient temperature overnight. Next, sat. aq. NaHCO3 solution was added. After stirring for ˜5 min, the mixture was evaporated to dryness. The residue was purified by preparative HPLC (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give the product (0.090 g, 0.13 mmol) with 92% purity determined via 1H NMR integration. (˜5% on UV via SFC). Additional purification via prep. SFC (Stationary phase: Chiralcel Diacel IH 20×250 mm, Mobile phase: CO2, EtOH+0.4 iPrNH2) yielded pure Compound 76 (0.061 g, 0.098 mmol).


Preparation of Compound 77, 77a, 77b, 77c, 77d:



embedded image


embedded image


A mixture of intermediate 130 (77.0 mg, 0.183 mmol) and tetrahydropyran-4-carbaldehyde (27.5 mg, 0.241 mmol) in MeOH (1.20 mL) was stirred for 30 min after which sodium cyanoborohydride (15.1 mg, 0.241 mmol) was added. The reaction mixture was stirred at rt overnight. The reaction was quenched with water and used as such for reversed-phase prep HPLC purification (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN), followed by purification by silica gel column chromatography eluting with ethyl acetate, followed by 20% methanol in dichloromethane to give Compound 77 (32 mg, 50.5% yield) as a white solid.


Compound 77 was further purified by Prep SFC (Stationary phase: Chiralpak Diacel AD 20×250 mm, Mobile phase: CO2, iPrOH+0.4 iPrNH2). The first fraction was collected at Compound 77a, the second fraction as Compound 77b, the third fraction as Compound 77c and the fourth fraction as Compound 77d.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 77














Compound No.
Structure
Starting Materials

















Compound 78


embedded image


Intermediate 131





Compound 78a


embedded image


Compound 78 was purified by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 78a, the second fraction as Compound 78b, the third fraction as Compound 78c and the fourth fraction as Compound 78d





Compound 78b


embedded image








Compound 78c &


embedded image








Compound 78d


embedded image








Compound 79


embedded image


Intermediate 1, tert-butyl 3- formylazetidine-1-carboxylate & intermediate 124





Compound 80


embedded image


Intermediate 1, tert-butyl 3- formylazetidine-1-carboxylate & intermediate 125









Preparation of Compound 84:



embedded image


At 0° C., to a solution of Compound 430 (256 mg, 0.506 mmol) in dichloromethane (10 mL) were added acetyl chloride (40 mg, 0.510 mmol) and triethylamine (155 mg, 1.532 mmol). After stirring at r.t. for 1 hr, the mixture was quenched with saturated aqueous sodium hydrogen carbonate solution and extracted with dichloromethane three times. The combined organic layers were washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by prep. HPLC (Column: Waters Xbridge C18 OBD 5 μm, 19*150 mm, Mobile Phase A: water (0.1% NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 20% B to 60% B) to give Compound 84 (88 mg, 30.8% yield) as a white solid.


Preparation of Compound 88:



embedded image


Intermediate 27 (50 mg, 0.12 mmol), intermediate 134 (69.3 mg, 0.24 mmol), DIEA (0.105 mL, 0.61 mmol) and potassium iodide (20.3 mg, 0.12 mmol) were added to NMP (2 mL). The mixture was stirred at 70° C. for 16 hours. The mixture was separated by HPLC (Column: Waters Xbridge C18 5 μm, 19*150 mm, Mobile Phase A: water (0.1% NH3H2O+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 17 mL/min, gradient condition from 20% B to 50% B). The pure fraction was collected and lyophilized to afford Compound 88 (20 mg, yield 29.80%).


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 88














Compound No.
Structure
Starting Materials







Compound 89


embedded image


Intermediate 27 & 135





Compound 95


embedded image


Intermediate 25 & 135





Compound 105


embedded image


Intermediate 28 & 159





Compound 110


embedded image


Intermediate 28 & 162





Compound 164a & Compound 164b


embedded image


Intermediate 236 & 116 The product was separated by SFC (separation condition: DAICEL CHIRALPAK IG (250 mm* 30 mm, 10 um); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O MeOH, A:B = 55:45 at 80 mL/min; Column Temp: 38° C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 164a and the second fraction as Compound 164b.








embedded image








Compound 186a & Compound 186b


embedded image


Intermediate 28 & 249 The product was separated by SFC (Column ID: IH Column Size: 4.6 mm* 250 mm 5 um, Method: CAN-IPA-DEA-50- 50-0.3-30 MIN, Flow: 1 ml/min, Temperature: 30° C.). The first fraction was collected as Compound 186a and the second as Compound 186b.








embedded image








Compound 195


embedded image


Intermediate 28 & 134





Compound 212


embedded image


Intermediate 202 & 273





Compound 254


embedded image


Intermediate 336 & 338









Preparation of Compound 91:



embedded image


A mixture of intermediate 25 (127 mg, 0.322 mmol), 2-(Boc-amino)-6-oxospiro[3.3]heptane (145 mg, 0.644 mmol) and AcOH (36.9 μL, 0.644 mmol) in MeOH (3.2 mL) was stirred for 30 min after which sodium cyanoborohydride (40.5 mg, 0.644 mmol) was added. The reaction mixture was stirred at 50° C. overnight. The reaction was cooled down to r.t., quenched with water, and evaporated to dryness. The residue was purified by silica gel column chromatography eluting with methanol (+1% NH3 in MeOH) in dichloromethane from 1% to 50%. The purest fractions were collected, evaporated to dryness to afford Compound 91 (64 mg, yield 32.6%) as a white solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 91














Compound No.
Structure
Starting Materials







Compound 92


embedded image


Intermediate 25 & 1,4- dioxaspiro[4.5]decane-8- carbaldehyde





Compound 104


embedded image


Intermediate 25 & 7- oxoazepane-4-carbaldehyde





Compound 114


embedded image


Intermediate 25 & 1- acetylazetidine-3-carbaldehyde





Compound 120


embedded image


Intermediate 25 & 6- oxopiperidine-3-carbaldehyde









Preparation of Compound 93:



embedded image


Compound 490 (55 mg, 0.107 mmol) was dissolved in DCM (1.2 mL). Then, DIPEA (0.11 mL, 0.64 mmol) was added, followed by Ac2O (0.051 mL, 0.536 mmol). The resulting mixture was then stirred at ambient temperature for 2 h. Next, a small amount of MeOH was added, and the mixture evaporated to dryness. The compound was purified by silica gel column chromatography eluting with methanol (+1% 7N NH3 in MeOH) in dichloromethane from 1% to 20% to afford the product (80 mg), which was triturated with DEE to give Compound 93 (52.3 mg, yield 83.5%).


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 93














Compound No.
Structure
Starting Materials







Compound 94


embedded image


Compound 432





Compound 99


embedded image


Compound 434





Compound 100


embedded image


Compound 435





Compound 103


embedded image


Compound 437





Compound 107


embedded image


Compound 438





Compound 112


embedded image


Compound 439





Compound 113


embedded image


Compound 502





Compound 116a


embedded image


Compound 442





Compound 116b


embedded image


Compound 443





Compound 118


embedded image


Compound 441





Compound 119a & Compound 119b


embedded image


Compound 444 The mixture was separated by SFC (separation condition: DAICEL CHIRALPAK AS (250 mm*30 mm, 10 um)); Mobile phase: A: Supercritical CO2, B: 0.1% NH3H2O EtOH, A:B = 85:15 at 60 mL/min; Column Temp: 38 °C.; Nozzle Pressure: 100 Bar; Nozzle Temp: 60° C.; Evaporator Temp: 20° C.; Trimmer Temp: 25° C.; Wavelength: 220 nm). The first fraction was collected as Compound 119a and the second fraction as Compound 119b








embedded image








Compound 127


embedded image


Compound 446





Compound 137a & Compound 137b


embedded image


Compound 391 The mixture was purified by the Prep. HPLC (Column: SunFire C18 150*19 mm*5 um, Mobile Phase A: water (0.1% NH4Oac), Mobile Phase B: acetonitrile, Flow rate: 15 mL/min, gradient condition from 10% B to 50% B). The first fraction was collected as Compound 137a & the second fraction as Compound 137b. The absolute stereochemistry was not determined.








embedded image








Compound 140


embedded image


Compound 447





Compound 150


embedded image


Compound 449





Compound 151


embedded image


Compound 450





Compound 175


embedded image


Compound 452





Compound 176


embedded image


Compound 453





Compound 189


embedded image


Compound 456





Compound 192


embedded image


Compound 457





Compound 215


embedded image


Compound 466





Compound 242


embedded image


Compound 475





Compound 243


embedded image


Compound 476





Compound 246


embedded image


Compound 477





Compound 247


embedded image


Compound 478









Preparation of Compound 101:



embedded image


To a solution of Compound 485 (100 mg, 0.20 mmol), triethylamine (61 mg, 0.59 mmol) in dichloromethane (10 mL) was added a solution of methylaminoformyl chloride (23 mg, 0.22 mmol) in 2 mL of DCM. After stirring at 20° C. for 5 hr, the mixture was diluted with water (20 mL) and extracted with DCM (10 mL) for three times. The combined organic layers were washed with brine (30 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum, which was purified by Prep-HPLC (Prep HPLC (Column: Xbridge C18 (5 m 19*150 mm), Mobile Phase A: Water (0.1% NH4HCO3), Mobile Phase B: acetonitrile, UV: 214 nm, Flow rate: 15 mL/min, Gradient: 15% B to 55% B) to give Compound 101 (90 mg, 0.15 mmol, 76.8% yield) as a white solid.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 101














Compound No.
Structure
Starting Materials







Compound 168


embedded image


Compound 451 & methylcarbamic chloride





Compound 197


embedded image


Compound 458 & methylcarbamic chloride





Compound 202


embedded image


Compound 433 & methylcarbamic chloride









Preparation of Compound 129:



embedded image


A mixture of intermediate 179 (85 mg, 0.16 mmol), 2 M methanamine in tetrahydrofuran (0.16 mL, 0.32 mmol), HATU (90 mg, 0.24 mmol, 1.5 equivalent), triethylamine (48 mg, 0.48 mmol, 3.0 equivalent) and DMF (10 mL) was stirred at room temperature overnight. The mixture was diluted with water (20 mL) and extracted with ethyl acetate (25 mL) for three times. The combined organic layers were dried over Na2SO4, filtered and concentrated to give the crude product, which was purified by prep. TLC eluting with 10% methanol in dichloromethane to give Compound 129 (55.3 mg, yield: 61.7%).


Preparation of Compound 134, 134a, 134b, 134c, 134d:



embedded image


TFA (1.39 mL, 18.13 mmol) was added to a solution of intermediate 199 (550 mg, 0.906 mmol) in DCM (10 mL) and stirred at rt for 3 h. The reaction mixture was concentrated under reduced pressure to give the TFA salt. TFA removal was done using SiliaBond@ propylsulfonic acid resin. The product was dissolved in MeOH and transferred to a column loaded with SiliaBond® propylsulfonic acid resin. The column was first eluted with MeOH after which the product was released by elution with ammoniated methanol (7 N). Tubes containing the product were concentrated under reduced pressure. The crude product was purified by silica gel column chromatography eluting with methanol (+1% 7N NH3 in methanol) in dichloromethane from 0% to 10% to give Compound 134 (350 mg, yield 76%). Compound 134 was further separated via Prep. SFC (Stationary phase: Chiralpak Diacel AD 20×250 mm, Mobile phase: CO2, EtOH+0.4 iPrNH2) and Prep. SFC (Stationary phase: Chiralcel Diacel IH 20×250 mm, Mobile phase: CO2, iPrOH+0.4 iPrNH2) to afford Compound 134a (25 mg, 5.4% yield), Compound 134b (95 mg, 21% yield), Compound 134c (115 mg, 25% yield) & Compound 134d (36 mg, 7.7% yield).


Preparation of Compound 142:



embedded image


A stir bar, intermediate 209 (50 mg, 0.086), EDCI (22 mg, 0.115 mmol), HOBt (21 mg, 0.114 mmol), DIEA (60 mg, 0.464 mmol), DCM (1 mL) and dimethylamine hydrochloride (16 mg, 0.196 mmol) were added into a 8 mL glass. The resulting mixture was stirred at room temperature for 4 h. The reaction mixture was poured it into water (5 m&), separated the layers, and the aqueous layers was extracted with DCM (5 mL×2). The combined organic extracts were dried over anhydrous Na2S4, filtered, and concentrated to dryness under reduced pressure to give the crude product which was purified by prep. HPLC (Column: Welch Xtimate C18 150*30 mm*5 um, Mobile Phase A: water (NH3H2O+NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 35 mE/min, gradient condition from 43% B to 73% B). The pure fractions were collected and the solvent was evaporated under vacuum to give a residue. The residue was partitioned between acetonitrile (2 mE) and water (10 mL). The solution was lyophilized to dryness to give Compound 142 (19 mg, 38.8% yield) as white power.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 142


Alternatively, purification can also be performed using the following method: prep-HPLC (Column: Welch Xtimate C18 150*25 mm*5 um, Mobile Phase A: water (+HCOOH), Mobile Phase B: acetonitrile Flow rate: 25 mL/min gradient condition from 2% B to 32% B).














Compound No.
Structure
Starting Materials







Compound 143


embedded image


Intermediate 209 & ethanamine hydrochloride





Compound 155


embedded image


Intermediate 209 & 3- aminopropanenitrile





Compound 158


embedded image


Intermediate 209 & 2- methoxyethanamine





Compound 159


embedded image


Intermediate 209 & cyclopropanamine





Compound 178


embedded image


Intermediate 247 & azetidine hydrochloride





Compound 179


embedded image


Intermediate 248 & azetidine hydrochloride





Compound 185


embedded image


Intermediate 247 & methylamine hydrochloride





Compound 221


embedded image


intermediate 292 & dimethylamine hydrochloride





Compound 236


embedded image


Intermediate 316 & methanamine hydrochloride





Compound 241


embedded image


Intermediate 292 & methanamine hydrochloride









Preparation of Compound 144:



embedded image


Intermediate 211 (35.8 mg, 0.0582 mmol) and AcOH (6.66 μL, 0.116 mmol) were stirred in MeOH (0.581 mL) at rt for 30 min. Sodium cyanoborohydride (7.3 mg, 0.116 mmol) was added and the reaction was heated at 50° C. The reaction mixture was quenched with water and used as such for reversed-phase prep HPLC purification (Stationary phase: RP XBridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give Compound 144 (13.2 mg, 44.8% yield) as a white solid.


Preparation of Compound 148:



embedded image


To a solution of intermediate 202 (60 mg, 0.14 mmol) in acetonitrile (5 mL) was added intermediate 216 (354 mg, 0.21 mmol), potassium carbonate (59 mg, 0.42 mmol) and potassium iodide (14 mg, 0.09 mmol). The reaction mixture was heated at 80° C. for 16 h. The mixture was cooled to room temperature, diluted with EtOAc and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 6% methanol in dichloromethane and prep. HPLC (Column: Xbridge C18 (5 μm 19*150 mm), Mobile Phase A: Water (0.1% ammonium bicarbonate), Mobile Phase B: acetonitrile, UV: 214 nm, Flow rate: 15 mL/min, Gradient: 15% B to 75% % B).


The Following Compound was Synthesized by an Analogous Method as Described for Compound 148

The product obtained from the alkylation step was immediately treated with 7M HCl in ethyl acetate.














Compound No.
Structure
Starting Materials







Compound 199


embedded image


Intermediate 202 & 260









Preparation of Compound 152:



embedded image


To a solution of Compound 448 (80 mg, 0.152 mmol) in tetrahydrofuran (4 mL) was added intermediate 221 (55 mg, 0.304 mmol). The reaction mixture was heated to 80° C. and stirred at this temperature overnight. The resulting mixture was concentrated and the residue was purified by silica gel column chromatography eluting with methanol in dichloromethane from 0% to 10% to give Compound 152 (65 mg, 70.6% yield) as a white solid.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 152














Compound No.
Structure
Starting Materials







Compound 157


embedded image


Compound 451 & 221









Preparation of Compound 153:



embedded image


Intermediate 222 (50 mg, 0.2 mmol) was added to a stirred mixture of intermediate 202 (81.6 mg, 0.2 mmol), sodium iodide (32.9 mg, 0.22 mmol) and K2CO3 (55.2 mg, 0.399 mmol) in MeCN (1.6 mL) and the mixture was heated at 80° C. overnight. The mixture was cooled down to rt, quenched with water, and extracted with EtOAc (×3). Reunited organic phases were dried over anhydrous sodium sulfate, filtered, evaporated to dryness and purified by silica gel column chromatography eluting with methanol (+1% NH3 in MeOH) in dichloromethane from 1% to 10% to give Compound 153 (71 mg, yield 58%) as a white solid.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 153














Compound No.
Structure
Starting Materials







Compound 154


embedded image


Intermediate 202 & 223









Preparation of Compound 162:



embedded image


A stir bar, Compound 526a (50.0 mg, 0.089 mmol) and methanamine (2 mL, 30% in ethanol) were added to a 8 mL glass bottle. The reaction mixture was heated and stirred at 70° C. for 4 days. The reaction mixture was cooled down to room temperature and concentrated under reduced pressure to give a residue, which was purified by preparative-HPLC (Column: Boston Prime C18 150*30 mm*5 um, Mobile Phase A: water (CH3COOH+CH3COONH4), Mobile Phase B: acetonitrile, Flow rate: 25 mL/min, gradient condition from 35% B to 65% B). The pure fractions were collected and the solvent was evaporated under vacuum. The residue was partitioned between acetonitrile (2 mL) and water (10 mL). The mixture was lyophilized to dryness to give Compound 162 (20.0 mg, 97.66% purity, 39.13% yield) as a white powder.


