Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PIPERIDINYLPYRIDINYLCARBONITRILE DERIVATIVES AS INHIBITORS OF GLUTAMINYL-PEPTIDE CYCLOTRANSFERASE AND GLUTAMINYL-PEPTIDE CYCLOTRANSFERASE LIKE PROTEIN
Document Type and Number:
WIPO Patent Application WO/2024/028245
Kind Code:
A1
Abstract:
The present disclosure provides certain piperidinylpyridinylcarbonitrile derivatives, and pharmaceutically acceptable salts thereof, that are inhibitors of Glutaminyl -peptide cyclotransferase (QPCT) and glutaminyl-peptide cyclotransferase-like protein (QPCTL), and are therefore useful for the treatment of diseases treatable by inhibition of QPCT/L. Also provided are pharmaceutical compositions containing the same, and processes for preparing said compounds.

Inventors:
WILLWACHER JENS (DE)
BINDER FLORIAN PAUL CHRISTIAN (DE)
DAHMANN GEORG (DE)
YANG HAMILTON JAMES YOUNG SOO (DE)
HANDSCHUH SANDRA RUTH (DE)
REINDL SOPHIA ASTRID (DE)
Application Number:
PCT/EP2023/071118
Publication Date:
February 08, 2024
Filing Date:
July 31, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOEHRINGER INGELHEIM INT (DE)
International Classes:
C07D401/14; A61K31/444; A61P35/00; C07D471/04; C07D487/04
Domestic Patent References:
WO2018178384A12018-10-04
WO2018178384A12018-10-04
Other References:
MARIA JIMENEZ-SANCHEZ ET AL: "siRNA screen identifies QPCT as a druggable target for Huntington's disease", NATURE CHEMICAL BIOLOGY, vol. 11, no. 5, 1 May 2015 (2015-05-01), New York, pages 347 - 354, XP055479437, ISSN: 1552-4450, DOI: 10.1038/nchembio.1790
STEPHAN SCHILLING ET AL.: "Identification of Human Glutaminyl Cyclase as a Metalloenzyme POTENT INHIBITION BY IMIDAZOLE DERIVATIVES AND HETEROCYCLIC CHELATORS", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 278, no. 50, 2003, pages 49773 - 79, Retrieved from the Internet
HOLGER CYNIS ET AL.: "Isolation of an Isoenzyme of Human Glutaminyl Cyclase: Retention in the Golgi Complex Suggests Involvement in the Protein Maturation Machinery", JOURNAL OF MOLECULAR BIOLOGY, vol. 379, no. 5, 2008, pages 966 - 80, XP022697726, Retrieved from the Internet DOI: 10.1016/j.jmb.2008.03.078
ANETT STEPHAN ET AL.: "Mammalian Glutaminyl Cyclases and Their Isoenzymes Have Identical Enzymatic Characteristics", FEBS JOURNAL, vol. 276, no. 22, 2009, pages 6522 - 36, XP055013009, Retrieved from the Internet DOI: 10.1111/j.1742-4658.2009.07337.x
MEIKE E. W. LOGTENBERG ET AL.: "Glutaminyl Cyclase Is an Enzymatic Modifier of the CD47- SIRPa Axis and a Target for Cancer Immunotherapy", NATURE MEDICINE, vol. 25, no. 4, 2019, pages 612 - 19, XP036749910, Retrieved from the Internet DOI: 10.1038/s41591-019-0356-z
ROSA BARREIRA DA SILVA ET AL.: "Loss of the Intracellular Enzyme QPCTL Limits Chemokine Function and Reshapes Myeloid Infiltration to Augment Tumor Immunity", NATURE IMMUNOLOGY, vol. 1-13, no. 4, 2022, pages 568 - 80, XP037798893, Retrieved from the Internet DOI: 10.1038/s41590-022-01153-x
ASTRID KEHLEN ET AL.: "N-Terminal Pyroglutamate Formation in CX3CL1 Is Essential for Its Full Biologic Activity", BIOSCIENCE REPORTS, vol. 37, no. 4, 2017, XP055915875, Retrieved from the Internet DOI: 10.1042/BSR20170712
CYNIS ET AL., ISOLATION OF AN ISOENZYME OF HUMAN GLUTAMINYL CYCLASE: RETENTION IN THE GOLGI COMPLEX SUGGESTS INVOLVEMENT IN THE PROTEIN MATURATION MACHINERY
ANDREAS BECKER ET AL.: "IsoQC (QPCTL) Knock-out Mice Suggest Differential Substrate Conversion by Glutaminyl Cyclase Isoenzymes", BIOLOGICAL CHEMISTRY, vol. 397, no. 1, 2016, pages 45 - 55, Retrieved from the Internet
MEIKE E.W. LOGTENBERGFERENC A. SCHEERENTON N. SCHUMACHER: "The CD47-SIRPa Immune Checkpoint", IMMUNITY, vol. 52, no. 5, 2020, pages 742 - 52, Retrieved from the Internet
DEBORAH HATHERLEY ET AL.: "Paired Receptor Specificity Explained by Structures of Signal Regulatory Proteins Alone and Complexed with CD47", MOLECULAR CELL, vol. 31, no. 2, 2008, pages 266 - 77, XP055175089, Retrieved from the Internet DOI: 10.1016/j.molcel.2008.05.026
LOGTENBERG ET AL., GLUTAMINYL CYCLASE IS AN ENZYMATIC MODIFIER OF THE CD47- SIRPA AXIS AND A TARGET FOR CANCER IMMUNOTHERAPY, 2019
KATIE-MAY MCLAUGHLIN ET AL.: "A Potential Role of the CD47/SIRPalpha Axis in COVID-19 Pathogenesis", CURRENT ISSUES IN MOLECULAR BIOLOGY, vol. 43, no. 3, 2021, pages 1212 - 25, Retrieved from the Internet
GERLINDE WERNIG ET AL.: "Unifying Mechanism for Different Fibrotic Diseases", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 114, no. 18, 2017, pages 4757 - 62, XP055459914, Retrieved from the Internet DOI: 10.1073/pnas.1621375114
LU CUI ET AL.: "Activation of JUN in Fibroblasts Promotes Pro-Fibrotic Programme and Modulates Protective Immunity", NATURE COMMUNICATIONS, vol. 11, no. 1, 2020, pages 2795, Retrieved from the Internet
WERNIG ET AL., UNIFYING MECHANISM FOR DIFFERENT FIBROTIC DISEASES
TRISTAN LERBS ET AL.: "CD47 Prevents the Elimination of Diseased Fibroblasts in Scleroderma", JCI INSIGHT, vol. 5, no. 16, 2020, pages 140458, Retrieved from the Internet
TAESIK GWAG ET AL.: "Anti-CD47 Antibody Treatment Attenuates Liver Inflammation and Fibrosis in Experimental Non-alcoholic Steatohepatitis Models", LIVER INTERNATIONAL, vol. 42, no. 4, 2022, pages 829 - 41, Retrieved from the Internet
ALEXAN-DRA L. MCCUBBREYJEFFREY L. CURTIS: "Efferocytosis and Lung Disease", CHEST, vol. 143, no. 6, 2013, pages 1750 - 57, Retrieved from the Internet
BRENNAN D. GERLACH ET AL.: "Efferocytosis Induces Macrophage Proliferation to Help Resolve Tissue Injury", CELL METABOLISM, 2021, Retrieved from the Internet
LERBS ET AL., CD47 PREVENTS THE ELIMINATION OF DISEASED FIBROBLASTS IN SCLERODERMA
LUCA RICHELDI ET AL.: "Efficacy and Safety of Nintedanib in Idiopathic Pulmonary Fibrosis", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 370, no. 22, 2014, pages 2071 - 82, XP055753710, Retrieved from the Internet DOI: 10.1056/NEJMoa1402584
KEVIN R FLAHERTY ET AL.: "Nintedanib in Progressive Fibrosing Interstitial Lung Diseases", NEW ENGLAND JOURNAL OF MEDICINE, vol. 381, no. 18, 2019, pages 1718 - 27, XP055869025, Retrieved from the Internet DOI: 10.1056/NEJMoa1908681
LUCA RICHELDI ET AL.: "Trial of a Preferential Phosphodiesterase 4B Inhibitor for Idiopathic Pulmonary Fibrosis", NEW ENGLAND JOURNAL OF MEDICINE, vol. 386, no. 23, 2022, pages 2178 - 87, XP093048263, Retrieved from the Internet DOI: 10.1056/NEJMoa2201737
STEPHEN B. WILLING-HAM ET AL.: "The CD47-Signal Regulatory Protein Alpha (SIRPa) Interaction Is a Therapeutic Target for Human Solid Tumors", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 109, no. 17, 2012, pages 6662 - 67, Retrieved from the Internet
HOLGER CYNIS ET AL.: "The Isoenzyme of Glutaminyl Cyclase Is an Important Regulator of Monocyte Infiltration under Inflammatory Conditions", EMBO MOLECULAR MEDICINE, vol. 3, no. 9, 2011, pages 545 - 58, XP055459940, Retrieved from the Internet DOI: 10.1002/emmm.201100158
KASPAR BRESSER ET AL.: "QPCTL Regulates Macrophage and Monocyte Abundance and Inflammatory Signatures in the Tumor Microenvironment", ONCOIMMUNOLOGY, vol. 11, no. 1, 2022, pages 2049486, Retrieved from the Internet
JIMENEZ-SANCHEZ ET AL., NATURE CHEMICAL BIOLOGY, vol. 11, 2015, pages 347 - 357
POZZI, C ET AL., JOURNAL OF BIOLOGICAL INORGANIC CHEMISTRY, vol. 23, no. 8, 2018, pages 1219 - 1226
RICHARD C. LAROCK: "Comprehensive Organic Transformations", 2010, JOHN WILEY & SONS
MICHAEL B. SMITH: "March's Advanced Organic Chemistry", 2013, JOHN WILEY & SONS
PHILIP J. KOCIENSKI: "Thieme", 2005, article "Protecting Groups"
PETER G. M. WUTSTHEODORA W. GREENE: "Protective Groups in Organic Synthesis", 2006, JOHN WILEY & SONS
Attorney, Agent or Firm:
SIMON, Elke Anna Maria et al. (DE)
Download PDF:
Claims:
WHAT IS CLAIMED 1. A compound of formula (I) wherein Y is N and Z is R4C or Y is HC and Z is N; A is A1a which is a 5- or 6-membered mono-heteroaryl ring containing one nitrogen or containing two heteroatom members wherein one is nitrogen and the second is selected from nitrogen or sulphur; or A is A1b which is a 9- or 10-membered fused bicyclic-heteroaryl ring containing one to four nitrogens; and wherein A1a or A1b is independently substituted with one or two R3; R1 is selected from the group R1a, consisting of H, C1-4-alkyl and halo; R2 is selected from the group R2a, consisting of halo, H, C1-4-alkyl, C3-4-cycloalkyl and F1- 9-fluoro-C1-4-alkyl; R3 is selected from the group R3a, consisting of H, halo, C1-4-alkyl, C3-4-cycloalkyl, C3-4- fluorocycloalkyl, F1-9-fluoro-C1-4-alkyl, C1-4-alkyloxy, C3-4-cycloalkyloxy and pyrazolyl, and R4 is selected from the group R4a, consisting of halo, H, C1-4-alkyl, C3-4-cycloalkyl and F1- 9-fluoro-C1-4-alkyl; or a salt thereof. 2. The compound of formula (I) according to claim 1, wherein A is selected from the group A3 consisting of pyridinyl, pyrazinyl, pyrazolyl, isothiazolyl, imidazo[1,2‐a]pyrimidyl, py- razolo[3,4‐b]pyridinyl, [1,2,4]triazolo[4,3‐a]pyrimidyl, pyrazolo[1,5-b]pyridazinyl, [1,2,4]triazolo[1,5‐a]pyrimidinyl, 2H‐[1,2,3]triazolo[4,5‐b]pyridinyl, 1H‐[1,2,3]tria- zolo[4,5‐b]pyridinyl and 1H‐imidazo[4,5‐b]pyridinyl and wherein A3 is independently substituted with one or two R3; or a salt thereof. 3. The compound of formula (I) according to claim 1, wherein A is selected from the group A9 consisting of , , , , , , , , , and wherein A9 is independently substituted with one or two R3; or a salt thereof. 4. The compound of formula (I) according to any of claims 1 to 3, wherein R2 is selected from the group R2b, consisting of H, F, Cl, C1-4-alkyl and F1-9-fluoro-C1-4-alkyl; or a salt thereof. 5. The compound of formula (I) according to any of claims 1 to 4, wherein R3 is selected from the group R3b, consisting of H, halo, C1-4-alkyl, C3-4-cycloalkyl, F1-9-fluoro-C1-4-al- kyl, C1-4-alkyloxy, C3-4-cycloalkyloxy and pyrazolyl, and or a salt thereof. 6. The compound of formula (I) according to any of claims 1 to 5, having formula (I-a) or a salt thereof.