The Following Compound was Synthesized by an Analogous Method as Described for Compound 162














Compound No.
Structure
Starting Materials







Compound 163


embedded image


Compound 526b









Preparation of Compound 172:



embedded image


To a solution of Compound 448 (30 mg, 0.0593 mmol) and DIPEA (0.102 mL, 0.59 mmol) in DCM (6 mL) was added triphosgene (48.1 mg, 0.162 mmol). The mixture was stirred at r.t. for 0.5 hour. 2-methoxy-N-methylethan-1-amine (5.288 mg, 0.0593 mmol) was added and the mixture was stirred for further 2 hours. The solvent was removed and the residue was dissolved in MeOH (3 ml) and purified by preparative-HPLC (Column: Waters Sunfire C18 5 μm, 19*150 mm, Mobile Phase A: water (0.1% HCOOH), Mobile Phase B: acetonitrile, Flow rate: 17 mL/min, gradient condition from 0% B to 30% B) to afford Compound 172 (10 mg, 23% yield).


Preparation of Compound 184, 184a & 184b:



embedded image


NaH (60% dispersion in mineral oil) (7.9 mg, 0.197 mmol) was added, under nitrogen at 0° C., to a solution of Compound 169 (70 mg, 0.131 mmol) in anhydrous DMF (1 mL). After 10 min, Mel (9.8 μL, 0.157 mmol) was added, and the reaction was left under stirring at rt overnight. The reaction was quenched with ice, diluted with MeOH to give the crude product, which was used as such for reversed-phase prep HPLC purification (Stationary phase: RP Xbridge Prep C18 OBD-5 μm, 50×250 mm, Mobile phase: 0.5% NH4HCO3 solution in water, CH3CN) to give Compound 184 (30.5 mg, 41.1% yield) as a white solid. A purification was performed via Prep. SFC (Stationary phase: Chiralpak Diacel AD 20×250 mm, Mobile phase: CO2, EtOH-iPrOH (50-50)+0.4% iPrNH2). The first fraction was collected as Compound 184a (9.5 mg, 13% yield) and the second fraction as Compound 184b (9.5 mg, 13% yield) as white solids.


Preparation of Compound 205, 205a & 205b:



embedded image


NaBH4 (9.6 mg, 0.25 mmol) was added to a stirred solution of intermediate 264 (66 mg, 0.127 mmol) in MeOH (1.2 mL) at r.t. and the mixture was left under stirring for 20 min. The reaction was quenched with water and purified by Prep. HPLC to give Compound 205 (41 mg, yield 61.9%) as a white solid. A further purification was performed via Prep. SFC (Stationary phase: Chiralpak Diacel AD 20×250 mm, Mobile phase: CO2, EtOH-iPrOH (50-50)+0.4% iPrNH2) to give Compound 205a (25 mg, 38% yield) and Compound 205b (7 mg, 10.6% yield).


Preparation of Compound 207:



embedded image


Compound 448 (120 mg, 0.237 mmol) and DIPEA (0.12 mL, 0.71 mmol) were added to DCM (5 mL). Isocyanatotrimethylsilane (32.8 mg, 0.28 mmol) was added and the mixture was stirred at r.t. for 16 hours. The solvent was removed and the residue was purified by preparative-HPLC (Column: Waters Xbridge C18 5 μm, 19*150 mm, Mobile Phase A: water (0.1% NH4OH+10 mM NH4HCO3), Mobile Phase B: acetonitrile, Flow rate: 17 mL/min, gradient condition from 25% B to 35% B) to give Compound 207 (100 mg, 75% yield).


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 207














Compound No.
Structure
Starting Materials







Compound 217


embedded image


Compound 468





Compound 218


embedded image


Compound 469





Compound 219


embedded image


Compound 430









Preparation of Compound 261:



embedded image


1-azaspiro[3.3]heptane (0.165 mmol, 1.2 eq.) was pre-weighed into a 2-dram vial. A stock solution (23 mL) of intermediate 366 (1.38 g, 0.14 M), HATU (1.8 g, 0.2 M) and DIPEA (1.32 mL, 0.36 M) was prepared in DMF and stirred for 1 h. A 2nd stock solution of DIPEA (1.32 mL in 11.5 mL DMF) was also prepared. The DIPEA stock solution (0.5 mL) was added to each vial to solubilize the amine HCl salt. Intermediate 366/HATU/DIPEA solution (1 mL) was then added to each amine well. The reactions were stirred for 2 h, whereupon an extra 1.5 eq. HATU was added, and stirring continued overnight. The solvent was evaporated, and the samples redissolved in DMSO (0.5 mL) and MeCN (2.5 mL) for purification. Purification was performed via Prep HPLC (Stationary phase: RP XBridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN) to give Compound 261 (3.6 mg, 4.8% yield) after lyophilization.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 261


Alternative purification methods that can be employed for the purification of examples listed below are as follows:


Purifications can also be performed via Prep HPLC (Stationary phase: RP Xselect CSH Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.1% FA solution in water, CH3CN or MeOH).


Purifications can also be performed via Prep HPLC: (Stationary phase: RP Xbridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, MeOH).


Purifications can also be performed via Prep SFC (Stationary phase: Torus Diol 30×150 mm, Mobile phase: CO2, MeOH+20 mM NH4OH).


These purification methods can also be used in combination.














Compound No.
Structure
Starting Materials







Compound 262


embedded image


intermediate 366 & 2- azabicyclo[3.1.0]hexane hydrochloride





Compound 263


embedded image


intermediate 366 & 3-oxa-6-aza- bicyclo[3.1.1]heptane tosylate





Compound 264


embedded image


intermediate 366 & 2-oxa-5- azabicyclo[2.2.1]heptane, hydrochloride (1:1)





Compound 265


embedded image


intermediate 366 & N- cyclopropylmethylamine





Compound 266


embedded image


intermediate 366 & 6-oxa-1- azaspiro[3.3]heptane oxalate(2:1)





Compound 267


embedded image


intermediate 366 & 2- (methoxymethyl)pyrrolidine





Compound 268


embedded image


intermediate 366 & 2- (hydroxymethyl)pyrrolidine





Compound 269


embedded image


intermediate 366 & N- methyltetrahydrofuran-3-amine





Compound 270


embedded image


intermediate 366 & (3R,5R)-5- methylpyrrolidin-3-ol hydrochloride





Compound 271


embedded image


intermediate 366 & rel-(2R,3S)- 2-methylpyrrolidin-3-ol hydrochloride





Compound 272


embedded image


intermediate 366 & 2,2- dimethylpyrrolidin-3-ol





Compound 273


embedded image


intermediate 366 & 2- azabicyclo[2.1.1]hexan-4-ol hydrochloride





Compound 274


embedded image


intermediate 366 & cis-3- (methylamino)cyclobutan-1-ol hydrochloride





Compound 275


embedded image


intermediate 366 & trans-3- (methylamino)cyclobutanol





Compound 276


embedded image


intermediate 366 & [1- (methylamino)cyclobutyl] methanol





Compound 277


embedded image


intermediate 366 & (1- (methylamino)cyclopropyl) methanol hydrochloride





Compound 278


embedded image


intermediate 366 & 2- (cyclopropylamino)ethanol





Compound 279


embedded image


intermediate 366 & (R)-(−)-2- methylpyrrolidine





Compound 280


embedded image


intermediate 366 & 2- methylpiperidine





Compound 281


embedded image


intermediate 366 & 2- (isopropylamino)ethanol





Compound 282


embedded image


intermediate 366 & (3R,5S)-5- methylpyrrolidin-3-ol hydrochloride









Preparation of Compound 283:



embedded image


The 3-formyl-N-methylbenzamide (0.4 mmol, 2 eq.) was pre-weighed into a 2-dram vial with a stirrer bar. Stock solutions of intermediate 25 (0.79 g, 7.5 mL, 0.27 M) and sodium cyanoborohydride (0.23 g, 7.5 mL, 0.48 M) were prepared in MeOH. 0.75 mL of intermediate stock solution was added and the solutions stirred for 2 h. Next, sodium cyanoborohydride stock solution (0.75 mL) was then added. The reaction mixture was then stirred at room temperature overnight. After reaction completion, the solution was added to MeOH-washed ethylbenzenesulfonic acid resin cartridge (Isolute® SCX-3), and eluted with MeOH (3×2 mL) followed by 3.5 M NH3 in MeOH (3×2 mL). The basic washes containing the product was evaporated and re-dissolved in 3 mL 1:1 MeCN/MeOH for purification. Purification was performed via Prep SFC (Stationary phase: Torus Diol 30×150 mm, Mobile phase: CO2, MeOH+20 mM NH4OH) to give Compound 283 (41 mg, 38% yield), after lyophilization.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 283


For reactions employing ketone building blocks, the following applies: acetic acid was added (23 μL, 2 eq.) into the reaction mixture before the addition of the sodium cyanoborohydride stock solution. The reaction mixture was stirred at 50° C. overnight (during the reductive amination step).


Alternative purification methods that can be employed for the purification of examples listed below are as follows:


Purifications can also be performed via Prep HPLC (Stationary phase: RP Xselect CSH Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.1% FA solution in water, CH3CN or MeOH).


Purifications can also be performed via Prep HPLC: (Stationary phase: RP Xbridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN or MeOH).


These purification methods can also be used in combination.














Compound No.
Structure
Starting Materials







Compound 284


embedded image


Intermediate 25 & 6- methylpyridazine-3- carbaldehyde





Compound 285


embedded image


Intermediate 25 & 5-methyl-1H- pyrazole-3-carbaldehyde





Compound 286


embedded image


Intermediate 25 & 3- methyloxetane-3-carbaldehyde





Compound 287


embedded image


Intermediate 25 & tetrahydrofuran-3- carboxaldehyde





Compound 288


embedded image


Intermediate 25 & tetrahydro-2- furancarbaldehyde





Compound 289


embedded image


Intermediate 25 & tetrahydropyran-3-carbaldehyde





Compound 290


embedded image


Intermediate 25 & azepane-2,4- dione





Compound 291


embedded image


Intermediate 25 & 1- methylazepane-2,4-dione









Preparation of Compound 292a:



embedded image


(R)-tert-Butyl 2-methyl-4-oxopiperidine-1-carboxylate (0.4 mmol, 2 eq.) was pre-weighed into a 2-dram vial with a stirrer bar. Stock solutions of intermediate 25 (0.63 g, 6.0 mL, 0.27 M) and sodium cyanoborohydride (0.18 g, 6.0 mL, 0.48 M) were prepared in MeOH. 0.75 mL of intermediate 25 stock solution was added to the reaction vial, together with acetic acid (23 μL, 2 eq.) and the solutions stirred for 1 h. The sodium cyanoborohydride stock solution (0.75 mL) was then added. The reaction mixtures were stirred at 50° C. overnight. After reaction completion, the solutions were added to MeOH-washed ethylbenzenesulfonic acid resin cartridge (Isolute® SCX-3), and eluted with MeOH (3×2 mL) followed by 3.5 M NH3 in MeOH (3×2 mL). The basic washes containing the product were evaporated.


The crude products from the reductive amination were dissolved in DCM (1 mL) and TFA (2 mL), and stirred at 50° C. for 1 h. The solvents were evaporated and redissolved in MeCN (2 mL). Siliamet® Diamine resin was added and the mixture stirred for 0.5 h. The resin was removed via filtration on a 24-well filter plate, and the filtrate concentrated.


The Boc deprotected products were dissolved in 1 mL DCM, and DIPEA (0.55 mL, 3.2 mmol), and Ac2O (0.25 mL, 2.6 mmol) were added. The reaction mixture was stirred for 2 h at room temperature, at which time they were quenched with MeOH (2 mL) and concentrated. The samples were re-dissolved in 3 mL 1:1 MeCN/MeOH for purification. Purification was performed via Prep SFC (Stationary phase: Torus Diol 30×150 mm, Mobile phase: CO2, MeOH+20 mM NH4OH) to give Compound 292a (22.9 mg, yield: 21%), after lyophilization.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 292a


For reactions employing ketone building blocks, the following applies: acetic acid was added (23 μL, 2 eq.) into the reaction mixture before the addition of the sodium cyanoborohydride stock solution. The reaction mixture was stirred at 50° C. overnight (during the reductive amination step).


Alternative purification methods that can be employed for the purification of examples listed below are as follows:


Purifications can also be performed via Prep HPLC (Stationary phase: RP Xselect CSH Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.100 FA solution in water, CH3CN or MeOH).


Purifications can also be performed via Prep HPLC: (Stationary phase: RP Xbridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN or MeOH).


These purification methods can also be used in combination.














Compound No.
Structure
Starting Materials







Compound 292b


embedded image


Intermediate 25 & (R)-tert-Butyl 2-methyl-4-oxopiperidine-1- carboxylate





Compound 293a


embedded image


Intermediate 25 & (S)-tert-Butyl 2-methyl-4-oxopiperidine-1- carboxylate





Compound 293b


embedded image


Intermediate 25 & (S)-tert-Butyl 2-methyl-4-oxopiperidine-1- carboxylate





Compound 294a


embedded image


Intermediate 25 & 2-Boc-5- oxohexahydrocyclopenta[c]pyrrole





Compound 294b


embedded image


Intermediate 25 & 2-Boc-5- oxohexahydrocyclopenta[c]pyrrole





Compound 295


embedded image


Intermediate 25 & N-Boc- hexahydro-1H-azepin-4-one





Compound 296


embedded image


Intermediate 25 & tert-butyl 1- oxo-7-azaspiro[3.5]nonane-7- carboxylate









Preparation of Intermediate 393:



embedded image


1-(1-[(tert-butoxy)carbonyl]piperidin-2-yl)cyclopropane-1-carboxylic acid (0.40 g, 1.5 mmol) was dissolved in anhydrous THF (20 mL). Then, the mixture was cooled to 0° C. in an ice bath. Next, BH3-THF (1M solution, 2.2 mL, 2.2 mmol) was added dropwise. The mixture was then allowed to stir at ambient temperature for ˜2 h, after which LC/MS showed full conversion of the starting material. Then, water was carefully added. After gas formation ceased, solid K2CO3 (0.26 g) was added and the mixture stirred at ambient temperature for −30 min. Then, EA was added and the mixture transferred to a separatory funnel. The layers were separated and the water layer was extracted twice more with EA. Organic layers were combined, dried over Na2SO4, filtered and evaporated. The residue was purified by silica gel chromatography eluting with ethyl acetate in petroleum ether from 30% to 80% to give Intermediate 393 (0.36 g, 1.4 mmol, yield: 95%) as an oil.


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 393














Intermediate No.
Structure
Starting Materials







Intermediate 394


embedded image


1-Boc-(1carboxy-cyclopropyl)- piperidine





Intermediate 395


embedded image


3-(1,1-Dimethylethyl)(7-exo)-3- azabicyclo[3.3.1]nonane-3,7- dicarboxylate









Preparation of Intermediate 396:



embedded image


Intermediate 393 (0.15 g, 0.59 mmol) was dissolved in ethyl acetate (4 mL). Next, IBX (0.49 g, 1.8 mmol) was added and the resulting mixture was heated to 80° C., open to air. After 3 h, TLC analysis (50% EA/Heptane) showed full conversion of the starting material. The mixture was cooled to ambient temperature, and filtered. The filter cake was washed once with EA. The filtrate was evaporated to dryness to give intermediate 396 (0.13 g, 0.51 mmol, yield: 87%).


The Following Intermediates were Synthesized by an Analogous Method as Described for Intermediate 396














Intermediate No.
Structure
Starting Materials







Intermediate 397


embedded image


Intermediate 394





Intermediate 398


embedded image


Intermediate 395









Preparation of Compound 297:



embedded image


Intermediate 396 (0.4 mmol, 2 eq.) was pre-weighed into 2-dram vials with a stirrer bar. Stock solutions of intermediate 25 (0.63 g, 6.0 mL, 0.27 M) and sodium cyanoborohydride (0.18 g, 6.0 mL, 0.48 M) were prepared in MeOH. 0.75 mL of intermediate 25 stock solution was added to the vial and the solution stirred for 1 h. The sodium cyanoborohydride stock solution (0.75 mL) was then added. The reaction mixture was stirred at room temperature (for aldehydes). After reaction completion, the solutions were added to MeOH-washed ethylbenzenesulfonic acid resin cartridge (Isolute® SCX-3) cartridge, and eluted with MeOH (3×2 mL) followed by 3.5 M NH3 in MeOH (3×2 mL). The basic washes containing the product were evaporated to dryness.


The crude products from the reductive amination were dissolved in DCM (1 mL) and TFA (2 mL), and stirred at 50° C. for 1 h. The solvents were evaporated and redissolved in MeCN (2 mL). Siliamet® Diamine resin was added and the mixtures stirred for 0.5 h. The resin was removed via filtration on a 24-well filter plate, and the filtrate concentrated.