7. The compound of formula (I) according to any of claims 1 to 5, having formula (I-b) or a salt thereof. 8. The compound of formula (I) according to claim 1, selected from the group consisting of

, , and or a salt thereof.

9. A pharmaceutically acceptable salt of a compound according to one or more of claims 1 to 8. 10. A pharmaceutical composition comprising one or more compounds according to one or more of claims 1 to 8, or pharmaceutically acceptable salts thereof, optionally together with one or more inert carriers and/or diluents. 11. A pharmaceutical composition comprising one or more compounds according to one or more of the claims 1 to 8, or pharmaceutically acceptable salts thereof, and one or more ad- ditional therapeutic agents, optionally together with one or more inert carriers and/or dilu- ents. 12. The pharmaceutical composition according to claim 11 wherein the one or more addi- tional therapeutic agents are selected from the group consisting of anticancer agents and antifibrotic agents. 13. The compound according to one or more of claims 1 to 8 or a pharmaceutically ac- ceptable salt thereof for use as a medicament. 14. A method for the treatment of diseases, such as cancer or fibrotic diseases, and condi- tions associated with these diseases, in a patient in need thereof, the method being charac- terized in that one or more compounds according to one or more of claims 1 to 8 or phar- maceutically acceptable salts thereof are administered to the patient. 15. A compound according to one or more of claims 1 to 8 or a pharmaceutically accepta- ble salt thereof for use in a method for the treatment of cancer, fibrotic diseases, neuro- degenerative diseases, atherosclerosis, infectious diseases, or chronic kidney diseases.

Description:
PIPERIDINYLPYRIDINYLCARBONITRILE DERIVATIVES AS INHIBITORS OF GLUTAMINYL-PEPTIDE CYCLOTRANSFERASE AND GLUTAMINYL- PEPTIDE CYCLOTRANSFERASE LIKE PROTEIN TECHNICAL FIELD The present disclosure provides certain piperidinylpyridinylcarbonitrile derivatives, and pharmaceutically acceptable salts thereof, that are inhibitors of Glutaminyl-peptide cy- clotransferase (QPCT) and glutaminyl-peptide cyclotransferase-like protein (QPCTL), and are therefore useful for the treatment of diseases treatable by inhibition of QPCT/L. Also provided are pharmaceutical compositions containing the same, and processes for preparing said compounds. BACKGROUND INFORMATION Glutaminyl-peptide cyclotransferase (QPCT) and glutaminyl-peptide cyclotransferase-like protein (QPCTL) catalyze the intramolecular cyclization of N-terminal glutamine (Q) resi- dues into pyroglutamic acid (pE) liberating ammonia [Stephan Schilling et al., “Identifica- tion of Human Glutaminyl Cyclase as a Metalloenzyme POTENT INHIBITION BY IMID- AZOLE DERIVATIVES AND HETEROCYCLIC CHELATORS,” Journal of Biological Chemistry 278, no. 50 (2003): 49773–79, https://doi.org/10.1074/jbc.m309077200; Holger Cynis et al., “Isolation of an Isoenzyme of Human Glutaminyl Cyclase: Retention in the Golgi Complex Suggests Involvement in the Protein Maturation Machinery,” Journal of Molecular Biology 379, no. 5 (2008): 966–80, https://doi.org/10.1016/j.jmb.2008.03.078; Anett Stephan et al., “Mammalian Glutaminyl Cyclases and Their Isoenzymes Have Identi- cal Enzymatic Characteristics,” FEBS Journal 276, no. 22 (2009): 6522–36, https://doi.org/10.1111/j.1742-4658.2009.07337.x.]. While QPCT is a secreted protein, QPCTL is retained within the Golgi complex. Both enzymes share a high homology in the active site and similar catalytic specificity. Because of the high homology in the active site, inhibition of the active site blocks the enzymatic activity of both enzymes: QPCT and QPCTL. Hence the term “QPCT/L” describes both enzymes at once. Due to their different cellular localisation, differences in their relevance for modification of biological substrates have been reported. Known substrates of the intracellular QPCTL and/or extracellular QPCT are CD47 [Meike E. W. Logtenberg et al., “Glutaminyl Cyclase Is an Enzymatic Modifier of the CD47- SIRPα Axis and a Target for Cancer Immunotherapy,” Nature Medicine 25, no. 4 (2019): 612–19, https://doi.org/10.1038/s41591-019-0356-z.], different chemokines (like for example CCL2 and 7 or CX3CL1) [Rosa Barreira da Silva et al., “Loss of the In- tracellular Enzyme QPCTL Limits Chemokine Function and Reshapes Myeloid Infiltration to Augment Tumor Immunity,” Nature Immunology 23, no. 4 (2022): 568–80, https://doi.org/10.1038/s41590-022-01153-x; Astrid Kehlen et al., “N-Terminal Pyrogluta- mate Formation in CX3CL1 Is Essential for Its Full Biologic Activity,” Bioscience Reports 37, no. 4 (2017): BSR20170712, https://doi.org/10.1042/bsr20170712.], Amyloid-b pep- tides [Cynis et al., “Isolation of an Isoenzyme of Human Glutaminyl Cyclase: Retention in the Golgi Complex Suggests Involvement in the Protein Maturation Machinery.”] or hor- mones like TRH [Andreas Becker et al., “IsoQC (QPCTL) Knock-out Mice Suggest Differ- ential Substrate Conversion by Glutaminyl Cyclase Isoenzymes,” Biological Chemistry 397, no.1 (2016): 45–55, https://doi.org/10.1515/hsz-2015-0192.]. The modification of N-termi- nal glutamine to pyroglutamate on the substrates has functional consequences for the pro- teins and could impact different pathomechanisms in several diseases. CD47 is expressed on the cell surface of virtually all cells of the body, including apoptotic cells, senescent cells or cancer cells. [Meike E.W. Logtenberg, Ferenc A. Scheeren, and Ton N. Schumacher, “The CD47-SIRPα Immune Checkpoint,” Immunity 52, no. 5 (2020): 742–52, https://doi.org/10.1016/j.immuni.2020.04.011]. The main ligand for CD47 is signal-regula- tory protein alpha (SIRP ^), an inhibitory transmembrane receptor present on myeloid cells, such as macrophages, monocytes, neutrophils, dendritic cells and others. QPCTL mediated N-terminal pyroglutamate modification on CD47 is required for SIRP ^ binding [Deborah Hatherley et al., “Paired Receptor Specificity Explained by Structures of Signal Regulatory Proteins Alone and Complexed with CD47,” Molecular Cell 31, no. 2 (2008): 266–77, https://doi.org/10.1016/j.molcel.2008.05.026; Meike E. W. Logtenberg et al., “Glutaminyl Cyclase Is an Enzymatic Modifier of the CD47- SIRPα Axis and a Target for Cancer Immu- notherapy,” Nature Medicine 25, no.4 (2019): 612–19, https://doi.org/10.1038/s41591-019- 0356-z.] This signaling axis induces a “Don’t Eat Me Signal”, preventing engulfment of CD47 expressing cells by macrophages. Thus, high expression of CD47 is connected to the pathogenesis of cancer [Logtenberg et al., “Glutaminyl Cyclase Is an Enzymatic Modifier of the CD47- SIRPα Axis and a Target for Cancer Immunotherapy,” 2019; Meike E.W. Log- tenberg, Ferenc A. Scheeren, and Ton N. Schumacher, “The CD47-SIRPα Immune Checkpoint,” Immunity 52, no. 5 (2020): 742–52, https://doi.org/10.1016/j.im- muni.2020.04.011.], COVID-19 [Katie-May McLaughlin et al., “A Potential Role of the CD47/SIRPalpha Axis in COVID-19 Pathogenesis,” Current Issues in Molecular Biology 43, no.3 (2021): 1212–25, https://doi.org/10.3390/cimb43030086.], lung fibrosis [Gerlinde Wernig et al., “Unifying Mechanism for Different Fibrotic Diseases,” Proceedings of the National Academy of Sciences 114, no. 18 (2017): 4757–62, https://doi.org/10.1073/pnas.1621375114; Lu Cui et al., “Activation of JUN in Fibroblasts Promotes Pro-Fibrotic Programme and Modulates Protective Immunity,” Nature Communi- cations 11, no. 1 (2020): 2795, https://doi.org/10.1038/s41467-020-16466-4.], systemic sclerosis [Wernig et al., “Unifying Mechanism for Different Fibrotic Diseases”; Tristan Lerbs et al., “CD47 Prevents the Elimination of Diseased Fibroblasts in Scleroderma,” JCI Insight 5, no. 16 (2020): e140458, https://doi.org/10.1172/jci.insight.140458.] and liver fi- brosis [Taesik Gwag et al., “Anti‐CD47 Antibody Treatment Attenuates Liver Inflammation and Fibrosis in Experimental Non‐alcoholic Steatohepatitis Models,” Liver International 42, no.4 (2022): 829–41, https://doi.org/10.1111/liv.15182.]. Since enhanced CD47 expression blocks the clearance of apoptotic cells, there is an accrual of apoptotic lung epithelial cells, leading to a pro-fibrotic stimulus and accelerating lung inflammation and -scaring [Alexan- dra L. McCubbrey and Jeffrey L. Curtis, “Efferocytosis and Lung Disease,” Chest 143, no. 6 (2013): 1750–57, https://doi.org/10.1378/chest.12-2413; Brennan D. Gerlach et al., “Ef- ferocytosis Induces Macrophage Proliferation to Help Resolve Tissue Injury,” Cell Metab- olism, 2021, https://doi.org/10.1016/j.cmet.2021.10.015.]. Since CD47 half-life and function is majorly dependent on QPCTL enzyme activity, QPCT and QPCTL inhibition could be a suitable mechanism as a treatment in lung fibrosis such as IPF or SSC-ILD [Lerbs et al., “CD47 Prevents the Elimination of Diseased Fibroblasts in Scleroderma.”], alone or together with current standard of care in pulmonary fibrosis like Nintedanib [Luca Richeldi et al., “Efficacy and Safety of Nintedanib in Idiopathic Pulmonary Fibrosis,” The New England Journal of Medicine 370, no.22 (2014): 2071–82, https://doi.org/10.1056/nejmoa1402584; Kevin R Flaherty et al., “Nintedanib in Progressive Fibrosing Interstitial Lung Diseases,” New England Journal of Medicine 381, no. 18 (2019): 1718–27, https://doi.org/10.1056/nejmoa1908681.] or future treatments like a PDE4 inhibitor [Luca Richeldi et al., “Trial of a Preferential Phosphodiesterase 4B Inhibitor for Idiopathic Pulmo- nary Fibrosis,” New England Journal of Medicine 386, no. 23 (2022): 2178–87, https://doi.org/10.1056/nejmoa2201737]. By expression of CD47, cancer cells can evade destruction by the immune system or evade immune surveillance, e.g. by evading phagocytosis by immune cells [Stephen B. Willing- ham et al., “The CD47-Signal Regulatory Protein Alpha (SIRPa) Interaction Is a Therapeutic Target for Human Solid Tumors,” Proceedings of the National Academy of Sciences 109, no.17 (2012): 6662–67, https://doi.org/10.1073/pnas.1121623109]. In addition to CD47, chemokines, such as CCL2 and CX3CL1, have been identified as QPCTL and/or QPCT substrates [Holger Cynis et al., “The Isoenzyme of Glutaminyl Cyclase Is an Important Regulator of Monocyte Infiltration under Inflammatory Condi- tions,” EMBO Molecular Medicine 3, no. 9 (2011): 545–58, https://doi.org/10.1002/emmm.201100158]. The formation of the N-terminal pGlu was shown to increase in vivo activity, both by conferring resistance to aminopeptidases and by increasing its capacity to induce chemokine receptor signaling. Two main monocyte chem- oattractants CCL2 and CCL7 are insensitive to DPP4-inactivation in vivo because of an in- tracellular mechanism of N-terminal cyclization mediated by the Golgi-associated enzyme QPCTL. It has been shown that QPCTL is a critical regulator of monocyte migration into solid tumors [Kaspar Bresser et al., “QPCTL Regulates Macrophage and Monocyte Abun- dance and Inflammatory Signatures in the Tumor Microenvironment,” Oncoimmunology 11, no. 1 (2022): 2049486, https://doi.org/10.1080/2162402x.2022.2049486; Rosa Barreira da Silva et al., “Loss of the Intracellular Enzyme QPCTL Limits Chemokine Function and Re- shapes Myeloid Infiltration to Augment Tumor Immunity,” Nature Immunology, 2022, 1– 13, https://doi.org/10.1038/s41590-022-01153-x]. Targeting of chemokines has long been pursued as a potential strategy for modulating cellular trafficking in different disease set- tings. It is therefore desirable to provide potent QPCT/L inhibitors. Jimenez-Sanchez, et al., Nature Chemical Biology, 2015, 11, 347-357, (hereinafter “J-S, NCB 2015”) discloses the human glutaminyl cyclase (hQC) inhibitors SEN177 and SEN180:

SEN177 is disclosed therein (supplementary information) as having a IC50 on isolated hQC of 53 nM and on isolated QPCTL of 13 nM. SEN180 is disclosed therein (supplementary information) as having a IC50 on hQC of 170 nM and on QPCTL of 58 nM. Pozzi, C, et al, Journal of Biological Inorganic Chemistry, 2018, 23, (8), 1219-1226, (here- inafter “P, JBIC 2018”), further discloses SEN177 and its binding mode within the hQC cavity. SEN177 is disclosed therein as having a Ki on isolated hQC of 20 nM. WO 2018/178384 discloses QPCTL inhibitors of the general formula A-B-D-E, which in- clude examples 1094 and 1095 (Formula (XIIa) on page 123 and table on page 125): . WO 2018/178384 does not disclose any biological data for examples 1094 or 1095. DETAILED DESCRIPTION OF THE INVENTION The present invention discloses novel piperidinylpyridinylcarbonitrile derivatives of for- mula (I) that are inhibitors of Glutaminyl-peptide cyclotransferase (QPCT) and glutaminyl-peptide cyclotransferase-like protein (QPCTL), possessing appropriate pharmacological and phar- macokinetic properties enabling their use as medicaments for the treatment of conditions and/or diseases treatable by inhibition of QPCT/L. The compounds of the present invention may provide several advantages, such as enhanced potency, cellular potency, high metabolic and/or chemical stability, high selectivity, safety and tolerability, enhanced solubility, enhanced permeability, desirable plasma protein bind- ing, enhanced bioavailability, suitable pharmacokinetic profiles, and the possibility to form stable salts. Compounds of the invention The present invention provides novel piperidinylpyridinylcarbonitrile derivatives that sur- prisingly, are potent inhibitors of QPCT and QPCTL (Assay A), as well as potent inhibi- tors of QPCT/L in cells relevant for, but not limited to, lung diseases or cancer, (Assay B). Furthermore, the present novel piperidinylpyridinylcarbonitrile derivatives have appropri- ate membrane permeability and a low in vitro efflux (Assay C). Consequently, compounds of the present invention are more viable for human use. Compounds of the present invention differ structurally from SEN177 in Pozzi, C, et al, Jour- nal of Biological Inorganic Chemistry, 2018, 23, (8), 1219-1226, in that the pyridinyl ring attached to the piperidinyl ring contains a ring-nitrogen which is in either of two meta-posi- tions to the piperidinyl ring attachment position. Furthermore, a carbonitrile substituent is attached at the ortho-position to the piperidinyl ring attachment position of said pyridinyl ring. Still further, R1 and R2 are not limited to hydrogen or methyl and A represents hetero- cyclic ring systems beyond pyridinyl. Compounds of the present invention differ structurally from examples 1094 and 1095 in WO 2018/178384 in that the pyridinyl ring attached to the piperidinyl ring contains a ring-nitro- gen which is in either of two meta-positions to the piperidinyl ring attachment position. Fur- thermore, a carbonitrile substituent is attached at the ortho-position to the piperidinyl ring attachment position of said pyridinyl ring. Still further, R1 and R2 are not limited to hydrogen and A represents heterocyclic ring systems beyond pyridinyl. Still further, the 5-membered heterocyclic ring attached to the piperidinyl ring at the 4-position relative to the piperidinyl nitrogen is in example 1094 an aminothiazolyl ring and in example 1095 an aminothiadia- zolyl ring whereas in compounds of the present invention it is a 3-substituted-4‐methyl‐4H‐ 1,2,4‐triazolyl ring. These structural differences between compounds of the present invention and the prior art unexpectedly lead to a favourable combination of (i) potent inhibition of QPCT and QPCTL, (ii) potent inhibition of QPCT/L in cells relevant for, but not limited to, lung diseases or cancer, and (iii) appropriate membrane permeability and a low in vitro efflux. Compounds of the invention are thus superior to those disclosed in the prior art in terms of the combination of the following parameters: ^ potent inhibition of QPCT and QPCTL (Assay A) ^ potent inhibition of QPCT/L in cells relevant for, but not limited to, lung diseases or cancer (Assay B) ^ appropriate membrane permeability and a low in vitro efflux (Assay C) The present invention provides novel compounds according to formula (I)

wherein Y is N and Z is R4C or Y is HC and Z is N; A is A1a which is a 5- or 6-membered mono-heteroaryl ring containing one nitrogen or containing two heteroatom members wherein one is nitrogen and the second is selected from nitrogen or sulphur; or A is A1b which is a 9- or 10-membered fused bicyclic-heteroaryl ring containing one to four nitrogens; and wherein A1a or A1b is independently substituted with one or two R3; R1 is selected from the group R1a, consisting of H, C1-4-alkyl and halo; R2 is selected from the group R2a, consisting of halo, H, C 1-4 -alkyl, C 3-4 -cycloalkyl and F 1- 9 -fluoro-C 1-4 -alkyl; R3 is selected from the group R3a, consisting of H, halo, C1-4-alkyl, C3-4-cycloalkyl, C3-4- fluorocycloalkyl, F 1-9 -fluoro-C 1-4 -alkyl, C 1-4 -alkyloxy, C 3-4 -cycloalkyloxy and pyrazolyl, and R4 is selected from the group R4a, consisting of halo, H, C1-4-alkyl, C3-4-cycloalkyl and F1- 9-fluoro-C 1-4 -alkyl; or a salt thereof, particularly a pharmaceutically acceptable salt thereof. Another embodiment of the present invention relates to a compound of formula (I), wherein A is A2a which is a 5- or 6-membered mono-heteroaryl ring containing one nitrogen or containing two heteroatom members wherein one is nitrogen and the second is selected from nitrogen or sulphur; and wherein A2a is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is A2b which is a 9- or 10-membered fused bicyclic-heteroaryl ring containing three to four nitrogens; and wherein A2b is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A3 consisting of pyridinyl, pyrazinyl, pyrazolyl, iso- thiazolyl, imidazo[1,2‐a]pyrimidyl, pyrazolo[3,4‐b]pyridinyl, [1,2,4]triazolo[4,3‐a]pyrim- idyl, pyrazolo[1,5-b]pyridazinyl, [1,2,4]triazolo[1,5‐a]pyrimidinyl, 2H‐[1,2,3]triazolo[4,5‐ b]pyridinyl, 1H‐[1,2,3]triazolo[4,5‐b]pyridinyl and 1H‐imidazo[4,5‐b]pyridinyl and wherein A3 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A4 consisting of pyridinyl, pyrazinyl, pyrazolyl, isothiazolyl, imidazo[1,2‐a]pyrimidyl, pyrazolo[3,4‐b]pyridinyl, [1,2,4]triazolo[4,3‐a]py- rimidyl and pyrazolo[1,5-b]pyridazinyl; and wherein A4 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A5 consisting of pyridinyl, imidazo[1,2‐a]pyrimidyl, pyrazolo[3,4‐b]pyridinyl, [1,2,4]triazolo[4,3‐a]pyrimidyl and pyrazolo[1,5-b]pyridazinyl; and wherein A5 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A6 consisting of pyridinyl and pyrazolo[1,5-b]pyri- dazinyl; and wherein A6 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A7 consisting of pyridinyl; and wherein A7 is inde- pendently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A8 consisting of pyrazolo[1,5-b]pyridazinyl; and wherein A8 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A9 consisting of

and wherein A9 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A10 consisting of , , , , , , and wherein A10 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A11 consisting of and ; wherein A11 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A12 consisting of and wherein A12 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A13 consisting of and wherein A13 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A14 consisting of wherein A14 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group A15 consisting of wherein A15 is independently substituted with one or two R3; and substituents R1, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R1 is selected from the group R1b, consisting of H, H3C-, H3CH2C-, H3CH2CH2C-, (H3C)2HC-, Cl and F; and substituents A, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R1 is selected from the group R1c, consisting of H, H 3 C- and F; and substituents A, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R1 is selected from the group R1d, consisting of H; and substituents A, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R1 is selected from the group R1e, consisting of F; and substituents A, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R1 is selected from the group R1f, consisting of H3C- and F; and substituents A, R2, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R2 is selected from the group R2b, consisting of H, F, Cl, C 1-4 -alkyl and F 1-9 - fluoro-C1-4-alkyl; and substituents A, R1, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R2 is selected from the group R2c, consisting of H, C1-4-alkyl and F1-9-fluoro-C1-4- alkyl; and substituents A, R1, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R2 is selected from the group R2d, consisting of H, H 3 C-, F 3 C-; and substituents A, R1, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R2 is selected from the group