The Boc deprotected products were dissolved in 1 mL DCM, and DIPEA (0.55 mL, 3.2 mmol), and Ac2O (0.25 mL, 2.6 mmol) were added. The reaction mixtures were stirred for 2 h at room temperature, at which time they were quenched with MeOH (2 mL) and concentrated. The samples were re-dissolved in 3 mL 1:1 MeCN/MeOH for purification. Purification was performed via Prep SFC (Stationary phase: Torus Diol 30×150 mm, Mobile phase: CO2, MeOH+20 mM NH4OH) to give Compound 297 (91 mg, yield=79%) after lyophilization.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 297 For reactions employing ketone building blocks, the following applies: acetic acid was added (23 μL, 2 eq.) into the reaction mixture before the addition of the sodium cyanoborohydride stock solution. The reaction mixture was stirred at 50° C. overnight (during the reductive amination step).


Alternative purification methods that can be employed for the purification of examples listed below are as follows:


Purifications can also be performed via Prep HPLC (Stationary phase: RP Xselect CSH Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.1% FA solution in water, CH3CN or MeOH).


Purifications can also be performed via Prep HPLC: (Stationary phase: RP Xbridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN or MeOH).


These purification methods can also be used in combination.














Compound No.
Structure
Starting Materials







Compound 298


embedded image


Intermediate 25 & Intermediate 397





Compound 299a


embedded image


Intermediate 25 & tert-butyl cis 3,5-dimethyl-4-oxopiperidine-1- carboxylate





Compound 300


embedded image


Intermediate 25 & Intermediate 398





Compound 301


embedded image


Intermediate 25 & cis-2,6- dimethyl-4-oxo-piperidine-1- carboxylic acid tert-butyl ester





Compound 302


embedded image


Intermediate 25 & trans-2,6- dimethyl-4-oxo-piperidine-1- carboxylic acid tert-butyl ester





Compound 303


embedded image


Intermediate 25 & 2-Boc-7-oxo- 2-azaspiro[4.5]decane





Compound 304


embedded image


Intermediate 25 & (1R,4S,5S)- rel-tert-Butyl 5-acetyl-2- azabicyclo[2.1.1]hexane-2- carboxylate





Compound 305


embedded image


Intermediate 25 & tert-butyl rel- (1R,5S,6s)-6-formyl-3- azabicyclo[3.1.0]hexane-3- carboxylate





Compound 306


embedded image


Intermediate 25 & 4- oxohexahydrocyclopenta[c]pyrrole- 2-carboxylic acid tert-butyl ester





Compound 307


embedded image


Intermediate 25 & 4- hydroxytetrahydro-2H-pyran-4- carbaldehyde





Compound 308


embedded image


Intermediate 25 & 3-methyl- 1,2,4-oxadiazole-5-carbaldehyde





Compound 309a


embedded image


Intermediate 25 & hexahydroindolizine-3.7-dione





Compound 309b


embedded image


Intermediate 25 & hexahydroindolizine-3,7-dione





Compound 299b


embedded image


Intermediate 25 & tert-butyl cis 3,5-dimethyl-4-oxopiperidine-1- carboxylate









Preparation of Compound 310:



embedded image


1-azaspiro[3.3]heptane hydrochloride (0.17 mmol, 1.2 eq.) were pre-weighed into a 2-dram vial. A stock solution (21 mL) of intermediate 370 (1.26 g, 0.12 M), HATU (1.46 g, 0.18 M) and DIPEA (1.1 mL) was prepared in DMF and stirred for 1 h. A 2nd stock solution of DIPEA (1.1 mL in 10.5 mL DMF) was also prepared. The DIPEA stock solution (0.5 mL) was added to each the vial to solubilize the amine HCl salt. Intermediate 370/HATU/DIPEA solution (1 mL) was then added. The reaction was stirred overnight at room temperature. The DMF was removed by evaporation. The crude was redissolved in DCM/EtOAc 2/1 (2.2 mL), and water (2.2 mL) was added. The mixture was stirred for 10 minutes. Then, the mixture was left standing for 10 minutes. 2 mL of the organics were removed using a pipette and these were filtered over a fritted filter. The remaining water layer was extracted another time using 2 mL of the DCM/EtOAc mixture. Again, 2 mL was removed and filtered over the same fritted filter. The fritted filter was rinsed with 500 μL of DMSO. The obtained filtrates were concentrated in vacuo until only DMSO remained. The obtained crude were redissolved in MeOH/MeCN 1/1 (2.2 mL) and submitted for purification. Purification was performed via Prep HPLC (Stationary phase: RP XBridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN) to give Compound 310 (8.7 mg, yield: 12%) after lyophilization.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 310


Alternative purification methods that can be employed for the purification of examples listed below are as follows:


Purifications can also be performed via Prep HPLC (Stationary phase: RP Xselect CSH Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.1% FA solution in water, CH3CN or MeOH).


Purifications can also be performed via Prep HPLC: (Stationary phase: RP Xbridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, MeOH).


Purifications can also be performed via Prep SFC (Stationary phase: Torus Diol 30×150 mm, Mobile phase: CO2, MeOH+20 mM NH4OH).


These purification methods can also be used in combination.














Compound No.
Structure
Starting Materials







Compound 311


embedded image


Intermediate 370 & 2- azabicyclo[3.1.0]hexane hydrochloride





Compound 311a & Compound 311b


embedded image


Compound 311 was separated by Prep SFC (Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, EtOH- iPrOH (50-50) + 0.4% iPrNH2). The first fraction was collected as Compound 311a & the second fraction as Compound 311b.








embedded image








Compound 312


embedded image


Intermediate 370 & 2-oxa-5- azabicyclo[2.2.1]heptane HCl





Compound 313


embedded image


Intermediate 370 & 2,2- dimethylpyrrolidin-3-ol





Compound 313a & Compound 313b


embedded image


Compound 313 was separated by Prep SFC (Stationary phase: Chiralpak Daicel ID 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2). The first fraction was collected as Compound 313a & the second fraction as Compound 313b.





Compound 314


embedded image


Intermediate 370 & N-(methyl- d3)propan-2-amine





Compound 315


embedded image


Intermediate 370 & (R)-(−)-2- methylpyrrolidine





Compound 316


embedded image


Intermediate 370 & (2S,5S)-2,5- dimethylpyrrolidine hydrochloride





Compound 317


embedded image


Intermediate 370 & 2- methylpiperidine





Compound 318


embedded image


Intermediate 370 & 4-methoxy- 2-methyl-pyrrolidine





Compound 319


embedded image


Intermediate 370 & (S)-(+)-2- methylpyrrolidine





Compound 320


embedded image


Intermediate 370 & 1-(propan-2- ylamino)propan-2-ol





Compound 320a & Compound 320b


embedded image


Compound 320 was separated by Prep SFC(Stationary phase: Chiralpak Diacel AD 20 × 250 mm, Mobile phase: CO2, iPrOH + 0.4 iPrNH2). The first fraction was collected as Compound 320a & the second fraction as Compound 320b.








embedded image








Compound 321


embedded image


Intermediate 370 & 2- (methoxymethyl)pyrrolidine





Compound 322


embedded image


Intermediate 370 & (R)-2- pyrrolidinemethanol





Compound 323


embedded image


Intermediate 370 & (S)-2- pyrrolidinemethanol





Compound 324


embedded image


Intermediate 370 & (3R,5R)-5- methylpyrrolidin-3-ol hydrochloride





Compound 325


embedded image


Intermediate 370 & rel-(2R,3S)- 2-methylpyrrolidin-3-ol hydrochloride





Compound 326


embedded image


Intermediate 370 & 2- (isopropylamino)ethanol





Compound 327


embedded image


Intermediate 370 & (2R,4S)-4- hydroxy-2-methylpyrrolidine hydrochloride





Compound 354


embedded image


Intermediate 370 & (2R,5R)-2,5-dimethylpyrrolidine





Compound 355


embedded image


Intermediate 370 & N-(2- methoxyethyl)propan-2-amine





Compound 356


embedded image


Intermediate 370 & N- methylcyclopropanamine









Preparation of Compound 328:



embedded image


A stock solution of intermediate 374 (39.1 mg, 0.075 mmol), DIPEA (0.038 mL, 0.2 mmol) and HATU (42.8 mg, 0.1 mmol) in DMF (0.56 mL) was added to a pre-weighed (S)-(+)-2-pyrrolidinemethanol (0.15 mmol, 2 equiv). The resulting solution was stirred for 2 h at rt. Afterwards the solvent was removed under reduced pressure. To the vial was added 2 mL of a DCM/EtOAc=2/1 mix and 1 mL aq. 1N citric acid solution and mixture was stirred for 5 minutes. The organic phase was collected, and the solvent removed under reduced pressure. 3 mL of a 2/1 mix MeOH/MeCN was added. Purification was performed via Prep HPLC (Stationary phase: RP XBridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN) to give Compound 328 (31 mg, 69% yield) after lyophilization.


The Following Compounds were Synthesized by an Analogous Method as Described for Compound 328


Alternative purification methods that can be employed for the purification of examples listed below are as follows:


Purifications can also be performed via Prep HPLC (Stationary phase: RP Xselect CSH Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.1% FA solution in water, CH3CN or MeOH).


Purifications can also be performed via Prep HPLC: (Stationary phase: RP Xbridge Prep C18 OBD-10 μm, 30×150 mm, Mobile phase: 0.25% NH4HCO3 solution in water, MeOH).


Purifications can also be performed via Prep SFC (Stationary phase: Torus Diol 30×150 mm, Mobile phase: CO2, MeOH+20 mM NH4OH).


These purification methods can also be used in combination.














Compound No.
Structure
Starting Materials







Compound 329


embedded image


Intermediate 374 & 2- (isopropylamino)ethanol





Compound 330


embedded image


Intermediate 374 & (2S)-1-[(1- methylethyl)amino]-2-propanol





Compound 331


embedded image


Intermediate 374 & (2R)-1-[(1- methylethyl)amino]-2-propanol





Compound 332


embedded image


Intermediate 374 & (3R,5R)-5- methylpyrrolidin-3-ol hydrochloride





Compound 333


embedded image


Intermediate 374 & 5- methylpyrrolidin-3-ol





Compound 334


embedded image


Intermediate 376 & 2- (isopropylamino)ethanol





Compound 335


embedded image


Intermediate 376 & (2S)-1-[(1- methylethyl)amino]-2-propanol





Compound 336


embedded image


Intermediate 376 & (2R)-1-[(1- methylethyl)amino]-2-propanol





Compound 337


embedded image


Intermediate 380 & 2- (isopropylamino)ethanol





Compound 338


embedded image


Intermediate 380 & (2S)-1-[(1- methylethyl)amino]-2-propanol





Compoumd 339


embedded image


Intermediate 380 & (2R)-1-[(1- methylethyl)amino]-2-propanol





Compound 340


embedded image


Intermediate 382 & 4- azaspiro[2.4]heptane hydrochloride





Compound 341


embedded image


Intermediate 382 & 2- azabicyclo[3.1.0]hexane hydrochloride





Compound 342


embedded image


Intermediate 382 & 2- ethylpyrrolidine





Compound 343


embedded image


Intermediate 382 & (2S,5S)-2,5- dimethylpyrrolidine hydrochloride





Compound 344


embedded image


Intermediate 382 & cis-2,5- dimethyl-pyrrolidine hydrochloride





Compound 345


embedded image


Intermediate 382 & 2-oxa-5- azabicyclo[2.2.1]heptane





Compound 346


embedded image


Intermediate 382 & (R)-2- methylpyrrolidine-hydrochloride





Compound 347


embedded image


Intermediate 384 & (S)-(+)-2- pyrrolidinemethanol





Compound 348


embedded image


Intermediate 384 & 1-(propan-2- ylamino)propan-2-ol





Compound 349


embedded image


Intermediate 388 & (S)-(+)-2- pyrrolidinemethanol





Compound 350


embedded image


Intermediate 388 & 1-(propan-2- ylamino)propan-2-ol





Compound 351a & Compound 351b


embedded image


Intermediate 309 & 2- azabicyclo[3.1.0]hexane hydrochloride The product was separated by Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 351a & the second fraction as Compound 351b.








embedded image








Compound 352a & Compound 352b


embedded image


Intermediate 390 & (2R,5R)-2,5- dimethylpyrrolidine hydrochloride The product was separated by Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 352a & the second fraction as Compound 352b.








embedded image








Compound 353a & Compound 353b


embedded image


Intermediate 390 & R-(−)-2- methylpyrrolidine The product was separated by Prep SFC (Stationary phase: Chiralcel Diacel IH 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 353a & the second fraction as Compound 353b.








embedded image








Compound 357


embedded image


Intermediate 392 & 2- (isopropylamino)ethan-1-ol





Compound 358


embedded image


Intermediate 392 & rel-(2R,3S)- 2-methylpyrrolidin-3-ol hydrochloride





Compound 359


embedded image


Intermediate 392 & (3R,5R)-5- methylpyrrolidin-3-ol





Compound 360


embedded image


Intermediate 392 & (S)- pyrrolidin-2-ylmethanol





Compound 361


embedded image


Intermediate 392 & (R)- pyrrolidine-2-ylmethanol





Compound 362a & Compound 362b


embedded image


Intermediate 392 & (2-methyl-2- pyrrolidinyl)methanol The product was separated by Prep SFC (Stationary phase: Torus Diol 30 × 150 mm, Mobile phase: CO2, MeOH + 20 mM NH4OH). The first fraction was collected as Compound 362a & the second fraction as Compound 362b.








embedded image








Compound 363a & Compound 363b


embedded image


Intermediate 392 & 1-[(2R)- pyrrolidin-2-yl]ethan-1-ol The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IG 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 363a & the second fraction as Compound 363b.








embedded image








Compound 364a & Compound 364b


embedded image


Intermediate 392 & 2,2- dimethylpyrrolidin-3-ol The product was separated by Prep SFC (Stationary phase: Chiralcel Diacel OD 20 × 250 mm, Mobile phase: CO2, MeOH + 0.4 iPrNH2). The first fraction was collected as Compound 364a & the second fraction as Compound 364b.








embedded image








Compound 365a & Compound 365b


embedded image


Intermediate 392 & 1- isopropylamino-propan-2-ol The product was separated by Prep SFC (Stationary phase: Chiralpak Daicel IG 20 × 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2). The first fraction was collected as Compound 365a & the second fraction as Compound 365b.








embedded image











LCMS (Liquid Chromatography/Mass Spectrometry)
General Procedure

The High Performance Liquid Chromatography (HPLC) measurement was performed using a LC pump, a diode-array (DAD) or a UV detector and a column as specified in the respective methods. If necessary, additional detectors were included (see table of methods below).


Flow from the column was brought to the Mass Spectrometer (MS) which was configured with an atmospheric pressure ion source. It is within the knowledge of the skilled person to set the tune parameters (e.g. scanning range, dwell time . . . ) in order to obtain ions allowing the identification of the compound's nominal monoisotopic molecular weight (MW). Data acquisition was performed with appropriate software.


Compounds are described by their experimental retention times (Rt) and ions. If not specified differently in the table of data, the reported molecular ion corresponds to the [M+H]+ (protonated molecule) and/or [M−H] (deprotonated molecule). In case the compound was not directly ionizable the type of adduct is specified (i.e. [M+NH4]+, [M+HCOO], etc. . . . ). For molecules with multiple isotopic patterns (Br, Cl . . . ), the reported value is the one obtained for the lowest isotope mass. All results were obtained with experimental uncertainties that are commonly associated with the method used.


Hereinafter, “SQD” means Single Quadrupole Detector, “RT” room temperature, “BEH” bridged ethylsiloxane/silica hybrid, “HSS” High Strength Silica, “DAD” Diode Array Detector.









TABLE 1a







LCMS Method codes (Flow expressed in mL/min; column temperature (T) in ° C.;


Run time in minutes). “TFA” means trifluoroacetic acid; “FA” means formic acid


















Flow








(ml/min)








- - -
Run


Method


Mobile

Column
time


code
Instrument
Column
phase
Gradient
T (° C.)
(min)





1
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Techno-
5 um
CH3COONH4;
5% A in 4.00 min, held for
- - -




logies 1200
4.6*50 mm
B: CH3CN
1.50 min, back to 95% A in
40




Series,


0.10 min, held for 0.40





G6110A


min




2
Agilent
Xbridge C18,
A: 0.05%
95% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA;
50% A in 4.00 min, then
- - -




logies 1200
4.6*50 mm
B: CH3CN
to 5% A in 0.50 min, held
40




Series,


for 1.00 min, back to 95%





G6110A


A in 0.10 min, held for








0.40 min




3
Agilent
Xbridge C18,
A: 0.05%
95% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA;
5% A in 4.00 min, held for
- - -




logies 1200
4.6*50 mm
B: CH3CN
1.50 min, back to 95% A
40




Series,


in 0.10 min, held for 0.40





G6110A


min




4
Agilent
Xbridge C18,
A: 0.05%
90% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA; B:
70% A in 4.00 min, then
- - -




logies 1200
4.6*50 mm
CH3CN
to 5% A in 0.50 min, held
40




Series,


for 1.00 min, back to 90%





G6110A


A in 0.10 min, held for








0.40 min




5
Agilent
Xbridge C18,
A: 0.05%
98% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA; B:
60% A in 4.00 min, then
- - -




logies 1200
4.6*50 mm
CH3CN
to 5% A in 0.50 min, held
40




Series,


for 1.00 min, back to 98%





G6110A


A in 0.10 min, held for








0.40 min




6
Agilent
Xbridge C18,
A: 0.05%
95% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA; B:
5% A in 4.00 min, held for
- - -




logies 1200
4.6*50 mm
CH3CN
1.50 min, back to 95% A
40




Series,


in 0.10 min, held for 0.40





G6130A


min




7
Agilent
Xbridge C18,
A: 0.05%
95% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA; B:
50% A in 4.00 min, then
- - -




logies 1200
4.6*50 mm
CH3CN
to 5% A in 0.50 min, held
40




Series,


for 1.00 min, back to 95%





G6130A


A in 0.10 min, held for








0.40 min




8
Agilent
ZORBAX
A: 0.05%
90% A for 3.00 min, to
1.5
15



Techno-
SB-C8,
TFA; B:
5% A in 8.00 min, held for
- - -




logies 1200
3.5 um
CH3CN
3.60 min, back to 90% A
40




Series,
4.6*150 mm

in 0.10 min, held for 0.30





G6110A


min.




9
Agilent
Xbridge C18,
A: 0.05%
95% A for 0.50 min, to
1.5
6.5



Techno-
5 um
TFA; B:
40% A in 4.00 min, then
- - -




logies 1200
4.6*50 mm
CH3CN
to 5% A in 0.50 min, held
40




Series,


for 1.00 min, back to 95%





G6110A


A in 0.10 min, held for








0.40 min.




10
Agilent
Waters
mobile phase
First, 90% A was held for
0.8
10



1200-6100
XBridge
A: H2O with
0.8 min. Then a gradient
- - -





C18,
0.04% TFA;
was applied to 20% A and
50





(2.0 × 50 mm,
mobile phase B:
80% B in 3.7 min and held






5 uM)
ACN with
for 3 min. And then return







0.02% TFA
to 90% A in 2 min and








held for 0.5 min. The post








time is 0.5 min




11
Agilent
Agilent
mobile phase
a gradient condition from
1.5
3.0



Prime-
Poroshell
A:
95% A, 5% B to 20% A,
- - -




6125B
120 HPH-
water(4 L) + N
80% B in 1.2 min, then to
30





C18 1.9 um
H3•H2O(2.0
5% A, 95% B in 1.3 min,






3.0*30 mm
mL); mobile
and then to 95% A, 5% B







phase B:
in 0.01 min and held for







acetonitrile(4
0.49 min







L)





12
Agilent
Agilent
mobile phase
a gradient condition from
1.5
3.0



Prime-
Poroshell
A:
95% A, 5% B to 20% A,
- - -




6125B
120 EC-C18
water(4 L) + T
80% B in 1.2 min, then to
50





1.9 um
FA(1.5 mL)
5% A, 95% B in 1.3 min,






3.0*30 mm

and then to 95% A, 5% B








in 0.01 min and held for








0.49 min




13
Waters: H-
Waters:
A: 0.1%
Gradient start from 5% of
0.8
3.0



Class +
ACQUITY
NH4OH in
B increase to 95% within
- - -




Zspray
UPLC BEH
water, B:
1.5 min and keep at 95%
40





C18 (1.7 μm,
CH3CN
till 2.5 min, then decrease






2.1 × 30 mm)

to 5% within 0.01 min and








keep at 5% till 3.0 min




14
Agilent:
Waters:
A: 0.1% FA
Gradient start from 5% of
1.2
3.5



1260
Sunfire C18
solution in
B increase to 95% within
- - -




Infinity and 6120
(2.5 μm,
water; B:
2.5 min and keep at 95%
50




Quadrupole LC/MS
3.0 × 30 mm)
CH3CN
till 3.5 min




15
Waters:
Waters:
A: 10 mM
From 100% A to 5% A
0.6
3.5



Acquity ®
BEH
NH4HCO3 in
in 2.10 min, to 0% A in 0.9
- - -




UPLC ®-
(1.8 μm,
95% H2O +
min, to 5% A in 0.5 min
55




DAD and
2.1*100 mm)
5% CH3CN;






SQD

B: CH3CN





16
Waters:
Waters:
A: 10 mM
From 100% A to 5% A in
0.6
3.5



Acquity ®
BEH
NH4HCO3 in
2.10 min, to 0% A in 0.9
- - -




UPLC ® -
(1.8 μm,
95% H2O +
min, to 5% A in 0.5 min
55




DAD and
2.1*100 mm)
5% CH3CN;






SQD and

B: CH3CN






ELSD







17
Waters:
Waters:
A: 10 mM
From 100% A to 5% A in
0.6
3.5



Acquity ®
BEH
NH4HCO3 in
2.10 min, to 0% A in 0.9
- - -




UPLC ®-
(1.8 μm,
95% H2O +
min, to 5% A in 0.5 min
55




DAD and
2.1*100 mm)
5% CH3CN;






SQD

B: CH3CN





18
Agilent
Xbridge C18,
A: 0.02%

1.5
6.5



Technologies
3.5 um
CH3COONH4;
95% A for 0.50 min, to
- - -




1200
4.6*50 mm
B: CH3CN
5% A in 4.00 min, held for
40




Series,


1.50 min, back to 95% A





G6110A


in 0.10 min, held for 0.40








min.




19
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Technologies
3.5 um 4.6*50 mm
CH3COONH4;
5% A in 4.00 min, held for
- - -




1200

B: CH3CN
1.50 min, back to 95% A
40




Series,


in 0.10 min, held for 0.40





G6130A


min.




20
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Technologies
5 um 4.6*50 mm
CH3COONH4;
40% A in 4.00 min, then
- - -




1200

B: CH3CN
to 5% A in 0.50 min, held
40




Series,


for 1.00 min, back to 95%





G6110A


A in 0.10 min, held for








0.40 min.




21
Agilent
Xbridge C18,
A: 0.02%
85% A for 0.50 min, to
1.5
6.5



Technologies 1200
5 um 4.6*50 mm
CH3COONH4;
60% A in 4.00 min, then
- - -




Series,

B: CH3CN
to 5% A in 0.50 min, held
40




G6110A


for 1.00 min, back to 85%








A in 0.10 min, held for








0.40 min.




22
Agilent
Xbridge C18,
A: 0.05%
95% A for 0.50 min, to
1.5
6.5



Technologies 1200
3.5 um 4.6*50 mm
TFA; B:
5% A in 4.00 min, held for
- - -




Series,

CH3CN
1.50 min, back to 95% A
40




G6130A


in 0.10 min, held for 0.40








min




23
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Technologies 1200
3.5 um 4.6*50 mm
CH3COONH4;
5% A in 4.00 min, held for
- - -




Series,

B: CH3CN
1.50 min, back to 95% A
40




G6130A


in 0.10 min, held for 0.40








min.




24
Agilent
Xbridge C18,
A: 0.02%
80% A for 0.50 min, to
1.5
6.5



Technologies 1200
3.5 um 4.6*50 mm
CH3COONH4;
40% A in 4.00 min, then
- - -




Series,

B: CH3CN
to 5% A in 0.50 min, held
40




G6130A


for 1.00 min, back to 80%








A in 0.10 min, held for








0.40 min.




25
Agilent
Xbridge C18,
A: 0.02%
90% A for 0.50 min, to
1.5
6.5



Technologies 1200
5 um 4.6*50 mm
CH3COONH4;
50% A in 4.00 min, then
- - -




Series,

B: CH3CN
to 5% A in 0.50 min, held
40




G6130A


for 1.00 min, back to 90%








A in 0.10 min, held for








0.40 min.




26
Agilent
Xbridge C18,
A: 0.05%
85% A for 0.50 min, to
1.5
6.5



Technologies 1200
5 um 4.6*50 mm
TFA; B:
70% A in 4.00 min, then
- - -




Series,

CH3CN
to 5% A in 0.50 min, held
40




G6110A


for 1.00 min, back to 85%








A in 0.10 min, held for








0.40 min.




27
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Technologies 1200
5 um 4.6*50 mm
CH3COONH4;
5% A in 4.00 min, held for
- - -




Series,

B: CH3CN
1.50 min, back to 95% A
40




G6130A


in 0.10 min, held for 0.40








min.




28
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Technologies 1200
5 um 4.6*50 mm
CH3COONH4;
40% A in 4.00 min, then
- - -




Series,

B: CH3CN
to 5% A in 0.50 min, held
40




G6130A


for 1.00 min, back to 95%








A in 0.10 min, held for








0.40 min.




29
Agilent
Xbridge C18,
A: 0.02%
95% A for 0.50 min, to
1.5
6.5



Technologies 1200
5 um 4.6*50 mm
CH3COONH4;
50% A in 4.00 min, then
- - -




Series,

B: CH3CN
to 5% A in 0.50 min, held
40




G6110A


for 1.00 min, back to 95%








A in 0.10 min, held for








0.40 min.




30
Waters:
Waters:
A: 10 mM
From 100% A to 5% A in
0.6
3.5



Acquity ®
BEH
CH3COONH4
2.10 min, to 0% A in
- - -




UPLC ®-
(1.7 μm,
in 95% H2O + 5%
0.9 min, to 5% A in
55




DAD and
2.1*100 mm)
CH3CN
0.5 min





SQD

B: CH3CN





31
Waters:
Waters:
A: 10 mM
From 100% A to 5% A
0.6
3.5



Acquity ®
BEH
NH4HCO3 in
in 2.10 min, to 0% A in 0.9
- - -




UPLC ®-
(1.7 μm,
95% H2O +
min, to 5% A in 0.5 min
5




DAD and
2.1*100 mm)
5% CH3CN






SQD

B: CH3CN





32
Waters:
Waters:
A: 0.1%
From 95% A to
0.3
28



Acquity ®
BEH
NH4HCO3
5% A in 20 min, hold 3
- - -




UPLC ®-
(1.7 μm,
in 95% H2O + 5%
min,
45




DAD and
2.1*150 mm)
CH3CN
to 95% A in 1 min,





SQD

B: CH3CN
hold 4 min




33
Agilent
XBridge
A: 10 mM
From 95% A to 20% A in
1.0
30



Technologies
C18, 3.5
NH4HCO3 in
20 min, hold 2 min, to 95%
- - -




1260
um, 4.6 ×
water pH 9.5
A in 1 min, hold 7 min
45




Series
150 mm
B: CH3CN





34
Waters:
Waters:
A: 10 mM
From 100% A to
0.6
3.5



Acquity ®
BEH
CH3COONH4
5% A in 2.10 min,
- - -




UPLC ®-
(1.7 μm,
in 95% H2O +
to 0% A in 0.90 min,
55




DAD and
2.1*100 mm)
5% CH3CN
to 5% A in 0.5 min





SQD

B: CH3CN





35
Waters:
Waters:
A: 0.1%
From 100% A to
0.8
2.0



Acquity
BEH
NH4HCO3
5% A in 1.3 min,
- - -




UPLC ®-
(1.7 μm,
in 95% H2O +
hold 0.7 min
55




DAD and
2.1*50 mm)
5% CH3CN






SQD

B: CH3CN





36
Waters:
Waters:
A: 10 mM
From 95% A to
0.8
2.0



Acquity ®
BEH
CH3COONH4
5% A in 1.3 min,
- - -




UPLC ® -
(1.7 μm,
in 95% H2O +
held for 0.7 min
55




DAD and
2.1*50 mm)
5% CH3CN






SQD

B: CH3CN





37
SHIMADZ
MERCK,
A:water(4 L) +
a gradient condition from
1.5
1.5



U LC20-
RP-18e 25-
TFA (1.5 mL)
95% A, 5% B to 5% A,
- - -




MS2010
2 mm
B: aceto-
95% B in 0.7 minutes,
50






nitrile (4 L) +
hold at these conditions







TFA
for 0.4 minutes, and then







(0.75 mL)
to 95% A, 5% B in 0.01








min and held for 0.49 min.




38
Agilent
Xtimate C18
A: water(4 L) ) +
a gradient condition from
1.2
2.0



LC1200-
2.1*30 mm, 3
TFA (1.5 mL)
90% A, 10% B to 20% A,
- - -




MS6110
um
B: aceto-
80% B in 0.9 minutes, and
50






nitrile(4 L) + T
hold at these conditions







FA (0.75 mL)
for 0.6 minutes, then to








90% A, 10% B in 0.01








min, and held for 0.49








min.




39
SHIMADZ
Xbridge
A: water(4 L) +
a gradient condition from
0.8
7.0



U LC20-
Shield RP-
NH3•H2O
90% A to 20% A, 80% B
- - -




MS2020
18, 5 um, 2.1*
(0.8 mL)
in 6 minutes, and hold at
50





50 mm
B: aceto-
these conditions for 0.5







nitrile
minutes, then to 90% A








and 10% B in 0.1 min and








held for 0.49 min.




40
SHIMADZ
Xbridge
A: water(4 L) +
a gradient condition from
0.8
7.0



U LC20-
Shield RP-
NH3•H2O
70% A to 10% A, 90% B
- - -




MS2020
18, 5 um, 2.1*
(0.8 mL)
in 6 minutes, and hold at
50





50 mm
B: aceto-
these conditions for 0.5







nitrile
minutes, then to 70% A








and 30% B in 0.1 min and








held for 0.49 min.
















TABLE 1b







LCMS and melting point data. Co. No. means compound


number; Rt means retention time in min.