R2e, consisting of H; and substituents A, R1, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R2 is selected from the group R2f, consisting of H3C- and F3C-; and substituents A, R1, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R2 is selected from the group R2g, consisting of F, Cl, C1-4-alkyl and F1-9-fluoro- C 1-4 -alkyl; and substituents A, R1, R3 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R3 is selected from the group R3b, consisting of H, halo, C1-4-alkyl, C3-4-cycloal- kyl, F1-9-fluoro-C1-4-alkyl, C1-4-alkyloxy, C3-4-cycloalkyloxy and pyrazolyl, and ; and substituents A, R1, R2 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R3 is selected from the group R3c, consisting of H, F, Cl, H 3 C, (H 3 C) 3 C-, H 3 C-O-, F3C-, and and substituents A, R1, R2 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R3 is selected from the group R3d, consisting of H, F, Cl, H 3 C, (H 3 C) 3 C-, H 3 C-O-, F3C- and and substituents A, R1, R2 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R3 is selected from the group R3e, consisting of H, H3C-O-, and ; and substituents A, R1, R2 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R3 is selected from the group R3f, consisting of H, H 3 C, (H 3 C) 3 C-, H 3 C-O- and F3C-; and substituents A, R1, R2 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R3 is selected from the group R3g, consisting of H, F and Cl; and substituents A, R1, R2 and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R4 is selected from the group R4b, consisting of H, F, Cl, C 1-4 -alkyl and F 1-9 - fluoro-C1-4-alkyl; and substituents A, R1, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R4 is selected from the group R4c, consisting of H, C1-4-alkyl and F1-9-fluoro-C1-4- alkyl; and substituents A, R1, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R4 is selected from the group R4d, consisting of H, H3C-, F3C-; and substituents A, R1, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R4 is selected from the group R4e, consisting of H; and substituents A, R1, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I), wherein R4 is selected from the group R4f, consisting of H 3 C- and F 3 C-; and substituents A, R1, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I) above, having formula (I-a) wherein substituents A, R1, R2, R3 and R4 are defined as in any of the preceding embodi- ments. Another embodiment of the present invention relates to a compound of formula (I) above, having formula (I-b) wherein substituents A, R1, R2 and R3 are defined as in any of the preceding embodi- ments. Another embodiment of the present invention relates to a compound of formula (I) above, having formula (I-c) wherein substituents A, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I) above, having formula (I-d) wherein substituents A, R2, R3and R4 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I) above, having formula (I-e) wherein substituents A, R2 and R3 are defined as in any of the preceding embodiments. Another embodiment of the present invention relates to a compound of formula (I) above, having formula (I-f) wherein substituents R2, R3 and R4 are defined as in any of the preceding embodiments. Further preferred embodiments of the compounds of formula (I) are encompassed as em- bodiments (EMB-1) to (EMB-20) in the following Table 1, wherein the substituent defini- tions above are employed. Table 1: further preferred embodiments For example, compounds of the embodiment EMB-1 have for R1 the genus group R1e as defined above, in combination with the other genus groups for the other substituents in for- mula (I) as defined within the same row of the table. The same applies analogously to the other variables incorporated in the general formulae. Particularly preferred is the compound according to formula (I) selected from the group consisting of

and Particularly preferred is the compound according to formula (I) selected from the group consisting of example 1, example 2, example 3, example 4, example 5, example 6, example 7, example 8, example 9, example 10, example 11, example 12, example 13, example 14, example 15, example 16, example 17, example 18, example 19, example 20, example 21, example 22, example 23, example 24, example 25, example 26, example 27, example 28, example 29, example 30, example 31 and example 32, as described hereinafter in EXAM- PLES. Particularly preferred is the compound according to formula (I) selected from the group consisting of example 1, example 3, example 4, example 5, example 6, example 7, example 8, example 12, example 13, example 15, example 16, example 17, example 19, example 21, example 25, example 26, example 27, example 28, example 29, example 30, example 31 and example 32, as described hereinafter in EXAMPLES. Particularly preferred is the compound according to formula (I) selected from the group consisting of example 1, example 2, example 3, example 4, example 6, example 7, example 10, example 14, example 16, example 17, example 18, example 26 and example 27 as de- scribed hereinafter in EXAMPLES. The present invention provides novel piperidinylpyridinylcarbonitrile derivatives of for- mula (I) that are surprisingly potent QPCT/L inhibitors. Another aspect of the invention refers to compounds according to formula (I) as surpris- ingly having potent inhibition of QPCT/L in cells relevant for, but not limited to, lung dis- eases or cancer. Another aspect of the invention refers to compounds according to formula (I) as surpris- ingly cellular potent QPCT/L inhibitors having appropriate membrane permeability and low in vitro efflux. Another aspect of the invention refers to pharmaceutical compositions, containing at least one compound according to formula (I) optionally together with one or more inert carriers and/or diluents. A further aspect of the present invention refers to compounds according to formula (I), for the use in the prevention and/or treatment of disorders associated with QPCT/L inhibition. Another aspect of the invention refers to processes of manufacture of the compounds of the present invention. Further aspects of the present invention will become apparent to the skilled artisan directly from the foregoing and following description and the examples. USED TERMS AND DEFINITIONS General Definitions Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to. In the groups, radicals, or moieties defined below, the number of carbon atoms is often specified preceding the group, for example, C1-6-alkyl means an alkyl group or radical hav- ing 1 to 6 carbon atoms. In general in groups like HO, H2N, (O)S, (O)2S, NC (cyano), HOOC, F 3 C or the like, the skilled artisan can see the radical attachment point(s) to the molecule from the free valences of the group itself. For combined groups comprising two or more subgroups, the last named subgroup is the radical attachment point, for example, the substituent "aryl-C 1-3 -alkylene" means an aryl group which is bound to a C 1-3 -alkyl- group, the latter of which is bound to the core or to the group to which the substituent is at- tached. In case a compound of the present invention is depicted in the form of a chemical name and as a formula, in case of any discrepancy the formula shall prevail. An asterisk may be used in sub-formulas to indicate the bond which is connected to the core molecule as de- fined. The numeration of the atoms of a substituent starts with the atom which is closest to the core or to the group to which the substituent is attached. For example, the term "3-carboxypropyl-group" represents the following substituent: wherein the carboxy group is attached to the third carbon atom of the propyl group. The terms "1-methylpropyl-", "2,2-dimethylpropyl-" or "cyclopropylmethyl-" group represent the following groups: The asterisk may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined. The term "substituted" as used herein, means that one or more hydrogens on the designated atom are replaced by a group selected from a defined group of substituents, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound. Likewise, the term “substituted” may be used in connection with a chemical moiety instead of a single atom, e.g. “substituted alkyl”, “substituted aryl” or the like. Unless specifically indicated, throughout the specification and the appended claims, a given chemical formula or name shall encompass tautomers and all stereo, optical and geo- metrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc…) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantio- mers exist, as well as solvates thereof such as for instance hydrates. Unless specifically indicated, also “pharmaceutically acceptable salts” as defined in more detail below shall encompass solvates thereof such as for instance hydrates. In general, substantially pure stereoisomers can be obtained according to synthetic princi- ples known to a person skilled in the field, e.g. by separation of corresponding mixtures, by using stereochemically pure starting materials and/or by stereoselective synthesis. It is known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, e.g. starting from optically active starting materials and/or by using chiral reagents. Enantiomerically pure compounds of this invention or intermediates may be prepared via asymmetric synthesis, for example by preparation and subsequent separation of appropriate diastereomeric compounds or intermediates which can be separated by known methods (e.g. by chromatographic separation or crystallization) and/or by using chiral reagents, such as chiral starting materials, chiral catalysts or chiral auxiliaries. Further, it is known to the person skilled in the art how to prepare enantiomerically pure compounds from the corresponding racemic mixtures, such as by chromatographic separa- tion of the corresponding racemic mixtures on chiral stationary phases; or by resolution of a racemic mixture using an appropriate resolving agent, e.g. by means of diastereomeric salt formation of the racemic compound with optically active acids or bases, subsequent resolution of the salts and release of the desired compound from the salt; or by derivatiza- tion of the corresponding racemic compounds with optically active chiral auxiliary rea- gents, subsequent diastereomer separation and removal of the chiral auxiliary group; or by kinetic resolution of a racemate (e.g. by enzymatic resolution); by enantioselective crystal- lization from a conglomerate of enantiomorphous crystals under suitable conditions; or by (fractional) crystallization from a suitable solvent in the presence of an optically active chi- ral auxiliary. The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio. As used herein, "pharmaceutically acceptable salt" refers to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. For example, such salts include salts from benzenesulfonic acid, benzoic acid, citric acid, ethanesulfonic acid, fumaric acid, gentisic acid, hydrobromic acid, hydrochloric acid, ma- leic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, 4-methyl-ben- zenesulfonic acid, phosphoric acid, salicylic acid, succinic acid, sulfuric acid and tartaric acid. Further pharmaceutically acceptable salts can be formed with cations from ammonia, L-arginine, calcium, 2,2’-iminobisethanol, L-lysine, magnesium, N-methyl-D-glucamine , potassium, sodium and tris(hydroxymethyl)-aminomethane. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical meth- ods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an or- ganic diluent such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof. Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention (e.g. trifluoro acetate salts,) also com- prise a part of the invention. The term halogen denotes fluorine, chlorine, bromine and iodine. The term "C1-n-alkyl", wherein n is an integer selected from 2, 3, 4, 5 or 6, preferably 4, 5, or 6, either alone or in combination with another radical, denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to n C atoms. For example the term C 1-5 -al- kyl embraces the radicals H3C-, H3C-CH2-, H3C-CH2-CH2-, H3C-CH(CH3)-, H3C-CH2-CH2-CH2-, H3C-CH2-CH(CH3)-, H3C-CH(CH3)-CH2-, H3C-C(CH3)2-, H 3 C-CH 2 -CH 2 -CH 2 -CH 2 -, H 3 C-CH 2 -CH 2 -CH(CH 3 )-, H 3 C-CH 2 -CH(CH 3 )-CH 2 -, H3C-CH(CH3)-CH2-CH2-, H3C-CH2-C(CH3)2-, H3C-C(CH3)2-CH2-, H3C-CH(CH3)-CH(CH3)- and H3C-CH2-CH(CH2CH3)-. The term "C 3-k -cycloalkyl", wherein k is an integer selected from 3, 4, 5, 7 or 8, preferably 4, 5 or 6, either alone or in combination with another radical, denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to k C atoms. For example the term C 3-7 -cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. The term "halo" added to an "alkyl", "alkylene" or "cycloalkyl" group (saturated or unsatu- rated) defines an alkyl, alkylene or cycloalkyl group wherein one or more hydrogen atoms are replaced by a halogen atom selected from among fluorine, chlorine or bromine, prefer- ably fluorine and chlorine, particularly preferred is fluorine. Examples include: H2FC-, HF 2 C-, F 3 C-. The term "mono-heteroaryl ring" means a monocyclic aromatic ring system, containing one or more heteroatoms selected from N or S, consisting of 5 to 6 ring atoms. The term "mono-heteroaryl ring" is intended to include all the possible isomeric forms. Thus, the term "mono-heteroaryl ring" includes the following exemplary structures (not de- picted as radicals as each form is optionally attached through a covalent bond to any atom so long as appropriate valences are maintained): The term "fused bicyclic-heteroaryl ring" means a bicyclic aromatic ring system, contain- ing one or more heteroatoms selected from N or S, consisting of 9 to 10 ring atoms. The term "fused bicyclic-heteroaryl ring" is intended to include all the possible isomeric forms. Thus, the term "bicyclic-heteroaryl ring" includes the following exemplary structures (not depicted as radicals as each form is optionally attached through a covalent bond to any atom so long as appropriate valences are maintained):

The term pyridinyl refers to the radical of the following ring: . The term pyrazinyl refers to the radical of the following ring: The term pyrazolyl refers to the radical of the following ring: The term isothiazolyl refers to the radical of the following ring: . The term imidazo[1,2‐a]pyrimidyl refers to the radical of the following ring: The term pyrazolo[3,4‐b]pyridinyl refers to the radical of the following ring: . The term [1,2,4]triazolo[4,3‐a]pyrimidyl refers to the radical of the following ring: . The term pyrazolo[1,5-b]pyridazinyl refers to the radical of the following ring: . The term [1,2,4]triazolo[1,5‐a]pyrimidinyl refers to the radical of the following ring: . The term 2H‐[1,2,3]triazolo[4,5‐b]pyridinyl refers to the radical of the following ring: . The term 1H‐[1,2,3]triazolo[4,5‐b]pyridinyl refers to the radical of the following ring: . The term 1H‐imidazo[4,5‐b]pyridinyl refers to the radical of the following ring: Many of the terms given above may be used repeatedly in the definition of a formula or group and in each case have one of the meanings given above, independently of one an- other. BIOLOGICAL ASSAYS Evaluation of inhibitory activity on QPCT and QPCTL Assay A: Biochemical QPCT and QPCTL Activity Assay The activity of the compounds of the invention may be demonstrated using the following biochemical enzyme activity assay: QPCT or QPCTL dependent conversion of N-terminal glutamine to pyroglutamate of CD47 was monitored via MALDI-TOF MS. Test compounds were dissolved in 100 % DMSO and serially diluted into clear 1,536-well microtiter plates. Enzymatic reactions were set up in assay buffer containing 20 mM Tris pH 7.5, 0.1 mM TCEP, 0.01% BSA, and 0.001% Tween20.2.5 µL of 2x concentrated QPCTL (in-house) or QPCT (Origine #TP700028) enzyme in assay buffer (0.5 nM final concentration, columns 1-23) or plain assay buffer (columns 24) were added to each well. The plates were incubated for 10 min in a humidified incubator at 24°C. Subsequently, 2.5 µL of CD47 peptide substrate surro- gate (19QLLFNKTKSVEFTFC33) was added to each well (final concentration: 10 µM for QPCTL / 20 µM for QPCT). The plates were mixed for 30 sec at 1,000 rpm and subse- quently incubated for 40 min in a humidified incubator at 24°C. After incubation, the enzy- matic reaction was stopped by adding 1 µL containing stable isotope labeled internal stand- ard peptide 19[Pyr]LLFN(K)TKSVEFTFC33 (final concentration 4.0 µM) as well as SEN177 (final concentration 10 µM). The plates were sealed with an adhesive foil, mixed for 30 s at 1,000 rpm and stored at room temperature until preparation of the MALDI tar- get plates. MALDI target plates were prepared as described previously.1 Mass spectra were acquired with a rapifleX MALDI-TOF/TOF instrument tracking the signals of the product (19[Pyr]LLFNKTKSVEFTFC33, m/z 1,787.9037) as well as internal standard (19[Pyr]LLFN(K)TKSVEFTFC33, m/z 1,795.9179) peptide. QPCT or QPCTL activity was monitored by calculating the ratio between product and internal standard signals followed by normalization to high (100% activity) and low (0% activity) controls. Determination of compound potencies was obtained by fitting the dose-response data to a four-parameter lo- gistical equation. Table 2: Biological data for compounds of the invention as obtained in Assay A. Table 3: Biological data for prior art compounds as obtained in Assay A. Assay B: SIRP ^ signalling assay (using either Raji or A549 cells) The activity of the compounds of the invention may be demonstrated using the following SIRP ^ signalling assay that measures SIRPα engagement induced by CD47 presented via cell-cell interaction. Two cell types are independently used: the Raji cell line (lymphoblast- like human cell line derived from B lymphocytes from a Burkitt’s lymphoma patient in 1963) and A549 cells (adenocarcinomic human alveolar basal epithelial cells). Test compounds were dissolved in 100 % DMSO and serially diluted into a white 384-well microtiter cell culture plate (PerkinElmer #60076780 in case of Raji assay; PDL-coated plates Greiner #781945 in case of A549 assay). 5000 Raji cells (ATCC #CC86) or 5000 A549 cells (ATCC #CCL-185) in Assay Complete Cell Plating reagent 30 (DiscoverX 93- 0563R30B) were added per well. The assay plate was incubated for 48 h at 37 °C, 95% humidity and 5 % CO 2 .15000 reporter cells (Jurkat PathHunter SIRPαV1, DiscoverX #93- 1135C19) were added to each well, and the plate was incubated for 5 h at 37 °C, 95 % humidity and 5 % CO2. Bioassay reagent 1 of the PathHunter Bioassay detection kit (Dis- coverX 93-0001) was added to each well of the plate using a multichannel pipette followed by a 15 min incubation at room temperature. Afterwards bioassay reagent 2 was added fol- lowed by 60 min incubation at room temperature (incubation in the dark). The analysis of the data was performed using the luminescence signal generated by beta- galactosidase in the PathHunter reporter cell line. The luminescence measurement was done using a Pherastar Multi-Mode Reader. Dose-response curves & IC50 data were calculated with 4-parameter sigmoidal dose response formula. Table 4: Biological data for compounds of the invention as obtained in Assay B. Table 5: Biological data for prior art compounds as obtained in Assay B. Evaluation of permeability Assay C: Permeability in CACO-2 cells Caco-2 cells (1 - 2 x 105 cells/1 cm2 area) are seeded on filter inserts (Costar transwell polycarbonate or PET filters, 0.4 μm pore size) and cultured (DMEM) for 10 to 25 days. Compounds are dissolved in appropriate solvent (like DMSO, 1 - 20 mM stock solutions). Stock solutions are diluted with HTP-4 buffer (128.13 mM NaCl, 5.36 mM KCl, 1 mM MgSO4, 1.8 mM CaCl2, 4.17 mM NaHCO3, 1.19 mM Na2HPO4 x 7H2O, 0.41 mM NaH 2 PO 4 xH 2 O, 15 mM HEPES, 20 mM glucose, 0.25% BSA, pH 7.2) to prepare the transport solutions (0.1 - 300 μM compound, final DMSO <= 0.5 %). The transport solution (TL) is applied to the apical or basolateral donor side for measuring A-B or B-A permeability (3 filter replicates), respectively. Samples are collected at the start and end of experiment from the donor and at various time intervals for up to 2 hours also from the receiver side for concentration measurement by HPLC-MS/MS or scintillation counting. Sampled receiver volumes are replaced with fresh receiver solution. Efflux ratio (ER) = permeability B-A / permeability A-B Table 7: Biological data for compounds of the invention as obtained in Assay C. Table 8: Biological data for prior art compounds as obtained in Assay C. Evaluation of Microsomal Clearance Microsomal clearance: The metabolic degradation of the test