Co. No.
Rt
[M + 1]+
LCMS method
















 1
2.81
594.5
9



 2
2.81
594.5
9



 2a
2.89
594.6
2



 3
1.11
614.5
12



 4
1.11
614.5
12



 4a
1.12
614.5
12



 4b
1.11
614.5
12



 6
1.67
598.9
13



 7
1.33
648.3
14



 8
3.02
633.5
4



 8a
1.70
633.5
11



 8b
1.00
633.5
12



 9
3.04
633.5
4



 9a
1.00
633.5
12



 9a
0.98
633.5
12



 9b
1.00
633.5
12



 10
2.99
668.6
1



 11
2.65
668.6
9



 12
2.76
659.5
2



 13
2.26
659.6
3



 14
3.02
580.6
2



 15
2.88
580.5
2



 16
3.20
645.5
2



 17
2.79
645.6
2



 18a
1.09
679.5
12



 18b
1.08
679.5
12



 19a
1.09
679.5
12



 19b
1.09
679.5
12



 20
2.87
671.3
2



 21
3.10
671.3
4



 22
2.70
657.6
2



 23
2.23
657.5
6



 24
2.73
631.3
2



 25
2.74
631.3
2



 26a
1.07
677.5
12



 26b
1.07
677.5
12



 27a
1.69
677.5
11



 27b
1.75
677.5
11



 27c
1.69
677.5
11



 27d
1.75
677.5
11



 28
2.60
643.5
2



 28b
2.62
643.6
2



 30
3.00
629.3
5



 31
2.23
629.6
6



 32a
2.83
631.3
2



 32b
3.02
631.2
4



 33a
2.31
666.2
3



 33b
2.31
666.2
3



 33c
2.96
666.2
2



 34a
2.91
667.2
2



 34b
2.92
667.2
2



 35a
2.91
645.3
2



 35b
2.91
645.3
2



 36a
2.99
680.5
7



 36b
2.97
680.5
7



 37a
3.05
681.5
7



 37b
3.05
681.5
7



 38a
2.89
671.6
7



 38b
2.92
671.6
7



 39a
1.08
677.5
12



 39b
1.09
677.5
12



 40a
1.07
677.5
12



 40b
1.08
677.5
12



 41
0.85
568.3
10



 42
2.85
507.4
1



 43
3.05
507.4
4



 44
0.96
465.3
14



 45
1.57
465.7
13



 46
1.23
479.7
13



 47
1.23
479.8
13



 48
3.05
521.2
2



 49
3.03
521.2
2



 49a
1.081
521.4
12



 49b
1.081
521.4
12



 50
1.29
548.8
13



 51
7.28
548.2
8



 58
1.33
562.8
13



 59
1.29
578.9
13



 60
1.31
573.9
13



 61
1.57
493.4
15



 62
2.00
592.5
15



 63
1.50
534.4
16



 64
1.61
570.4
16



 65
1.70
550.4
16



 66
1.01
564.4
12



 67
1.01
559.4
12



 68
1.64
598.4
11



 69
1.63
598.4
11



 70
1.55
562.4
11



 71
1.55
562.4
11



 72
1.05
562.4
12



 73
1.04
562.4
12



 74
1.48
548.5
11



 75
1.64
613.4
15



 76
1.62
625.5
15



 77
1.81
527.4
17



 77a
1.73
527.5
15



 77b
1.73
527.5
15



 77c
1.73
527.5
15



 77d
1.73
527.5
15



 78
1.52
513.4
15



 78a
1.57
513.5
15



 78b
1.56
513.5
15



 78c
1.56
513.5
15



 78d
1.57
513.5
15



 79
1.55
513.4
15



 80
1.41
499.4
15



 81
1.14
534.78
13



 82
1.16
578.84
13



 83
1.48
522.4
16



 84
2.681
548.2
1



 85
3.399
578.2
1



 86
2.771
578.2
1



 87
2.734
564.2
1



 88
1.11
520.82
13



 89
1.21
520.85
13



 90a
1.6
534.6
15



 90b
1.6
534.6
15



 91
1.92
604.5
16



 92
1.69
549.4
16



 93
1.72
555.4
17



 94
1.49
520.5
15



 95
3.774
506.4
1



 96
1.67
542.4
17



 97
1.67
518.4
17



 98
1.73
520.4
17



 99
1.15
534.89
13



100
1.15
534.89
13



101
2.739
563.2
1



102
2.579
534.2
1



103
1.44
508.4
11



104
1.45
520.5
16



105
2.989
520.1
1



106
2.858
652.1
1



107
0.9
560.3
13



108
2.77
652.2
1



109
2.435
521.4
1



110
2.220
520.1
1



111
2.262
564.2
1



112
2.644
548.1
1



113
2.645
548.2
1



114
1.36
506.4
30



116a
1.589
562.4
12



116b
1.503
564.4
12



117a
1.065
592.5
12



117b
1.068
592.5
12



118
1.044
548.4
11



119a
1.01
508.4
12



119b
0.998
508.4
12



120
2.536
506.1
1



121
3.083
576.2
1



122
3.068
562.2
1



123
2.954
578.2
1



124
1.38
556.09
13



125
1.36
593.06
13



126
1.52
602.07
13



127
1.36
546.4
30



128
1.58
568.4
16



128a
1.6
568.5
15



128b
1.6
568.5
15



128c
1.6
568.5
15



128d
1.6
568.5
15



129
7.343
548.2
1



130
1.76
562.6
15



131
1.8
562.6
15



132
1.8
562.6
15



133
1.76
562.6
15



134
1.45
507.6
15



134a
1.44
507.5
15



134b
1.41
507.5
15



134c
1.44
507.5
15



134d
1.44
507.5
15



135
2.986
574.2
1



136
2.813
578.2
1



137a
1.31
548.93
13



137b
1.4
548.83
13



138
1.85
507.4
16



138a
1.87
507.5
15



138b
1.88
507.3
15



139
1.45
520.4
16



139a
1.52
520.5
17



139b
1.52
520.5
17



140
1.535
562.5
11



141
1.4
566.98
13



142
1.589
562.5
11



143
1.562
562.6
11



144
1.45
507.4
16



145
1.59
562.5
17



146
1.66
562.4
30



147a
1.83
507.5
15



147b
1.83
507.5
15



148
3.885
506.4
1



149
2.493
577.2
1



150
1.046
548.4
11



151
1.03
548.4
11



152
2.979
589.5
1



153
1.97
563.4
16



154
1.96
563.4
16



155
1.531
587.4
11



156
1.16
534.87
13



157
3.635
589.5
2



158
1.5
592.4
11



159
1.546
574.4
11



160
1.83
576.5
30



161
1.37
571.09
13



162
1.071
562.4
11



163
1.068
562.3
11



164a
1.089
562.4
12



164b
1.088
562.4
12



165a
1.61
551.5
15



165b
1.62
551.6
15



166a
1.59
513.5
15



166b
1.59
513.5
15



167a
1.45
499.5
15



167b
1.45
499.5
15



168
2.628
563.2
2



169
1.51
534.4
16



169a
1.54
534.5
16



169b
1.49
534.5
16



170
2.853
577.2
2



171
2.826
534.1
3



172
1.48
621.9
13



173
1.51
593.95
13



174
1.42
619.8
13



175
1.003
534.3
11



176
1.005
534.3
11



177
1.01
564.4
11



178
1.52
588.4
30



179
1.65
588.5
16



180a
1.59
527.5
15



180b
1.6
527.5
15



181a
1.57
527.3
16



181b
1.56
527.4
16



182a
1.31
527.5
15



182b
1.31
527.5
15



183a
1.6
527.5
15



183b
1.6
527.5
15



184a
1.63
548.5
15



184b
1.63
548.6
15



184
1.6
548.4
16



185
1.54
562.4
16



186a
3.449
520.1
26



186b
2.803
520.1
27



187
3.634
546.1
3



188a
1.35
548.88
13



188b
1.44
548.67
13



189
1.41
562.5
17



190a
1.73
493.5
16



190b
1.73
493.5
15



191a
1.43
479.5
17



191b
1.42
479.5
17



192
1.47
547.3
16



193
2.2
505.97
13



194
2.22
505.95
13



195
1.3
534.88
13



196
2.977
506.1
6



197
1.056
557.4
12



198a
1.57
537.4
16



198b
1.57
537.4
16



199
2.569
506.2
8



200
2.708
548.1
9



201
2.758
548.2
9



202
1.37
577.97
13



203
1.55
607.9
13



204
1.46
633.9
13



205
1.61
521.5
17



205a
1.62
521.5
17



205b
1.63
521.5
17



206a
1.33
578.5
15



206b
1.32
578.6
15



207
1.3
549.95
13



208
3.334
546.2
9



209
3.370
546.1
9



210
2.685
548.2
9



211
2.734
548.2
9



212
2.588
520.1
9



213a
1.483
555.3
12



213b
1.481
555.3
12



214
1.37
552.4
16



214a
1.42
552.4
16



214b
1.37
552.4
16



215
1.29
534.88
13



216
2.763
548.2
19



217
1.38
563.87
13



218
1.39
563.8
13



219
1.27
549.7
13



220
2.755
546.1
20



221
1.648
562.4
12



222
2.796
548.2
20



223
3.000
546.1
21



224
1.24
552.4
30



224a
1.24
552.4
30



224b
1.23
552.4
30



225
1.55
619.4
31



226
1.63
562.5
16



227
1.7
562.6
30



228
1.65
562.5
16



229
1.67
562.5
16



230a
1.3
568.5
30



230b
1.29
568.5
30



231a
1.3
568.5
30



231b
1.29
568.5
30



232a
1.29
568.5
30



232b
1.29
568.5
30



233a
1.29
568.5
30



233b
1.3
568.5
30



234
3.200
562.2
22



235
1.51
591.9
13



236
1.079
562.4
12



237
1.47
606.85
13



238
2.277
534.4
22



239
2.431
521.4
22



240a
1.73
564.6
17



240b
1.73
564.5
17



241
1.528
548.4
12



242
1.535
548.4
12



244
1.44
592.69
13



245
1.36
590.8
13



246
1.104
576.5
12



247
1.109
576.4
12



248
1.437
564.4
12



249
1.41
590.61
13



250
1.34
578.56
13



251
1.3
576.59
13



252a
2.618
565.5
28



252b
2.632
565.5
28



253a
2.789
551.2
29



253b
2.778
551.1
18



254
2.474
548.4
22



255
2.552
562.4
22



256
2.770
562.5
22



257
2.710
521.4
23



258
2.932
576.5
23



259
2.840
562.5
26



260
3.092
562.4
25



261
0.81
517
15



262
0.74
503
16



263
0.68
519
16



264
0.7
519
15



265
0.79
491
15



266
0.67
519
16



267
0.75
535
16



268
0.67
521
16



269
0.69
521
16



270
0.7
522
15



271
0.66
521
16



272
0.68
535
16



273
0.65
519
16



274
0.65
521
16



275
0.64
521
16



276
0.69
535
16



277
0.66
521
16



278
0.67
521
16



279
0.77
505
16



280
0.85
519
15



281
0.69
523
16



282
0.65
521
16



283
0.81
542
16



284
0.76
501
16



285
0.73
489
16



286
0.79
479
16



287
0.74
479
16



288
0.76
479
16



289
0.78
493
16



290
0.73
506
16



291
0.76
520
16



292a
0.83
534
16



292b
0.77
534
16



293a
0.83
534
16



293b
0.77
534
16



294a
0.72
546
16



294b
0.73
546
16



295
0.73
534
16



296
0.81
560
16



297
0.87
575
15



298
0.83
575
15



299
0.84
549
15



300
0.78
575
15



301
0.82
549
15



302
0.83
549
15



303
0.75
575
15



304
0.73
547
15



305
0.7
533
15



306
0.75
547
15



307
0.71
510
15



308
0.87
491
15



309a
0.85
533
15



309b
0.75
533
15



310
0.81
573
15



311
0.74
559
15



311a
1.42
558.5
17



311b
1.41
558.5
17



312
0.66
575
15



313
0.68
591
15



313a
1.31
590.6
17



313b
1.32
590.6
17



314
0.75
552
15



315
0.75
561
15



316
0.79
575
15



317
0.81
575
15



318
0.74
591
15



319
0.75
561
15



320
0.73
593
15



320a
1.39
592.6
17



320b
1.5
592.6
17



321
0.75
591
15



322
0.67
577
15



323
0.67
577
15



324
0.65
577
15



325
0.66
577
15



326
0.69
579
15



327
0.65
577
15



328
0.74
605
16



329
0.74
607
16



330
0.79
621
16



331
0.79
621
16



332
0.72
605
16



333
0.72
605
16



334
0.75
595
16



335
0.79
609
16



336
0.79
609
16



337
0.75
593
16



338
0.8
607
16



339
0.8
607
16



340
0.83
617
15



341
0.73
602.5
16



342
0.98
619
15



343
0.96
619
15



344
0.82
619
16



345
0.66
619
16



346
0.76
619
16



347
0.8
630
15



348
0.84
646
15



349
0.71
617
15



350
0.76
633
15



351a
6.47
544.4
32



351b
7.78
544.4
32



352a
1.4
560.6
17



352b
1.39
560.6
17



353a
1.34
546.5
17



353b
1.34
546.5
17



354
1.46
574.6
17



357
0.69
537.4
32



358
0.65
535.4
32



359
0.65
535.4
32



360
0.67
535.3
32



361
0.68
535.4
32



362a
1.35
549.6
15



362b
1.3
549.6
15



363a
1.53
549.5
16



363b
1.55
549.5
16



364a
1.35
549.5
17



364b
1.34
549.5
17



365a
1.65
551.5
17



365b
1.65
551.6
17



366
3.53
565.2
2



367
3.53
565.2
2



368
3.52
565.2
2



369
4.94
565.4
39



370
0.707
585.1
37



371
0.703
585.2
37



372
0.693
585.2
37



373
0.701
585.1
37



374
0.658
585.2
37



375
0.658
585.1
37



378
1.69
606.84
14



380
1.74
620.89
14



383
2.11
592.5
15



386
1.464
618.3
14



387
0.708
620.4
37



388
0.708
620.4
37



392
1.490
624.3
14



394
2.29
606.4
15



395
4.560
606.5
39



396
4.635
606.5
39



398
0.689
592.3
37



399
0.693
592.3
37



400
2.10
620.5
15



401
2.00
606.5
15



402
0.758
620.5
37



403
0.757
620.4
37



407
1.83
563.4
15



414
1.97
620.5
14



415
1.98
620.4
14



420
0.701
606.2
37



421
0.699
606.2
37



422
0.664
634.9
37



423
0.660
634.9
37



428
2.754
620.5
1



429
1.37
492.4
16



431
1.37
492.4
15



433
1.12
520.5
14



434
0.334
492.3
14



435
0.409
492.3
14



437
0.609
466.3
37



438
0.364
518.3
14



441
0.566
506.2
37



442
0.321
520.3
37



443
0.427
520.3
37



449
0.351
506.4
40



444
0.609
466.3
37



446
1.35
504.4
15



448
0.98
506.4
35



452
0.595
492.2
37



453
0.591
492.3
37



454
0.396
522.3
38



456
1.41
520.4
15



457
1.35
506.4
15



458
0.321
520.3
37



459
0.427
520.3
37



468
1.69
520.2
14



469
1.68
520.54
14



486
0.665
620.3
37



487
0.743
606.8
37



488
0.577
520.3
37



489
2.24
613
15



490
1.44
513.4
15



491
1.487
592.3
14



492
1.51
592.9
14



497
1.95
563.4
15



498
4.209
549.4
39



499
4.282
549.5
39



500
4.266
622.6
39



503
1.33
423.3
15



504
1.34
423.3
31



505
1.33
423.3
15



507
1.33
423.3
15



508
1.29
423.3
15



509
1.34
423.4
34



510
1.34
423.4
34



516
1.08
535.5
35



517
1.09
523.3
35



519
1.51
547.5
34



520
0.74
551
36



521
1.76
560.5
34



522
1.33
423.3
15



525
0.765
640.3
37



526a
0.681
563.3
37



526b
0.674
563.3
37



527
0.76
452
36



528
1.44
507.4
34



529
1.00
565
36



530
0.61
465
36



532
1.30
565
35



533
0.68
493
36



534
1.51
466.3
16










Analytical SFC
General Procedure for SFC Methods

The SFC measurement was performed using an Analytical Supercritical fluid chromatography (SFC) system composed by a binary pump for delivering carbon dioxide (CO2) and modifier, an autosampler, a column oven, a diode array detector equipped with a high-pressure flow cell standing up to 400 bars. If configured with a Mass Spectrometer (MS) the flow from the column was brought to the (NIS), It is within the knowledge of the skilled person to set the tune parameters (e.g. scanning range, dwell time . . . ) in order to obtain ions allowing the identification of the compound's nominal monoisotopic molecular weight (MW). Data acquisition was performed with appropriate software.









TABLE 1c







Analytical SFC Methods (Flow expressed in mL/min; column temperature (T) in


° C.; Run time in minutes, Backpressure (BPR) in bars or pound-force per square


inch (psi). “ACN” means acetonitrile; “MeOH” means methanol; “EtOH” means


ethanol; “iPrNH2” means isopropylamine. All other abbreviations used in the table


below are as defined before
















Flow-







(ml/mn)
Run time







(min)


Method



Column T



code
Column
Mobile phase
Gradient
(° C.)
BPR (bar)















1
Daicel
A: supercritical
10%-50% B in 6
2.5
9.5



Chiralpak ®
CO2
min, hold 3.5 min





AD3
B: iPrOH + 0.2%

40
130



column (3.0
iPrNH2






μm, 150 × 4.6







mm)






2
Daicel
A: supercritical
10%-50% B in 6
2.5
9.5



Chiralpak ®
CO2
min, hold 3.5 min





IG3
B: EtOH + 0.2%

40
130



column (3.0
iPrNH2






μm, 150 × 4.6







mm)









NMR:
NMR-Methods

Some NMR experiments were carried out using a Bruker Avance III 400 spectrometer at ambient temperature (298.6 K), using internal deuterium lock and equipped with BBO 400 MHz S1 5 mm probe head with z gradients and operating at 400 MHz for the proton and 100 MHz for carbon. Chemical shifts (d) are reported in parts per million (ppm). J values are expressed in Hz.


Some NMR experiments were carried out using a Varian 400-MR spectrometer at ambient temperature (298.6 K), using internal deuterium lock and equipped with Varian 400 4NUC PFG probe head with z gradients and operating at 400 MHz for the proton and 100 MHz for carbon. Chemical shifts (d) are reported in parts per million (ppm). J values are expressed in Hz.


Some NMR experiments were carried out using a Varian 400-VNMRS spectrometer at ambient temperature (298.6 K), using internal deuterium lock and equipped with Varian 400 ASW PFG probe head with z gradients and operating at 400 MHz for the proton and 100 MHz for carbon. Chemical shifts (d) are reported in parts per million (ppm). J values are expressed in Hz.













Compound



number
NMR data







Compound 8

1H NMR (400 MHZ, DMSO-d6) 8.43-8.33 (m, 1H), 7.92 (d, J = 7.2 Hz,




1H), 7.65-7.62 (m, 1H), 7.50-7.42 (m, 2H), 7.27-7.22 (m, 1H), 6.54 (s,



3.0H), 4.43-4.36 (m, 0.54H), 3.52-3.46 (m, 0.55H), 3.37-3.35 (m, 4H),



2.93-2.89 (m, 2H), 2.63-2.58 (m, 6H), 2.36-2.34 (m, 8H), 1.96 (s, 3H),



1.84 (s, 6H), 1.48-1.36 (m, 5H), 0.96-0.83 (m, 9H), 0.46-0.16 (m, 3H).


Compound 8a

1H NMR (400 MHz, METHANOL-d4) = 8.46 (s, 1H), 8.41 (d, J = 11.6




Hz, 1H), 7.98 (d, J = 4.8 Hz, 1H), 7.70-7.64 (m, 1H), 7.55-7.43 (m,



2H), 7.38 (dd, J = 2.4, 8.0 Hz, 1H), 3.84-3.46 (m, 8H), 3.26-3.07 (m,



4H), 2.89-2.71 (m, 4H), 2.69-2.47 (m, 9H), 2.31 (s, 2H), 2.10 (s, 3H),



1.96-1.54 (m, 5H), 1.31 (d, J = 18.0 Hz, 1H), 1.13-0.83 (m, 9H), 0.69-



0.20 (m, 2H)


Compound 9a

1H NMR (400 MHZ, METHANOL-d4) = 8.43-8.34 (m, 1H), 7.93 (d,




J = 8.0 Hz, 1H), 7.66-7.60 (m, 1H), 7.47-7.40 (m, 1H), 7.37-7.31 (m,



2H), 3.54 (d, J = 18.0 Hz, 4H), 3.10-2.91 (m, 3H), 2.81-2.67 (m, 6H),



2.51-2.33 (m, 8H), 2.09 (s, 3H), 2.03 (d, J = 5.2 Hz, 2H), 1.75-1.40 (m,



4H), 1.31 (d, J = 18.0 Hz, 5H), 1.05-0.88 (m, 9H), 0.53 (s, 1H), 0.22 (dd,



J = 6.4, 12.0 Hz, 2H)


Compound 32a

1H NMR (400 MHZ, DMSO-d6) 8.42-8.34 (m, 1H), 7.93-7.92 (m, 1H),




7.65-7.62 (m, 1H), 7.48-7.41 (m, 2H), 7.18-7.15 (m, 1H), 4.42-4.38 (m,



0.54H), 3.50-3.45 (m, 0.55H), 3.37 (s, br, 5H), 3.07-3.05 (m, 2H), 2.89-



2.81 (m, 2H), 2.60-2.56 (m, 5H), 2.33-2.30 (m, 2H), 2.18-2,11 (m, 4H),



1.96 (s, 3H), 1.87 (s, 10H), 1.67-1.60 (m, 2H), 1.52-1.47 (m, 1H), 0.94-



0.81 (m, 7H), 0.48-0.18 (m, 1H).