compound was assayed at 37 °C with pooled liver microsomes from various species. The final incubation volume of 60 μl per time point con- tains TRIS buffer pH 7.6 at room temperature (0.1 M), magnesium chloride (5 mM), micro- somal protein (1 mg/mL for human and dog, 0.5 mg/mL for other species) and the test com- pound at a final concentration of 1 μM. Following a short preincubation period at 37°C, the reactions were initiated by addition of betanicotinamide adenine dinucleotide phosphate, re- duced form (NADPH, 1 mM), and terminated by transferring an aliquot into solvent after different time points. After centrifugation (10000 g, 5 min), an aliquot of the supernatant was assayed by LC-MS/MS for the amount of parent compound. The half-life was deter- mined by the slope of the semi-logarithmic plot of the concentration-time profile. The intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of pro- tein in the incubation: CL_INTRINSIC [µl/min/mg protein] = (Ln 2 / (half-life [min] * protein content [mg/ml])) * 1000 CL_INTRINSIC_INVIVO [ml/min/kg] = (CL_INTRINSIC [µL/min/mg protein] x MPPGL [mg protein/g liver] x liver factor [g/kg bodyweight]) / 1000 Qh [%] = CL [ml/min/kg] / hepatic blood flow [ml/min/kg]) Hepatocellularity, human: 120x10e6 cells / g liver Liver factor, human: 25.7 g / kg bodyweight Blood flow, human: 21 ml/(min x kg) Evaluation of Hepatocyte Clearance Hepatocyte clearance The metabolic degradation of a test compound is assayed in a human hepatocyte suspen- sion. After recovery from cryopreservation, human hepatocytes are diluted in Dulbecco´s modified eagle medium (supplemented with 3.5 µg glucagon/500 mL, 2.5 mg insulin/500 mL, 3.75 mg hydrocortisone/500 mL, 5% human serum) to obtain a final cell density of 1.0x106 cells/mL. Following a 30 minutes preincubation in a cell culture incubator (37 °C, 10 % CO 2 ), test compound solution is spiked into the hepatocyte suspension, resulting in a final test com- pound concentration of 1 µM and a final DMSO concentration of 0.05 %. The cell suspension is incubated at 37 °C (cell culture incubator, horizontal shaker) and samples are removed from the incubation after 0, 0.5, 1, 2, 4 and 6 hours. Samples are quenched with acetonitrile (containing internal standard) and pelleted by centrifugation. The supernatant is transferred to a 96-deepwell plate, and prepared for analysis of decline of parent compound by HPLC-MS/MS. The percentage of remaining test compound is calculated using the peak area ratio (test compound/internal standard) of each incubation time point relative to the time point 0 peak area ratio. The log-transformed data are plotted versus incubation time, and the absolute value of the slope obtained by linear regression analysis is used to estimate in vitro half- life (T1/2). In vitro intrinsic clearance (CLint) is calculated from in vitro T1/2 and scaled to whole liver using a hepatocellularity of 120x106 cells/g liver, a human liver per body weight of 25.7 g liver/kg as well as in vitro incubation parameters, applying the following equation: CL_INTRINSIC_IN VIVO [mL/min/kg] = (CL_INTRINSIC [µL/min/106 cells] x hepato- cellularity [106 cells/g liver] x liver factor [g/kg body weight]) / 1000 Hepatic in vivo blood clearance (CL) is predicted according to the well-stirred liver model considering an average liver blood flow (QH) of 20.7 mL/min/kg: CL [mL/min/kg] = CL_INTRINSIC_IN VIVO [mL/min/kg] x hepatic blood flow [mL/min/kg] / (CL_INTRINSIC_IN VIVO [mL/min/kg] + hepatic blood flow [mL/min/kg]) Results are expressed as percentage of hepatic blood flow: QH [%] = CL [mL/min/kg] / hepatic blood flow [mL/min/kg]) Evaluation of plasma protein binding Equilibrium dialysis technique is used to determine the approximate in vitro fractional binding of test compounds to plasma proteins applying Dianorm Teflon dialysis cells (micro 0.2). Each dialysis cell consists of a donor and an acceptor chamber, separated by an ultrathin semipermeable membrane with a 5 kDa molecular weight cutoff. Stock solutions for each test compound are prepared in DMSO at 1 mM and serially diluted to obtain a final test concentration of 1 µM. The subsequent dialysis solutions are prepared in plasma (supplemented with NaEDTA as anticoagulant), and aliquots of 200 µl test compound dialysis solution in plasma are dispensed into the donor (plasma) chambers. Aliquots of 200 µl dialysis buffer (100 mM potassium phosphate, pH 7.4, supplemented with up to 4.7 % Dextran) are dispensed into the buffer (acceptor) chamber. Incubation is carried out for 2 hours under rotation at 37°C for establishing equilibrium. At the end of the dialysis period, aliquots obtained from donor and acceptor chambers, respectively, are transferred into reaction tubes and processed for HPLC-MS/MS analysis. Analyte concentrations are quantified in aliquots of samples by HPLC-MS/MS against calibration curves. Percent bound is calculated using the formula: %bound = (plasma concentration - buffer concentration/ plasma concentration) X 100 Evaluation of solubility Saturated solutions are prepared in well plates (format depends on robot) by adding an ap- propriate volume of selected aqueous media (typically in the range of 0.25 - 1.5 ml) into each well which contains a known quantity of solid drug substance (typically in the range 0.5 - 5.0 mg). The wells are shaken or stirred for a predefined time period (typically in a range of 2 - 24 h) and then filtered using appropriate filter membranes (typically PTFE-fil- ters with 0.45 µm pore size). Filter absorption is avoided by discarding the first few drops of filtrate. The amount of dissolved drug substance is determined by UV spectroscopy. In addition, the pH of the aqueous saturated solution is measured using a glass-electrode pH meter. Evaluation of Metabolism in human hepatocytes in vitro The metabolic pathway of a test compound is investigated using primary human hepatocytes in suspension. After recovery from cryopreservation, human hepatocytes are incubated in Dulbecco´s modified eagle medium containing 5% human serum and supplemented with 3.5 µg glucagon/500ml, 2.5mg insulin/500ml and 3.75mg/500ml hydrocortisone. Following a 30 min preincubation in a cell culture incubator (37°C, 10% CO2), test com- pound solution is spiked into the hepatocyte suspension to obtain a final cell density of 1.0*106 to 4.0*106 cells/ml (depending on the metabolic turnover rate of the compound ob- served with primary human hepatocytes), a final test compound concentration of 10 µM, and a final DMSO concentration of 0.05%. The cells are incubated for six hours in a cell culture incubator on a horizontal shaker, and samples are removed from the incubation after 0, 0.5, 1, 2, 4 or 6 hours, depending on the metabolic turnover rate. Samples are quenched with acetonitrile and pelleted by centrifuga- tion. The supernatant is transferred to a 96-deepwell plate, evaporated under nitrogen and resuspended prior to bioanalysis by liquid chromatography-high resolution mass spectrom- etry for identification of putative metabolites. The structures are assigned tentatively based on Fourier-Transform-MSn data. Metabolites are reported as percentage of the parent in human hepatocyte incubation with a threshold of ≥ 4%. Evaluation of pharmacokinetic characteristics The test compound is administered either intravenously or orally to the respective test spe- cies. Blood samples are taken at several time points post application of the test compound, anticoagulated and centrifuged. The concentration of analytes - the administered compound and/or metabolites - are quanti- fied in the plasma samples. PK parameters are calculated using non compartment methods. AUC and Cmax are normalized to a dose of 1 μmol/kg. METHOD OF TREATMENT The present invention is directed to compounds of general formula ( ^) which are useful in the prevention and/or treatment of a disease and/or condition associated with or modulated by QPCT/L activity, including but not limited to the treatment and/or prevention of cancer, fibrotic diseases, neurodegenerative diseases, atherosclerosis, infectious diseases, chronic kidney diseases. The compounds of general formula (I) are useful for the prevention and/or treatment of: (1) Pulmonary fibrotic diseases such as pneumonitis or interstitial pneumonitis associated with collagenosis, e g. lupus erythematodes, systemic scleroderma, rheumatoid arthritis, polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as pulmonary lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory bronchiolitis associated interstitial lung disease, desquamative interstitial pneumonia, cryptogenic orgainizing pneumonia, acute interstitial pneumonia and lymphocytic interstitial pneumonia, lymangi- oleiomyomatosis, pulmonary alveolar proteinosis, Langerhan's cell histiocytosis, pleural parenchymal fibroelastosis, interstitial lung diseases of known cause, such as interstitial pneumonitis as a result of occupational exposures such as asbestosis, silicosis, miners lung (coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds) or other occupa- tional airbourne triggers such as metal dust or mycobacteria, or as a result of treatment such as radiation, methotrexate, amiodarone, nitrofurantoin or chemotherapeutics, or for granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss syndrome, sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused by different origins, e g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or inter- stitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, M. boeck or sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, or alpha-I-antitrypsin defi- ciency. (2) Other fibrotic diseases such as hepatic bridging fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture, keloid, scleroderma/ systemic sclerosis, mediastinal fibrosis, myelotibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis; spontaneous acute exacerba- tions in pulmonary fibrosis and progressive pulmonary fibrosis or induced by infection, microaspiration, surgical lung biopsy, surgical resection, bronchoscopy (BAL, cryobi- opsy), air pollution, prior exacerbation and medications. (3) Leukemia, acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), T-cell acute lym- phoblastic leukemia (T-ALL), lymphoma, B-cell lymphoma, T-cell lymphoma, Hodgkin’s disease, non-Hodgkin’s lymphoma (NHL), hairy cell lymphoma, Burkett’s lymphoma, multiple myeloma (MM), myelodysplastic syndrome, solid cancer, lung cancer, adenocar- cinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), mediastinum cancer, peritoneal cancer, mesothelioma, gastrointestinal cancer, gastric cancer, stomach cancer, bowel cancer, small bowel cancer, large bowel cancer, colon cancer, colon adeno- carcinoma, colon adenoma, rectal cancer, colorectal cancer, leiomyosarcoma, breast can- cer, gynaecological cancer, genito-urinary cancer, ovarian cancer, endometrial cancer, cer- vical cancer, prostate cancer, testicular cancer, seminoma, teratocarcinoma, liver cancer, kidney cancer, bladder cancer, urothelial cancer, biliary tract cancer, pancreatic cancer, ex- ocrine pancreatic carcinoma, esophageal cancer, nasopharyngeal cancer, head and neck squamous cell carcinoma (HNSCC), skin cancer, squamous cancer, squamous cell carci- noma, Kaposi's sarcoma, melanoma, malignant melanoma, xeroderma pigmentosum, kera- toacanthoma, bone cancer, bone sarcoma, osteosarcoma, rhabdomyosarcoma, fibrosar- coma, thyroid gland cancer, thyroid follicular cancer, adrenal gland cancer, nervous system cancer, brain cancer, astrocytoma, neuroblastoma, glioma, schwannoma, glioblastoma, or sarcoma, gastrointestinal cancer, gastric cancer, stomach cancer, esophageal cancer, head and neck squamous cell carcinoma (HNSCC), breast cancer, colorectal cancer, bowel can- cer, large bowel cancer, colon cancer, colon adenocarcinoma, colon adenoma, rectal can- cer, ovarian cancer, pancreatic cancer, exocrine pancreatic carcinoma, leukemia, acute my- eloid leukemia (AML), myelodysplastic syndrome, lymphoma, B-cell lymphoma, non- Hodgkin’s lymphoma (NHL), urothelial cancer, or peritoneal cancer. (4) Inflammatory, auto-immune or allergic diseases and conditions such as asthma, pediat- ric asthma, allergic bronchitis, alveolitis, hyperreactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bron- chitis or pneumonitis, non-allergic asthma, chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, pulmonary emphysema; autoimmune diseases, such as rheumatoid arthritis, Graves’ disease, Sjogren's syndrome psoriatic arthritis, multiple scle- rosis, systemic lupus Erythematosus, inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis, scleroderma; psoriasis (including T-cell mediated psoriasis) and in- flammatory dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e g, necrotizing, cutaneous, and hypersensitivity vasculitis), or erythemanodosum. (5) Neurodegenerative disorders such as amyotrophic lateral sclerosis, multiple sclerosis, Parkinson's disease, Alzheimer's disease, Huntington's disease, multiple system atrophy, or prion diseases. Accordingly, the present invention relates to a compound of general formula (I) or a phar- maceutically acceptable salt thereof for use as a medicament. Furthermore, the present invention relates to the use of a compound of general formula (I) for the treatment and/or prevention of a disease and/or condition associated with or modu- lated by QPCT/L activity. Furthermore, the present invention relates to the use of a compound of general formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for the treatment and/or prevention of cancer, fibrotic diseases, neurodegenerative diseases, atherosclerosis, infectious diseases, chronic kidney diseases. Furthermore, the present invention relates to the use of a compound of general formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for the treatment and/or prevention of: (1) Pulmonary fibrotic diseases such as pneumonitis or interstitial pneumonitis associated with collagenosis, e g. lupus erythematodes, systemic scleroderma, rheumatoid arthritis, polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as pulmonary lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory bronchiolitis associated interstitial lung disease, desquamative interstitial pneu- monia, cryptogenic orgainizing pneumonia, acute interstitial pneumonia and lymphocytic interstitial pneumonia, lymangioleiomyomatosis, pulmonary alveolar proteinosis, Langer- han's cell histiocytosis, pleural parenchymal fibroelastosis, interstitial lung diseases of known cause, such as interstitial pneumonitis as a result of occupational exposures such as asbestosis, silicosis, miners lung (coal dust), farmers lung (hay and mould), Pidgeon fanci- ers lung (birds) or other occupational airbourne triggers such as metal dust or mycobacte- ria, or as a result of treatment such as radiation, methotrexate, amiodarone, nitrofurantoin or chemotherapeutics, or for granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss syndrome, sarcoidosis, hypersensitivity pneumonitis, or intersti- tial pneumonitis caused by different origins, e g. aspiration, inhalation of toxic gases, va- pors, bronchitis or pneumonitis or interstitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, M. boeck or sarcoidosis, granulomatosis, cystic fibrosis or muco- viscidosis, or alpha-I-antitrypsin deficiency. (2) Other fibrotic diseases such as hepatic bridging fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture, keloid, scleroderma/ systemic sclerosis, mediastinal fibrosis, myelotibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis; spontaneous acute exacerba- tions in pulmonary fibrosis and progressive pulmonary fibrosis or induced by infection, microaspiration, surgical lung biopsy, surgical resection, bronchoscopy (BAL, cryobi- opsy), air pollution, prior exacerbation and medications. (3) Leukemia, acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), T-cell acute lym- phoblastic leukemia (T-ALL), lymphoma, B-cell lymphoma, T-cell lymphoma, Hodgkin’s disease, non-Hodgkin’s lymphoma (NHL), hairy cell lymphoma, Burkett’s lymphoma, multiple myeloma (MM), myelodysplastic syndrome, solid cancer, lung cancer, adenocar- cinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), mediastinum cancer, peritoneal cancer, mesothelioma, gastrointestinal cancer, gastric cancer, stomach cancer, bowel cancer, small bowel cancer, large bowel cancer, colon cancer, colon adeno- carcinoma, colon adenoma, rectal cancer, colorectal cancer, leiomyosarcoma, breast can- cer, gynaecological cancer, genito-urinary cancer, ovarian cancer, endometrial cancer, cer- vical cancer, prostate cancer, testicular cancer, seminoma, teratocarcinoma, liver cancer, kidney cancer, bladder cancer, urothelial cancer, biliary tract cancer, pancreatic cancer, ex- ocrine pancreatic carcinoma, esophageal cancer, nasopharyngeal cancer, head and neck squamous cell carcinoma (HNSCC), skin cancer, squamous cancer, squamous cell carci- noma, Kaposi's sarcoma, melanoma, malignant melanoma, xeroderma pigmentosum, kera- toacanthoma, bone cancer, bone sarcoma, osteosarcoma, rhabdomyosarcoma, fibrosar- coma, thyroid gland cancer, thyroid follicular cancer, adrenal gland cancer, nervous system cancer, brain cancer, astrocytoma, neuroblastoma, glioma, schwannoma, glioblastoma, or sarcoma, gastrointestinal cancer, gastric cancer, stomach cancer, esophageal cancer, head and neck squamous cell carcinoma (HNSCC), breast cancer, colorectal cancer, bowel cancer, large bowel cancer, colon cancer, colon adenocarcinoma, colon adenoma, rectal cancer, ovarian cancer, pancreatic cancer, exocrine pancreatic carcinoma, leukemia, acute myeloid leukemia (AML), myelodysplastic syndrome, lymphoma, B-cell lymphoma, non- Hodgkin’s lymphoma (NHL), urothelial cancer, or peritoneal cancer. (4) Inflammatory, auto-immune or allergic diseases and conditions such as asthma, pediat- ric asthma, allergic bronchitis, alveolitis, hyperreactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bron- chitis or pneumonitis, non-allergic asthma, chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, pulmonary emphysema; autoimmune diseases, such as rheumatoid arthritis, Graves’ disease, Sjogren's syndrome psoriatic arthritis, multiple scle- rosis, systemic lupus Erythematosus, inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis, scleroderma; psoriasis (including T-cell mediated psoriasis) and in- flammatory dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e g, necrotizing, cutaneous, and hypersensitivity vasculitis), or erythemanodosum. (5) Neurodegenerative disorders such as amyotrophic lateral sclerosis, multiple sclerosis, Parkinson's disease, Alzheimer's disease, Huntington's disease, multiple system atrophy, or prion diseases. In a further aspect the present invention relates to a compound of general formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for use in the treatment and/or prevention of above-mentioned diseases and conditions. In a further aspect the present invention relates to the use of a compound of general for- mula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for the preparation of a medicament for the treatment and/or prevention of above- mentioned diseases and conditions. In a further aspect of the present invention the present invention relates to methods for the treatment or prevention of above-mentioned diseases and conditions, which method com- prises the administration of an effective amount of a compound of general formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof to a hu- man being. COMBINATION THERAPY The compounds of the invention may further be combined with one or more, preferably one additional therapeutic agent. According to one embodiment the additional therapeutic agent is selected from the group of therapeutic agents useful in the treatment of diseases or conditions described hereinbefore, in particular associated with cancer, fibrotic diseases, Alzheimer’s diseases, atherosclerosis, infectious diseases, chronic kidney diseases and auto-immune disease. Additional therapeutic agents that are suitable for such combinations include in particular those, which, for example, potentiate the therapeutic effect of one or more active sub- stances with respect to one of the indications mentioned and/or allow the dosage of one or more active substances to be reduced. Therefore, a compound of the invention may be combined with one or more additional therapeutic agents selected from the group consisting of chemotherapy, targeted cancer therapy, cancer immunotherapy, irradiation, antifibrotic agents, anti-tussive agents, anti- inflammatory agents, anti-atopic dermatitis, and