Compound 43

1H NMR (400 MHZ, DMSO-d6) d 8.43-8.34 (m, 1H), 7.94-7.92 (m,




1H), 7.65-7.62 (m, 1H), 7.50-7.42 (m, 2H), 7.28-7.22 (m, 1H), 4.41-



4.39 (m, 0.5H), 3.80 (dd, J = 7.2 Hz; 3.6 Hz, 2H), 3.51-3.48 (m, 1H),



3.31-3.23 (m, 2.5H), 2.93-2.86 (m, 2H), 2.68-2.66 (m, 1H), 2.61-



2.58 (m, 5H), 2.55-2.51 (m, 0.5H), 2.48-2.46 (m, 0.5H), 2.44-2.37



(m, 1.5H), 2.34 (s, 1.5H), 2.33-2.18 (m, 3H), 1.96-1.95 (m,1H), 1.68-



1.60 (m, 3H), 1.49-1.45 (m, 1H), 1.16-1.06 (m, 2H), 0.96-0.94 (m,



2.5H), 0.48-0.47 (m, 1H), 0.20-0.17 (m, 1.5H).


Compound 51

1H NMR (400 MHZ, DMSO-d6): δ 8.43-8.34 (m, 1H), 7.94-7.92 (m,




1H), 7.67-7.62 (m, 1H), 7.50-7.42 (m, 2H), 7.28-7.22 (m, 1H), 4.42-4.39



(m, 0.43H), 4.34-4.31 (m, 1H), 3.78-3.74 (m, 1H), 3.51-3.46 (m, 0.53H),



3.00-2.84 (m, 3H), 2.71-2.55 (m, 7H), 2.48-2.34 (m, 4H), 2.28-2.14 (m,



3H), 2.00-1.90 (m, 4H), 1.75-1.65 (m, 3H), 1.50-1.44 (m, 1H), 1.06-0.84



(m, 5H), 0.48-0.17 (m, 3H).


Compound 51a

1H NMR (400 MHZ, DMSO-d6, 27° C.) δ ppm 0.18-0.53 (m, 3 H), 0.89-




1.29 (m, 5 H), 1.68-1.89 (m, 3 H), 1.98 (d, J = 1.3 Hz, 4 H), 2.07-2.29



(m, 1 H), 2.37 (s, 1 H), 2.57 (s, 2 H), 2.63 (s, 3 H), 2.94-3.15 (m, 6 H),



3.48-3.54 (m, 0.5 H), 3.56-3.75 (m, 2 H), 3.76-3.86 (m, 1 H), 4.35



(br d, J = 14.3 Hz, 2 H), 4.38-4.42 (m, 0.5 H), 7.32-7.42 (m, 1 H), 7.44-



7.54 (m, 2 H), 7.62-7.71 (m, 1 H), 7.97 (d, J = 5.5 Hz, 1 H), 8.34-8.47



(m, 1 H), 10.01-10.48 (m, 1 H).


Compound 59

1H NMR (400 MHZ, DMSO-d6) δ = 8.56-8.25 (m, 1H), 8.04-7.82 (m,




1H), 7.70-7.58 (m, 1H), 7.56-7.38 (m, 2H), 7.33-7.17 (m, 1H), 4.49-



4.35 (m, 0.5H), 4.35-4.24 (m, 1H), 4.10-3.93 (m, 2H), 3.82-3.66 (m,



1H), 3.55-3.37 (m, 0.5H), 3.27-3.21 (m, 3H), 2.99-2.79 (m, 3H),



2.73-2.55 (m, 7H), 2.43-2.30 (m, 3H), 2.30-2.09 (m, 4H), 2.04-1.84



(m, 1H), 1.81-1.59 (m, 3H), 1.54-1.36 (m, 1H), 1.12-0.81 (m, 5H),



0.63-0.10 (m, 3H).


Compound 60

1H NMR (400 MHZ, DMSO-d6) δ = 8.48-8.27 (m, 1H), 7.98-7.90 (m,




1H), 7.72-7.60 (m, 1H), 7.53-7.40 (m, 2H), 7.31-7.19 (m, 1H), 4.48-



4.33 (m, 0.5H), 4.33-4.23 (m, 1H), 4.08-3.92 (m, 2H), 3.66-3.57 (m,



1H), 3.55-3.42 (m, 0.5H), 3.08-2.79 (m, 3H), 2.74-2.55 (m, 7H),



2.43-2.11 (m, 6H), 2.06-1.82 (m, 1H), 1.80-1.61 (m, 3H), 1.56-1.37



(m, 1H), 1.16-0.82 (m, 5H), 0.62-0.09 (m, 3H).


Compound 117a

1H NMR (400 MHZ, METHANOL-d4) 8.44-8.32 (m, 1H), 7.92 (d, J =




7.6 Hz, 1H), 7.67-7.59 (m, 1H), 7.47-7.39 (m, 1H), 7.38-7.30 (m,



2H), 4.52-4.46 (m, 0.5H), 4.22-4.07 (m, 2H), 3.92-3.84 (m, 1H),



3.63-3.54 (m, 0.5H), 3.39 (s, 3H), 3.11-2.80 (m, 4H), 2.79-2.48 (m,



9H), 2.46-1.85 (m, 6H), 1.84-1.74 (m, 1H), 1.72-1.51 (m, 2H), 1.30-



1.12 (m, 2H), 1.10-0.87 (m, 6H), 0.59-0.18 (m, 3H)


Compound 125

1H NMR (400 MHZ, DMSO-d6) δ ppm −0.04 (br d, J = 2.1 Hz, 1 H), 0.13-




0.22 (m, 2 H), 0.36-0.60 (m, 1 H), 0.85-0.88 (m, 1 H), 0.90-0.99



(m, 3 H), 1.24 (br s, 1 H), 1.29 (s, 6 H), 1.41-1.53 (m, 1 H), 1.64 (br d,



J = 1.7 Hz, 1 H), 1.67-1.75 (m, 2 H), 1.87-2.02 (m, 1 H), 2.13-2.28



(m, 3 H), 2.32-2.42 (m, 3 H), 2.56-2.63 (m, 5 H), 2.64-2.74 (m, 1 H),



2.77-3.01 (m, 3 H), 3.14-3.27 (m, 1 H), 3.38-3.58 (m, 1 H), 4.33-



4.48 (m, 1 H), 5.30 (s, 1 H), 7.23 (d, J = 8.2 Hz, 1 H), 7.28 (s, 1 H), 7.42-



7.51 (m, 2 H), 7.61-7.68 (m, 1 H), 7.93 (d, J = 7.9 Hz, 1 H), 8.30-8.47



(m, 1 H), 8.34 (s, 1 H), 8.43 (d, J = 2.6 Hz, 1 H)


Compound 169a

1H NMR (400 MHZ, DMSO-d6, 27° C.) d ppm 0.12-0.25 (m, 2 H), 0.36-




0.63 (m, 1 H), 0.81-1.12 (m, 5 H), 1.33-1.55 (m, 1 H), 1.59-2.06



(m, 4 H), 2.08-2.27 (m, 4 H), 2.34 (s, 5 H), 2.56-2.65 (m, 5 H), 2.79-



3.16 (m, 4 H), 3.42 (s, 1 H), 4.32-4.48 (m, 1 H), 7.18-7.31 (m, 1 H),



7.32-7.54 (m, 3 H), 7.57-7.72 (m, 1 H), 7.86-7.99 (m, 1 H), 8.27-



8.51 (m, 1 H)


Compound 228

1H NMR (400 MHZ, DMSO-d6, 100° C.) d ppm 0.25-0.89 (m, 6 H),




0.95 (s, 2 H), 1.04 (s, 3 H), 1.39-1.52 (m, 1 H), 1.61-1.72 (m, 2 H),



1.78-1.85 (m, 1 H), 1.87-1.95 (m, 2 H), 1.97 (s, 3 H), 2.09 (dd,



J = 12.1, 4.5 Hz, 1 H), 2.16 (quin, J = 6.2 Hz, 1 H), 2.25 (q, J = 8.4 Hz, 1



H), 2.41 (br s, 1.5 H rotamer), 2.51-2.52 (m, 1 H), 2.60 (s, 1.5



Hrotamer), 2.62-2.65 (m, 3 H), 2.72 (br dd, J = 14.9, 9.3 Hz, 1 H), 3.02-



3.11 (m, 2 H), 3.51-3.62 (m, 0.5 H rotamer), 3.64-4.33 (m, 4 H), 4.35-



4.44 (m, 0.5 H rotamer), 7.25 (s, 1 H), 7.35 (dd, J = 8.5, 3.0 Hz, 1 H),



7.43 (td, J = 8.5, 3.0 Hz, 1 H), 7.58 (dd, J = 8.8, 5.0 Hz, 1 H), 7.94 (br s, 1



H), 8.29-8.44 (m, 1 H)


Compound 365b

1H NMR (400 MHZ, DMSO-d6, 27° C.) δ ppm 0.28 (br d, J = 6.2 Hz, 1 H),




0.39 (br d, J = 6.4 Hz, 1 H), 0.68 (d, J = 6.3 Hz, 1 H), 0.74-0.83 (m, 2 H),



0.83-0.91 (m, 2 H), 0.91-1.00 (m, 2 H), 1.04-1.16 (m, 2 H), 1.42-



1.52 (m, 1 H), 1.56-1.67 (m, 3 H), 1.92-2.02 (m, 1 H), 2.17-2.29 (m,



3 H), 2.32-2.48 (m, 2 H), 2.52-2.65 (m, 5 H), 2.81-2.92 (m, 3 H),



2.96 (br dd, J = 13.6, 5.7 Hz, 1 H), 3.10 (br dd, J = 13.6, 6.2 Hz, 1 H), 3.18-



3.30 (m, 3 H), 3.39-3.62 (m, 2 H), 3.67-3.89 (m, 3 H), 4.57-4.77



(m, 1 H), 7.24-7.29 (m, 1 H), 7.37-7.56 (m, 2 H), 7.56-7.64 (m, 1 H),



7.90-7.94 (m, 1 H), 8.26-8.37 (m, 1 H)









DSC

For a number of compounds, melting points (MP) were determined with a TA Instrument (Discovery DSC 250 or a DSC 2500). Melting points were measured with a temperature gradient of 10° C./minute. Maximum temperature was 300° C. Values are melting peak onset values.


XPRD

Compound 51 as a Crystalline Free Base Form Compound 51 as a crystalline free base Form may be characterized by an X-ray powder diffraction pattern.


X-ray powder diffraction (XRPD) analysis was carried out on a PANalytical Empyrean diffractometer. The compound was loaded onto a zero-background silicon wafer sample holder by gently pressing the powder sample onto the flat surface.


Samples were run on XRPD using the method below:

  • Radiation: Cu K-Alpha (λ=1.5418 Å)
  • Tube voltage/current: 45 kV/40 mA
  • Divergence slit: ⅛°
  • Geometry: Bragg-Brentano
  • Scan mode: Continuous Scan
  • Scan Range: 3-40° 2θ
  • Step size: 0.013° 2θ
  • Scan speed: 20.4 s/step
  • Rotation: On
  • Detector: PIXcel1D


One skilled in the art will recognize that diffraction patterns and peak positions are typically substantially independent of the diffractometer used and whether a specific calibration method is utilized. Typically, the peak positions may differ by about ±0.2° 2θ, or less. The intensities (and relative intensities) of each specific diffraction peak may also vary as a function of various factors, including but not limited to particle size, orientation, sample purity, etc.


The X-ray powder diffraction pattern comprises peaks at 9.3, 12.6, 15.7, 21.9 and 22.5° 2θ 0.2° 2θ. The X-ray powder diffraction pattern may further comprise at least one peak selected from 8.1, 11.6, 13.2, 16.8, 18.5, 18.7, 19.2, 19.9, 20.5° 2θ 0.2° 2θ.


Compound 51 as a crystalline free base Form may further be characterized by an X-ray powder diffraction pattern having four, five, six, seven, eight, nine or more peaks selected from those peaks identified in Table 2a.


Compound 51 as a crystalline free base Form may further be characterized by an X-ray powder diffraction pattern comprising those peaks identified in Table 2a, wherein the relative intensity of the peaks is greater than about 2%, preferably greater than about 5%, more preferably greater than about 10%, more preferably greater than about 15%. However, a skilled person will realize that the relative intensity of the peaks may vary between different samples and different measurements on the same sample.


Compound 51 as a crystalline free base Form may further be characterized by an X-ray powder diffraction pattern substantially as depicted in FIG. 1.


Table 2a provides peak listing and relative intensity for the XPRD of Compound 51 as a crystalline free base Form:





















Pos.
Rel. Int.

Pos.
Rel. Int.



No.
(° 2θ)
(%)
No.
(° 2θ)
(%)























1
7.411
4.6
16
18.744
63.6



2
8.056
31.4
17
19.15
48.8



3
9.304
100
18
19.57
16.4



4
9.893
18.2
19
19.912
32.2



5
11.574
28.1
20
20.503
38.2



6
11.969
6.4
21
21.881
65.4



7
12.598
55.9
22
22.485
57.1



8
13.163
37.6
23
23.693
13



9
14.804
7.9
24
24.205
5.6



10
15.723
89.6
25
24.915
15.2



11
16.195
24.4
26
25.401
19.6



12
16.762
34.5
27
26.068
5.3



13
17.076
21.2
28
26.400
14.5



14
17.694
9.5
29
28.276
8



15
18.454
33.2
30
28.499
11










Compound 51a Crystalline HCl Salt Form (Mono HCl Trihydrate Salt)

Compound 51a (Crystalline HCl salt Form —mono HCl trihydrate salt) may be characterized by an X-ray powder diffraction pattern.


X-ray powder diffraction (XRPD) analysis was carried out on a PANalytical Empyrean diffractometer. The compound was loaded onto a zero-background silicon wafer sample holder by gently pressing the powder sample onto the flat surface.


Samples were run on XRPD using the method below:

  • Radiation: Cu K-Alpha (λ=1.5418 Å)
  • Tube voltage/current: 45 kV/40 mA
  • Divergence slit: ⅛°
  • Geometry: Bragg-Brentano
  • Scan mode: Continuous Scan
  • Scan Range: 3-40° 2θ
  • Step size: 0.013° 2θ
  • Scan speed: 20.4 s/step
  • Rotation: On
  • Detector: PIXcel1D


One skilled in the art will recognize that diffraction patterns and peak positions are typically substantially independent of the diffractometer used and whether a specific calibration method is utilized. Typically, the peak positions may differ by about ±0.2° 2θ, or less. The intensities (and relative intensities) of each specific diffraction peak may also vary as a function of various factors, including but not limited to particle size, orientation, sample purity, etc.


The X-ray powder diffraction pattern comprises peaks at 5.2, 13.2, 14.1, 18.8 and 20.3° 2θ 0.2° 2θ. The X-ray powder diffraction pattern may further comprise at least one peak selected from 9.7, 10.0, 15.4, 15.8, 18.3, 21.3, 24.3° 2θ±0.2° 2θ.


Compound 51a may further be characterized by an X-ray powder diffraction pattern having four, five, six, seven, eight, nine or more peaks selected from those peaks identified in Table 2b.


Compound 51a may further be characterized by an X-ray powder diffraction pattern comprising those peaks identified in Table 2b, wherein the relative intensity of the peaks is greater than about 2%, preferably greater than about 5%, more preferably greater than about 10%, more preferably greater than about 15%. However, a skilled person will realize that the relative intensity of the peaks may vary between different samples and different measurements on the same sample.


Compound 51a may further be characterized by an X-ray powder diffraction pattern substantially as depicted in FIG. 2.


Table 2b provides peak listing and relative intensity for the XPRD of Compound 51a.





















Pos.
Rel. Int.

Pos.
Rel. Int.



No.
(° 2θ)
(%)
No.
(° 2θ)
(%)























1
5.151
36
13
19.505
16.6



2
9.749
37.2
14
20.305
100



3
9.984
58.9
15
21.331
16.6



4
13.217
34
16
21.855
6.8



5
14.095
64.4
17
22.905
4.5



6
15.393
20.4
18
23.419
8.2



7
15.842
16.4
19
24.310
17.4



8
16.315
10.4
20
25.136
10.6



9
17.471
10.1
21
25.595
7.1



10
18.296
19.4
22
26.529
12.9



11
18.810
34.3
23
29.496
4.5



12
19.19
10.8
24
30.179
6.1










Dynamic Vapor Sorption (DVS)

The moisture sorption analysis (DVS) was performed using a ProUmid GmbH & Co. KG Vsorp Enhanced dynamic vapor sorption apparatus. Results are shown in FIG. 3 and FIG. 4. The moisture profile was evaluated by monitoring vapor adsorption/desorption over the range of 0 to 90% relative humidity at 25° C. The sample weight equilibrium criteria were set at <0.01% change in 45 min with minimum and maximum time of acclimation at 50 min and 120 min, respectively. The moisture profile consisted of 2 cycles of vapor adsorption/desorption.


The DVS change in mass plot of crystalline HCl salt Form (Compound 51a) shows that the crystalline form is hygroscopic with the water content varying with relative humidity and dehydrates rapidly at below 10% RH (relative humidity) to complete dehydrated state at 0% RH. In the humidity range of 20-90% RH, the crystalline form adsorbs and desorbs moisture slowly and reversibly up to 2.5% by mass on average. Based on DVS, the crystalline HCl salt Form, at equilibrium, can contain around 3 equivalents of water (8.5-9.5% total moisture mass) at common ambient RH of 40% to 75%. The XRPD pattern of the fraction obtained after the DVS test was comparable to the starting material. No indication of a solid-state form change was observed.


Pharmacological Part
1) Menin/MLL Homogenous Time-Resolved Fluorescence (HTRF) Assay

To an untreated, white 384-well microtiter plate was added 40 nL 200× test compound in DMSO and 4 μL 2× terbium chelate-labeled menin (vide infra for preparation) in assay buffer (40 mM Tris HCl, pH 7.5, 50 mM NaCl. 1 mM DTT (dithiothreitol) and 0.05% Pluronic F-127). After incubation of test compound and terbium chelate-labeled menin for 30 min at ambient temperature, 4 μL 2×FITC-MBM1 peptide (FITC-β-alanine-SARWRFPARPGT-NH2) (“FITC” means fluorescein isothiocyanate) in assay buffer was added, the microtiter plate centrifuged at 1000 rpm for 1 min and the assay mixtures incubated for 15 min at ambient temperature. The relative amount of menin FITC-MBM1 complex present in an assay mixture is determined by measuring the homogenous time-resolved fluorescence (HTRF) of the terbium/FITC donor/acceptor fluorphore pair using an EnVision microplate reader (ex. 337 nm/terbium em. 490 nm/FITC em. 520 nm) at ambient temperature. The degree of fluorescence resonance energy transfer (the HTRF value) is expressed as the ratio of the fluorescence emission intensities of the FITC and terbium fluorophores (Fem520 nm/Fem490 nm). The final concentrations of reagents in the binding assay are 200 μM terbium chelate-labeled menin, 75 nM FITC-MBM1 peptide and 0.5% DMSO in assay buffer. Dose-response titrations of test compounds are conducted using an 11 point, four-fold serial dilution scheme, starting typically at 10 μM.


Compound potencies were determined by first calculating % inhibition at each compound concentration according to equation 1:





% inhibition=((HC−LC)−(HTRFcompound−LC))/(HC−LC))*100  (Eqn 1)


Where LC and HC are the HTRF values of the assay in the presence or absence of a saturating concentration of a compound that competes with FITC-MBM1 for binding to menin, and HTRFcompound is the measured HTRF value in the presence of the test compound. HC and LC HTRF values represent an average of at least 10 replicates per plate. For each test compound, % inhibition values were plotted vs. the logarithm of the test compound concentration, and the IC50 value derived from fitting these data to equation 2:





% inhibition=Bottom+(Top−Bottom)/(1+10{circumflex over ( )}((log/C50−log[cmpd])*h))  (Eqn 2)


Where Bottom and Top are the lower and upper asymptotes of the dose-response curve, respectively, IC50 is the concentration of compound that yields 50% inhibition of signal and h is the Hill coefficient.


Preparation of Terbium cryptate labeling of Menin: Menin (a.a 1-610-6xhis tag, 2.3 mg/mL in 20 mM Hepes (2-[4-(2-Hydroxyethyl)-1-piperazinyl]ethane sulfonic acid), 80 mM NaCl, 5 mM DTT (Dithiothreitol), pH 7.5) was labeled with terbium cryptate as follows. 200 μg of Menin was buffer exchanged into 1×Hepes buffer. 6.67 μM Menin was incubated with 8-fold molar excess NHS (N-hydroxysuccinimide)-terbium cryptate for 40 minutes at room temperature. Half of the labeled protein was purified away from free label by running the reaction over a NAP5 column with elution buffer (0.1M Hepes, pH 7+0.1% BSA (bovine serum albumin)). The other half was eluted with 0.1 M phosphate buffered saline (PBS), pH7. 400 μl of eluent was collected for each, aliquoted and frozen at −80° C. The final concentration of terbium-labeled Menin protein was 115 μg/mL in Hepes buffer and 85 μg/mL in PBS buffer, respectively.









MENIN Protein Sequence (SEQ ID NO: 1):


MGLKAAQKTLFPLRSIDDVVRLFAAELGREEPDLVLLSLVLGFVEHFLAV





NRVIPTNVPELTFQPSPAPDPPGGLTYFPVADLSIIAALYARFTAQIRGA





VDLSLYPREGGVSSRELVKKVSDVIWNSLSRSYFKDRAHIQSLFSFITGT





KLDSSGVAFAVVGACQALGLRDVHLALSEDHAWVVFGPNGEQTAEVTWHG





KGNEDRRGQTVNAGVAERSWLYLKGSYMRCDRKMEVAFMVCAINPSIDLH





TDSLELLQLQQKLLWLLYDLGHLERYPMALGNLADLEELEPTPGRPDPLT





LYHKGIASAKTYYRDEHIYPYMYLAGYHCRNRNVREALQAWADTATVIQD





YNYCREDEEIYKEFFEVANDVIPNLLKEAASLLEAGEERPGEQSQGTQSQ





GSALQDPECFAHLLRFYDGICKWEEGSPTPVLHVGWATFLVQSLGRFEGQ





VRQKVRIVSREAEAAEAEEPWGEEAREGRRRGPRRESKPEEPPPPKKPAL





DKGLGTGQGAVSGPPRKPPGTVAGTARGPEGGSTAQVPAPAASPPPEGPV





LTFQSEKMKGMKELLVATKINSSAIKLQLTAQSQVQMKKQKVSTPSDYTL





SFLKRQRKGLHHHHHH






2a) Proliferation Assay

The anti-proliferative effect of menin/MLL protein/protein interaction inhibitor test compounds was assessed in human leukemia cell lines. The cell line MOLM14 harbors a MLL translocation and expresses the MLL fusion protein MLL-AF9, respectively, as well as the wildtype protein from the second allele. OCI-AML3 cells that carry the NPM1c gene mutation were also tested. MLL rearranged cell lines (e.g. MOLM14) and NPM1c mutated cell lines exhibit stem cell-like HOXA/MEIS1 gene expression signatures. KO-52 was used as a control cell line containing two MLL (KMT2A) wildtype alleles in order to exclude compounds that display general cytotoxic effects.


MOLM14 cells were cultured in RPMI-1640 (Sigma Aldrich) supplemented with 10% heat-inactivated fetal bovine serum (HyClone), 2 mM L-glutamine (Sigma Aldrich) and 50 μg/ml gentamycin (Gibco). KO-52 and OCI-AML3 cell lines were propagated in alpha-MEM (Sigma Aldrich) supplemented with 20% heat-inactivated fetal bovine serum (HyClone), 2 mM L-glutamine (Sigma Aldrich) and 50 μg/ml gentamycin (Gibco). Cells were kept at 0.3-2.5 million cells per ml during culturing and passage numbers did not exceed 20.


In order to assess the anti-proliferative effects, 200 MOLM14 cells, 200 OCI-AML3 cells or 300 KO-52 cells were seeded in 200 μl media per well in 96-well round bottom, ultra-low attachment plates (Costar, catalogue number 7007). Cell seeding numbers were chosen based on growth curves to ensure linear growth throughout the experiment. Test compounds were added at different concentrations and the DMSO content was normalized to 0.3%. Cells were incubated for 8 days at 37° C. and 5% CO2. Spheroid like growth was measured in real-time by live-cell imaging (IncuCyteZOOM, Essenbio, 4×objective) acquiring images at day 8. Confluence (%) as a measure of spheroid size was determined using an integrated analysis tool.


In order to determine the effect of the test compounds over time, the confluence in each well as a measure of spheroid size, was calculated. Confluence of the highest dose of a reference compound was used as baseline for the LC (Low control) and the confluence of DMSO treated cells was used as 0% cytotoxicity (High Control, HC).


Absolute IC50 values were calculated as percent change in confluence as follows:


LC=Low Control: cells treated with e.g. 1 μM of the cytotoxic agent staurosporin, or e.g. cells treated with a high concentration of an alternative reference compound


HC=High Control: Mean confluence (%) (DMSO treated cells)





% Effect=100−(100*(Sample−LC)/(HC−LC))


GraphPad Prism (version 7.00) was used to calculate the IC50. Dose-response equation was used for the plot of % Effect vs Log10 compound concentration with a variable slope and fixing the maximum to 100% and the minimum to 0%.


2b) MEIS1 mRNA Expression Assay


MEIS1 mRNA expression upon treatment of compound was examined by Quantigene Singleplex assay (Thermo Fisher Scientific). This technology allows for direct quantification of mRNA targets using probes hybridizing to defined target sequences of interest and the signal is detected using a Multimode plate reader Envision (PerkinElmer). The MOLM14 cell line was used for this experiment. Cells were plated in 96-well plates at 3,750 cells/well in the presence of increasing concentrations of compounds. After incubation of 48 hours with compounds, cells were lysed in lysis buffer and incubated for 45 minutes at 55° C. Cell lysates were mixed with human MEIS specific capture probe or human RPL28 (Ribosomal Protein L28) specific probe as a normalization control, as well as blocking probes. Cell lysates were then transferred to the custom assay hybridization plate (Thermo Fisher Scientific) and incubated for 18 to 22 hours at 55° C. Subsequently, plates were washed to remove unbound materials followed by sequential addition of preamplifiers, amplifiers, and label probe. Signals (═gene counts) were measured with a Multimode plate reader Envision. IC50S were calculated by dose-response modelling using appropriate software. For all non-housekeeper genes response equal counts corrected for background and relative expression. For each sample, each test gene signal (background subtracted) was divided by the normalization gene signal (RPL28: background subtracted). Fold changes were calculated by dividing the normalized values for the treated samples by the normalized values for the DMSO treated sample. Fold changes of each target gene were used for the calculation of IC50S.









TABLE 3







Biological data - HTRF assay, proliferation assay, and MEIS1


mRNA expressionassay
















spheroid




HTRF-30 min
MEIS1
spheroid assay_
assay_OCI-
spheroid assay_KO-


Compound
incubation IC50
IC50
MOLM14 IC50
AML3
52 IC50


Number
(μM)
(μM)
(μM)
IC50 (μM)
(μM)















 1
0.000033
0.004
0.002

7.5


 2
0.000049
0.003
0.001

>15


 2a
0.000024
0.036





 3
0.000026
0.010
0.004

1.1


 4
0.000016
0.011
0.005

3.0


 4a
0.000301
0.247
0.132

7.8


 4b
0.000094
0.006
0.003

0.5


 6
0.000288
0.049
0.034

1.5


 7
0.000305
0.158





 8
0.000365
0.023
0.011

2.4


 8a
0.000024
0.013





 8b
~0.0073
0.044





 9
0.000138
0.023
0.013

5.6


 9a
0.000016
0.006
0.002

0.6


 9b
0.000975
0.042





 10
0.000021
0.017





 11
0.000034
0.006





 12
0.000018
0.005





 13
0.000124
0.006





 14
0.000054
<0.0036





 15
0.000040
<0.0008





 16
0.000051
0.070





 17
0.000031
0.048





 18a
0.000019
0.005





 18b
0.000017
0.110





 19a
0.000032
<0.0033
0.003

>15


 19b
0.000065
0.128





 20
~0.00019
<0.0017





 21
0.000052
<0.0023
<0.0018

2.0


 22
0.000024
~0.011





 23
0.000070
0.007





 24
0.000057
0.071
0.013

8.5


 25
0.000087
0.091





 26a
0.000036
0.007





 26b
~0.000036
0.029





 27a
0.000012
<0.0041





 27b
0.000038
0.019





 27c
0.000038
0.040





 27d
0.000013
0.080





 28
0.000055
0.107





 28b
0.000042
0.162





 30
~0.00049
>1





 31
0.000052
~0.47





 32a
0.000062
0.022
0.005

5.5


 32b
0.000472
>1





 33a
0.000070
0.011
0.010

3.0


 33b
0.000030
0.023





 33c
0.000072
~0.42





 34a
0.000019
0.016





 34b
0.000092
0.025





 35a
0.000059
0.021


1.5


 35b
0.000077
0.047





 36a
0.000030
0.016


0.6


 36b
0.000052
0.019
0.003

0.5


 37a
0.000126
0.027
0.008

0.8


 37b
0.000237
0.028





 38a
0.000036
0.006
<0.0018

0.8


 38b
0.000013

0.002

1.2


 39a
0.000034
0.005
0.002

>15


 39b
0.000052
0.035





 40a
0.000841
~0.54





 40b
0.000182
~0.68





 41
0.000202
0.015
0.014

1.4


 42
0.000030
0.045
0.031

2.8


 43
0.000027
0.024
0.014
0.023
3.3


 44
0.000060
0.043
0.071

4.7


 45
0.000054
0.056
0.053

6.2


 46
0.000047
0.092





 47
0.000042
0.055





 48
0.000060
0.020





 49
0.000048
0.017
0.003

3.0


 49a
0.000227
0.221
0.113




 49b
0.000060
0.009
0.003
0.007
4.1


 50
0.000079
0.089
0.039

2.3


 51
0.000042
0.011
0.008
0.024
1.9


 51a
0.000024
0.011
0.013
0.070
1.2


 58
0.000135
0.183
0.176

10.4


 59
0.000054
0.009
0.011
0.012
2.1


 60
<0.0000095
0.007
0.013
0.013
2.7


 61
0.000132
0.023
0.010

7.9


 62
0.000085
0.014





 63
0.000176
0.083
0.053
0.121
5.9


 64
0.000284
~0.039





 65
0.000079
0.045





 66
0.000037
0.046
0.018

7.7


 67
~0.000043
~0.068
0.048

12.0


 68
0.000043
0.032





 69
0.000049
0.017
0.005
0.021
11.0


 70
0.000114
0.048
0.040
0.068
8.5


 71
0.000202
0.048
0.083

>15


 72
0.000173
0.182





 73
0.000057
0.101





 74
0.000034
0.012
0.004

5.2


 75
0.000059
0.119
0.009

2.0


 76
0.000304






 77
0.000191
0.136
0.090

5.8


 77a
0.000105
0.206
0.117




 77b
0.000070
0.132
0.081
0.128
1.9


 77c
0.000546
0.669





 77d
0.000208
~0.26
0.197

>15


 78
0.000299
0.327
0.111

8.4


 78a
0.001217
~0.69





 78b
0.000552
0.361





 78c
0.000382
0.416
0.200




 78d
0.000875
0.538





 79
0.000168

0.092

9.4


 80
0.000116

0.099

7.2


 81
0.000425
0.077
0.058
0.100
4.0


 82
~0.000035
0.011





 83
0.001300
2.220





 84
0.000075
0.131
0.063
0.120
4.3


 85
0.000113
0.022
0.012
0.022
2.2


 86
0.000042
0.021
0.010
0.023
1.3


 87
0.000042
0.027





 88
0.000093
0.140
0.096




 89
0.000183
~0.895





 90a
0.000391
~0.127





 90b
0.000263
0.103





 91
0.000155
0.094





 92
0.000219
0.052





 93
0.000418
~0.360





 94
0.000079
0.309





 95
0.000601
~0.914





 96
0.004571
3.691





 97
0.001971
1.607





 98
0.000747
~0.535





 99
0.000217
~0.792





100
0.000165
0.434





101
0.000041
0.007
0.041
0.008
3.1


102
0.003025
>1





103
0.003272
~5.12





104
0.000147
0.254
0.275

>15


105
0.000728
>1





106
0.000039
0.104





107
0.000188
0.226





108
0.000161
0.153





109
0.000304
0.053





110
0.001802
~1.07





111
0.000745
~0.707





112
0.000555
0.345





113
0.000642
~0.493





114
0.000173
0.487





115
0.000021
0.011





116a
0.000075
0.005
0.008
0.009
3.8


116b
0.000329
0.377





117a
0.000088
0.007
0.005
0.007
1.6


117b
0.000270
0.411
0.300




118
0.000139
0.117





119a
0.000906
>1





119b
0.000569
>1





120
0.000054
0.149
0.539




121
0.000049
0.013





122
0.000050
0.018
0.012




123
0.000052
0.031
0.016




124
0.000156
0.114





125
0.000027
0.016
0.011
0.012
7.0


126
0.000091
0.032
0.018




128
0.000078
0.238
0.146

7.9


128a
0.000151
0.258
0.676

7.7


128b
0.000045
0.065
0.040




128c
0.001006
~0.4869





128d
0.000104
0.419
0.089




129
0.000291
0.277





130
0.001202
0.314





131
0.000132
0.129





132
0.001211
~0.5733





133
0.000051
0.042





134a
0.000032
0.086





134b
0.000988
>1





134c
0.000073
0.119





134d
0.000425
>1





135
~0.000056
0.007





136
~0.000094
0.071
0.028
0.054
1.4


137a
0.000063
0.014
0.015
0.023
1.1


137b
~0.00034
0.043





138
0.000067
0.047





138a
0.000054
0.044
0.032
0.072
1.7


138b
0.000109
~0.258
0.086
0.222
3.7


139
0.000119
0.444





139a
0.000122
~0.81





139b
0.000087
0.716





140
~0.00017
0.049
0.015




141
0.000027
0.015





142
0.000086
0.010





143
0.000064
0.005





144
0.000072
0.011
0.011

6.5


145
0.001056
~0.7485





146
0.001220
>1





147a
0.000082
0.052
0.010




147b
0.000073
~0.1048





148
0.000086
0.238
0.088




149
0.000037
0.023





150
0.000430
>1
1.474




151
0.003773
1.351





152
0.000053
0.197





153
0.000077
0.077
0.035

7.9


154
0.000140
0.244
0.091




155
0.000051
0.054
0.049




156
0.000042
0.187
0.042




157
0.000020
0.006
0.002




158
0.000045
0.024
0.006




159
0.000019
0.010
0.003




160
0.004021
>1





161
0.000167
0.256
0.053




162
0.000505
>1
0.424




163
0.000192
~0.623
0.456




164a
0.000209
0.249
0.154




164b
0.000025
0.017
0.010
0.012
10.4


165a
0.000173
~0.18





165b
0.000070
0.038
0.004

1.9


166a
0.000084
0.038
0.049




166b
0.000226
0.256
0.120




167a
0.000190
>1
0.210




167b
0.000118
0.117
0.099




168
0.000100
0.016
0.008




169
0.000061
0.071
0.016

10.0


169a
0.000061
0.015
0.017




169b
0.000195
~0.22
0.087




170
0.000155
0.079
0.014




171
0.000581
>1
0.337




172
0.000134
0.018
0.011




173
0.000056
0.032
0.005




174
0.000048
0.034
0.007




175
0.001332
~1.05
0.234




176
0.000200
0.306
0.046




177
0.000109
0.268
0.035




178
0.000055
0.014
0.012

2.9


179
0.000405
0.259





180a
0.000143
0.117
0.039

7.2


180b
0.007291
~2.806





181a
0.000066
0.111
0.044

4.1


181b
0.004582
1.914





182a
0.000462
0.287





182b
0.003095
1.767





183a
0.000370
0.307





183b
0.002614
~2.04





184
0.000060

0.026

7.4


184a
0.000041
0.023
0.013
0.026
>15


184b
0.000065
0.115
0.052
0.138
>15


185
0.000097
0.044
0.023

11.0


186a
0.000447
0.419





186b
0.000932
0.504





187
0.001016
1.937





189
0.000544
0.314
0.255

14.4


190a
0.000101
0.113
0.040

10.0


190b
0.000063
0.065
0.046

4.9


191a
0.000056
0.039
0.007

4.6


191b
0.000071
0.041
0.033

7.0


192
~0.000268
~0.403





193
0.000071
0.041





194
0.000072
0.066





195
0.000153
0.156





196
0.000190
0.438





197
0.000046
0.008





198a
0.000034
0.013
0.010
0.015



198b
0.000085
0.107

0.156
>15


199
0.000327
0.338
0.126

11.8


200
0.000909
~0.864
0.679

>15


201
0.000063
0.047
0.031
0.035



202
0.000081
0.025
0.007

3.4


203
0.000069
0.043
0.012

2.9


204
0.000091
0.079
0.016

1.6


205
0.000106
0.019
0.019

2.5


205a
0.000049
0.021
0.023

7.0


205b
0.000035
0.031
0.025

3.2


206a
0.000045
0.024
0.013

6.6


206b
0.000387
~0.524
0.148

>15


207
0.000016
0.029
0.016

14.3


208
0.000147
0.281
0.224

>15


209
0.000202
>1
0.458

12.8


210
0.000602
>1





211
0.000032
0.113
0.169

>15


212
0.000115
0.322
0.305

>15


213a
0.000567
1.149





213b
0.001629
~2.30





214

>1





214a
0.001293
~0.605





214b
0.011692
~7.28





215
0.000291
0.299





216
0.000187
0.100





217
0.000039
0.021





218
0.000094
0.048





219
0.000039
0.032





220
0.000932
>1





221
0.000311
0.109





222
0.000103
0.259





223
0.000239
0.279





224
0.008656
~3.88





224a
0.030697
>1





224b
0.002766
>1





225
0.000637
0.340





226
0.000255
0.269





227
0.000655
0.551





228
0.000061
0.008
0.010
0.011
5.8


229
0.000235
0.093





230a
0.000053
0.077





230b
0.005446
>1





231a
0.000311
0.052





231b
0.006942
>1





232a
0.000624
~0.448





232b
0.004044
>1





233a
0.000171
0.247





233b
0.002238
>1





234
~0.000068
0.018





235
0.000097
0.014





236
0.000086
~0.0989





237
0.000149
0.052





238
0.001935
>1





239
0.000305
0.138





240a
0.000407
0.291





240b
0.000151
0.038





241
0.000084
0.173





242
0.000811
>1





243
0.000097
0.037





244
0.000101
0.051





245
0.000185
0.065





246
0.000057
0.016





247
0.000179
0.116





248
0.000255
0.011





249
0.000023
0.007





250
0.000076
0.030





251
0.000066
0.083





252a
0.000341
0.069





252b
0.000250
0.315





253a
0.001474
~0.540





253b
0.001726
>1





254
0.000071
0.019





255
0.000102
0.010





256
0.000073
0.020





257
0.000031
0.007





258
0.000021
0.011





259
0.000031
0.138





260
0.000042
~0.0276





261
0.000213
0.217





262
0.000018
0.015
0.015

2.9


263
0.000258
0.175





264
0.000034
0.025
0.020

13.5


265
0.000097
0.046





266
0.000349
0.431





267
0.00024
0.166





268
0.000149
0.037
0.088

>15


269
0.001699
0.018





270
0.000311
0.177





271
0.000368
0.288





272
0.000310
~0.27





273
0.001080
>1





274
0.002422
>1





275
0.000945
>1





276
0.000532
~0.462





277
0.000481
~0.502





278
0.000323
0.342





279
0.000025
0.017
0.012

5.1


280
0.000077
0.051





281
0.000058
0.013
0.010

4.2


282
0.003272
>1





283
0.000610
0.440





284
0.004185
~2.36





285
0.001485
~1.33





286
0.000046
0.059





287
0.000034
0.026





288
0.000059
0.043





289
0.000055
0.031





290
0.000393
0.170





291
0.000594
0.468





292a
0.000132






292b
0.000562
~0.586





293a
0.000459
0.148





293b
0.000152
0.259





294a
0.000237
0.365





294b
0.000077
0.153





295
0.000171
0.338





296
0.000339
0.549





297
0.000477
>1





298
0.000083
0.178





299
0.000151
~0.281





300
0.000116
0.055





301
0.000191
0.144





302
0.000050
0.078





303
0.000179
~0.077





304
0.002803
>1





305
0.000146
~0.297





306
0.000468
0.159





307
0.000044
0.048





308
0.060201
>1





309a
0.001300






309b
0.000111
>1





310
0.000119
0.351





311
0.000031
0.018
0.017

5.2


311a
0.000037
0.014
0.018

7.7


311b
0.000077
0.026
0.035

>15


312
0.000058
0.055
0.033

>15


313
0.000155
0.552
0.179




313a
0.000111
~0.299





313b
0.000713
~0.713





314
0.000055
0.019
0.006




315
0.000071
0.037
0.010

2.4


316
0.000055
0.029
0.012

8.9


317
0.000291
0.065





318
0.000220
1.377





319
0.000293
0.240
0.139

>15


320
0.000078
0.023
0.019

6.9


320a
0.000189
0.024





320b
0.000104
0.062





321
0.000257
0.413
0.232




322
0.000152
~0.547
0.323




323
0.000080
~0.146
0.142




324
0.000267
0.642
0.553




325
0.000337
1.336
1.049




326
0.000038
0.023
0.016




327
0.000819
~0.616





328
0.000101
~0.154
0.050
0.052



329
0.000152
0.013





330
0.000130
0.010





331
0.000117
0.029





332
0.000130
0.196





333
0.000251
~0.394





334
0.000224
0.012





335
0.000193
0.018





336
0.000128
0.033





337
0.000092
0.024





338
0.000185
0.037





339
0.000193
0.068





340
0.000034
>1





341
0.000037
0.020





342
0.000188
0.107





343
0.000045
0.013





344
~0.000058
0.014





345
0.000204
0.036





346
0.000116
0.011





347
0.000045
0.144





348
0.000059
0.033





349
0.006685
>1





350
0.000583
~1.00





351a
0.000040
0.012





351b
0.000255
0.182





352a
0.000102
0.013
0.007
0.011
>15


352b
0.000247
0.266
0.140
0.263
>15


353a
0.000094
0.012
0.006
0.011
>15


353b
0.000238
0.139

0.236



354
0.000040
0.018





357
0.000035
0.015
0.008

3.0


358
0.000223
0.386





359
0.000166
0.358
0.079




360
0.000127
0.107





361
0.000149






362a
0.000149
0.184





362b
0.000067
0.019
0.012




363a
0.000228
0.359





363b
0.000093
~0.1745





364a
0.000098
0.225





364b
0.000292
~0.6699





365a
0.000102
0.035





365b
0.000081
0.011
0.008
0.011



366
0.000067
~0.3194





367
0.000201
>1





503
0.010457






504
0.023939
>1





505
0.003110






507
0.006605






508
0.003680






509
0.004525






510
0.002946






522
0.010590






523a
0.000376
>1





523b
0.000124
0.062





524b
0.008624
2.120





524a
0.019002
2.557





528
0.000514






533
0.000808






534
0.000556








Claims
  • 1. A compound of Formula (I)
  • 2. The compound according to claim 1, wherein Q represents —CHRy—, —O—, —C(═O)—, —NRq—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;R1a represents hydrogen; cyano; halo; Het; —C(═O)—NRxaRxb; —S(═O)2—R18;
  • 3. The compound according to claim 1, wherein Q represents —CHRy— or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;R1a represents hydrogen; halo; —C(═O)—NRaRxb; —S(═O)2—R18,—C(═O)—O—C1-4alkyl; or
  • 4. The compound according to claim 1, wherein Q represents —CHRy—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;R1a represents hydrogen; halo; —C(═O)—NRxaRxb; or
  • 5. The compound according to claim 4, wherein Q represents —CHRy—, or —CRy═; the dotted line is an optional additional bond to form a double bond in case Q represents —CRy═;R1a represents hydrogen; halo; or —C(═O)—NRxaRxb;Rxa and Rxb are each independently selected from the group consisting of hydrogen and C1-6alkyl;R1b represents F;R2 represents halo, C1-4alkyl, or C1-4alkyl substituted with one, two or three halo substituents;R21 represents hydrogen;Ry represents hydrogen;R5 represents hydrogen;R3 and R4 are each independently selected from the group consisting of Het1; Cy2;C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, —NR8aR8b, Het1, and Cy2;Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of—(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;or Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 6- to 11-membered bicyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three substituents selected from the group consisting of —(C═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;R8a and R8b are each independently selected from the group consisting of C1-6alkyl; and C1-6alkyl substituted with one —O—C1-4alkyl;Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8;R6 represents Het4; —C(═O)—NH—R8; or —S(═O)2—C1-4alkyl;R8 represents —O—C1-6alkyl, C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —O—C1-4alkyl, and cyano;Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b;Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one or two carbon atoms with in total one or two —S(═O)2—C1-4alkyl; and wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of—C(═O)—C1-4alkyl and —S(═O)2—C1-4alkyl;Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with—C(═O)—C1-4alkyl;Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, Het6a, Het6b, and —NR9aR9b;R9a and R9b are each independently selected from the group consisting of hydrogen;C1-4alkyl; —C(═O)—C1-4alkyl; and —S(═O)2—C1-4alkyl;R10a and R10b are each independently selected from the group consisting of hydrogen and C1-4alkyl.
  • 6. The compound according to claim 5, wherein Q represents —CHRy—;R1a represents —C(═O)—NRxaRxb;Rxa and Rx represent C1-6alkyl;R1b represents F;R2 represents halo or C1-4alkyl;R21 represents hydrogen;Ry represents hydrogen;R5 represents hydrogen;R3 is selected from the group consisting of Het1; Cy2; C1-6alkyl; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2;R4 represents C1-6alkyl; in particular isopropyl;Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said heterocyclyl is optionally substituted on one nitrogen with a substituent selected from the group consisting of R6 and —C(═O)—R8;R6 represents Het4 or —C(═O)—NH—RB;R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —O—C1-4alkyl, and cyano;Het4 represents a monocyclic C-linked 5- or 6-membered aromatic ring containing one, two or three heteroatoms each independently selected from O, S, and N, or a fused bicyclic C-linked 9- or 10-membered aromatic ring containing one, two, three or four heteroatoms each independently selected from O, S, and N; wherein said aromatic ring is optionally substituted on one or two carbon atoms with in total one or two —C(═O)—NR10aR10b;Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with—C(═O)—C1-4alkyl;Het6b represents a bicyclic N-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—C1-4alkyl;Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6, Het6a, Het6b, and —NR9aR9b;R9a and R9b are each independently selected from the group consisting of hydrogen; and —S(═O)2—C1-4alkyl;R10a and R10b are each independently selected from the group consisting of hydrogen and C1-4alkyl.
  • 7. The compound according to claim 6, wherein Q represents —CHRy—;R1a represents —C(═O)—NRxaRxb;Rxa and Rxb represent C1-6alkyl;R1b represents F;R2 represents C1-4alkyl;R21 represents hydrogen;Ry represents hydrogen;R5 represents hydrogen;R3 is selected from the group consisting of Cy2; and C1-6alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1, and Cy2;R4 represents C1-6alkyl; in particular isopropyl;Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8;R6 represents —C(═O)—NH—RB;R8 represents C1-6alkyl;Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2 wherein said heterocyclyl is optionally substituted on one nitrogen with—C(═O)—C1-4alkyl;Cy2 represents C3-7cycloalkyl optionally substituted with one, two, three or four substituents each independently selected from the group consisting of R6 and Het6a.
  • 8. The compound according to claim 1, wherein Q represents —CHRy—;R1a represents —C(═O)—NRxaRxb;Rxa and Rxb are C1-6alkyl optionally substituted with 1, 2 or 3 —OH;R1b represents F;R2 represents methyl;R21 represents hydrogen or methyl;Y represents a covalent bond;n1 is 1;n2 is selected from 1 and 2;Ry represents hydrogen;R3 is selected from C1-8alkyl substituted with one, two, three or four substituents each independently selected from the group consisting of —NRxcRxd, Het1 and Cy2;Rxc and Rxd are taken together to form together with the N-atom to which they are attached a 4- to 7-membered monocyclic fully or partially saturated heterocyclyl containing one N-atom and optionally one additional heteroatom selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted with one, two or three —(C═O)—C1-4alkyl;Het1 represents a monocyclic C-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; or a bicyclic C-linked 6- to 11-membered fully or partially saturated heterocyclyl containing one, two or three heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with —C(═O)—R8; and wherein said heterocyclyl is optionally substituted on one carbon atom with oxo;R8 represents C1-6alkyl; or C1-6alkyl substituted with one, two or three substituents each independently selected from —OH, —O—C1-4alkyl and cyano;Het6a represents a monocyclic N-linked 4- to 7-membered fully or partially saturated heterocyclyl containing one N-atom and optionally one or two additional heteroatoms each independently selected from O, S, and N, wherein said S-atom might be substituted to form S(═O) or S(═O)2; wherein said heterocyclyl is optionally substituted on one nitrogen with—C(═O)—C1-4alkyl;Cy2 represents C3-7cycloalkyl optionally substituted with one Het6a.
  • 9. The compound according to claim 1, wherein R21 represents hydrogen.
  • 10. The compound according to claim 1 wherein R2 represents methyl.
  • 11. The compound according to claim 1, wherein R1b represents F.
  • 12. The compound according to claim 1, wherein —Y—R3 is attached to the nitrogen atom of the ring.
  • 13. The compound according to claim 1, wherein Formula (I) is limited to Formula (I-x):
  • 14. A compound of Formula (A)
  • 15. A pharmaceutical composition comprising a compound as claimed in claim 1, or a pharmaceutically acceptable salt or a solvate thereof and a pharmaceutically acceptable carrier or excipient.
  • 16. (canceled)
  • 17. (canceled)
  • 18. (canceled)
  • 19. (canceled)
  • 20. (canceled)
  • 21. (canceled)
  • 22. A method of treating or preventing cancer in a subject, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound as claimed in claim 1 or a pharmaceutically acceptable salt or a solvate thereof.
  • 23. The method of claim 22 wherein the cancer is selected from leukemias, lymphomas, myelomas or solid tumor cancers such as prostate cancer, lung cancer, breast cancer, pancreatic cancer, colon cancer, liver cancer, melanoma and glioblastoma.
  • 24. A method of treating or preventing leukemia, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN) in a subject, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of claim 1 or a pharmaceutically acceptable salt or a solvate thereof.
  • 25. The method of claim 24 wherein the leukemia is selected from acute leukemias, chronic leukemias, myeloid leukemias, myelogeneous leukemias, lymphoblastic leukemias, lymphocytic leukemias, Acute myelogeneous leukemias (AML), Chronic myelogenous leukemias (CML), Acute lymphoblastic leukemias (ALL), Chronic lymphocytic leukemias (CLL), T cell prolymphocytic leukemias (T-PLL), Large granular lymphocytic leukemia, Hairy cell leukemia (HCL), MLL-rearranged leukemias, MLL-PTD leukemias, MLL amplified leukemias, MLL-positive leukemias, and leukemias exhibiting HOX/MEIS1 gene expression signatures.
  • 26. The method of claim 24 wherein the leukemia is (NPM1)-mutated leukemia.
Priority Claims (2)
Number Date Country Kind
PCT/CN2021/097679 Jun 2021 WO international
PCT/CN2022/085680 Apr 2022 WO international
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2022/095901 5/30/2022 WO