broncho dilators. Chemotherapy is a type of cancer therapy that uses one or more chemical anti-cancer drugs, such as cytostatic or cytotoxic substances, cell proliferation inhibitors, anti-angio- genic substances and the like. Examples include folic acid (Leucovorin), 5-Fluorouracil, Irinotecan, Oxaliplatin, cis-platin Azacytidine, gemcitabine, alkylation agents, antimitotic agents, taxanes and further state-of-the-art or standard-of-care compounds. Targeted therapy is a type of cancer treatment that uses drugs to target specific genes and proteins that help cancer cells survive and grow. Targeted therapy includes agents such as inhibitors of growth factors (e.g. platelet derived growth factor (PDGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), insulin-like growth factors (IGF), human epidermal growth factor (HER, e.g. HER2, HER3, HER4) and hepatocyte growth factor), tyrosine-kinases, KRAS, BRAF, BCR-ABL, mTOR, cyclin-dependent kinases, or MDM2. Cancer immunotherapy is a type of therapy that uses substances to stimulate or suppress the immune system to help the body fight cancer. Cancer immunotherapy includes a thera- peutic antibody, such as: anti-Her2 antibody, an anti-EGFR antibody, and an anti-PDGFR antibody; an anti-GD2 (Ganglioside G2) antibody. Examples include Dinutuximab, Olaratumab, Trastuzumab, Pertuzumab, Ertumaxomab, Cetuximab, Necitumumab, Nimotuzumab, Panitumumab, or rituximab. Cancer immunotherapy also includes a thera- peutic antibody which is a checkpoint inhibitor, such as an anti PD1, anti PD-L1 antibody or CTLA4 inhibitor. Examples include Atezolizumab, Avelumab, and Durvalumab, Ipili- mumab, nivolumab, or pembrolizumab. Cancer immunotherapy also includes agents which target (inhibit) the CD47-SIRPα signaling axis, such as agents which bind to CD47 or SIRPα. Non-limiting examples include antibodies such as anti-CD47 antibodies and anti- SIRPα antibodies, and recombinant Fc-fusion proteins such as CD47-Fc and SIRPα-Fc. Cancer immunotherapy also includes STING-targeting agent, or T cell engagers, such as blinatumomab. Antifibrotic agents are for example nintedanib, pirfenidone, phosphodiesterase-IV (PDE4) inhibitors such as roflumilast or specific PDE4b inhibitors like BI 1015550, autotaxin in- hibitors such as GLPG-1690 or BBT-877; connective tissue growth factor (CTGF) block- ing antibodies such as Pamrevlumab; B-cell activating factor receptor (BAFF-R) blocking antibodies such as Lanalumab, alpha-V/beta-6 blocking inhibitors such as BG-00011/STX- 100, recombinant pentraxin-2 (PTX-2) such as PRM-151; c-Jun-N-terminal kinase (JNK) inhibitors such as CC-90001; galectin-3 inhibitors such as TD-139; G-protein coupled re- ceptor 84 (GPR84) inhibitors ; G-protein coupled receptor 84/ G-protein coupled receptor 40 dual inhibitors such asPBI-4050, Rho Associated Coiled-Coil Containing Protein Ki- nase 2 (ROCK2) inhibitors such as KD-025, heat shock protein 47 (HSP47) small interfer- ing RNA such as BMS-986263/ND-L02-s0201; Wnt pathway inhibitor such as SM-04646; LD4/ PDE3/4 inhibitors such as Tipelukast; recombinant immuno-modulatory domains of histidyl tRNA synthetase(HARS) such as ATYR-1923, prostaglandin synthase inhibitors such as ZL-2102 /SAR-191801; 15-hydroxy-eicosapentaenoic acid (15-HEPE e.g. DS- 102); Lysyl Oxidase Like 2 (LOXL2) inhibitors such as PAT-1251, PXS-5382/PXS-5338; phosphoinositide 3-kinases (PI3K)/ mammalian target of rapamycin (mTOR) dual inhibi- tors such as HEC-68498; calpain inhibitors such as BLD-2660; mitogen-activated protein kinase kinase kinase (MAP3K19) inhibitors such as MG-S-2525; chitinase inhibitors such as OATD-01,mitogen-activated protein kinase-activated protein kinase 2 (MAPKAPK2) inhibitors such as MMI-0100; transforming growth factor beta I (TGF-beta I) small inter- fering RNA such as TRKZSO/BNC-1021; or lysophosphatidic acid receptor antagonists such as BMS986278. The dosage for the combination partners mentioned above is usually 1/5 of the lowest dose normally recommended up to 1/1 of the normally recommended dose. Therefore, in another aspect, this invention relates to the use of a compound according to the invention in combination with one or more additional therapeutic agents described hereinbefore and hereinafter for the treatment of diseases or conditions which may be af- fected or which are mediated by QPCT/L, in particular diseases or conditions as described hereinbefore and hereinafter. In a further aspect this invention relates to a method for treating a disease or condition which can be influenced by the inhibition of QPCT/L in a patient that includes the step of administering to the patient in need of such treatment a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with a therapeutically effective amount of one or more additional therapeutic agents. In a further aspect this invention relates to the use of a compound of formula (I) or a phar- maceutically acceptable salt thereof in combination with one or more additional therapeu- tic agents for the treatment of diseases or conditions which can be influenced by the inhibi- tion of QPCT/L in a patient in need thereof. In yet another aspect the present invention relates to a method for the treatment of a disease or condition mediated by QPCT/L activity in a patient that includes the step of administer- ing to the patient, preferably a human, in need of such treatment a therapeutically effective amount of a compound of the present invention in combination with a therapeutically ef- fective amount of one or more additional therapeutic agents described in hereinbefore and hereinafter. The use of the compound according to the invention in combination with the additional therapeutic agent may take place simultaneously or at staggered times. The compound according to the invention and the one or more additional therapeutic agents may both be present together in one formulation, for example a tablet or capsule, or separately in two identical or different formulations, for example as a so-called kit-of-parts. Consequently, in another aspect, this invention relates to a pharmaceutical composition that comprises a compound according to the invention and one or more additional thera- peutic agents described hereinbefore and hereinafter, optionally together with one or more inert carriers and/or diluents. Other features and advantages of the present invention will become apparent from the fol- lowing more detailed examples which illustrate, by way of example, the principles of the invention. PREPARATION The compounds according to the present invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the literature of organic synthesis. Preferably, the compounds are obtained in analogous fashion to the methods of preparation explained more fully hereinafter, in particular as de- scribed in the experimental section. In some cases, the order in carrying out the reaction steps may be varied. Variants of the reaction methods that are known to the one skilled in the art but not described in detail here may also be used. The general processes for preparing the compounds according to the invention will become apparent to the one skilled in the art studying the following schemes. Any functional groups in the starting materials or intermediates may be protected using conventional pro- tecting groups. These protecting groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the one skilled in the art. The compounds according to the invention are prepared by the methods of synthesis de- scribed hereinafter in which the substituents of the general formulae have the meanings given herein before. These methods are intended as an illustration of the invention without restricting its subject matter and the scope of the compounds claimed to these examples. Where the preparation of starting compounds is not described, they are commercially ob- tainable or may be prepared analogously to known compounds or methods described herein. Substances described in the literature are prepared according to the published meth- ods of synthesis. Abbreviations are as defined in the Examples section. Examples 1 - 25 may be prepared as shown in Scheme I below. Scheme I:

In scheme I, N-Methyl triazolyl piperidine (Z=C-H, C-F, C-Me) (A) undergo a nucleophilic aromatic substitution with heteroaryl fluoride (one Y = N, one Y = CR; X= Cl, Br, I) (B). The reaction can typically be run at ambient temperature or at elevated temperature (up to 110 °C) in the presence of a base (e.g. diisopropylethylamine). The intermediate (C) is then subjected to a Suzuki-cross coupling with a hetero-aryl boronic acid derivative in the pres- ence of a suitable catalyst (e.g. Pd(dppf)Cl2) and a suitable base at elevated temperature (e.g. 100 °C) to afford compounds of general formula (I). Intermediates I may be prepared as shown in Scheme II below: Scheme II: Compounds of formula (A) with Z = C-F, C-Me, C-H can be prepared from the correspond- ing piperidinyl esters (R= Me, Et) (D) equipped with a suitable protecting group (PG, e.g. BOC) by treatment with a suitable hydrazine source (e.g. N 2 H 4 *H 2 O) at elevated tempera- ture (e.g.50 °C). The obtained hydrazide (E) is then activated with DMF/DMA at elevated temperature (e.g.50 °C) and subsequently treated with methyl amine at elevated temperature (e.g.90 °C) to yield the triazole derivative (F). Compounds of formula (A) can be obtained by cleaving the protecting group under suitable conditions (e.g. 4 M HCl in dioxane for PG=BOC). The obtained hydrochloride salts of (A) are treated with ammonia and passed through a Biotage SNAP Cartridge KP-NH column to release the free amine of (A). Com- pounds of formula (A) with Z = C-H and C-Me can also be obtained as hydrochlorides salts from commercial sources. Intermediates II may be prepared as shown in Scheme III below: Scheme III: In case of R=H, intermediates of formula (B) can be prepared from the corresponding car- boxylic acids (G). The carboxylic acid moiety is transformed into the corresponding amide (H) using a suitable combination of reagents, e.g.1,1’-carbonyldiimidazole and ammonia at ambient temperature. Compounds of formula (B) are subsequently obtained by treatment of (H) with a suitable dehydrating agent, e.g. Burgess reagent at ambient temperature. In case of R = Me or CF 3 , deprotonation of pyridine (J) at low temperature (e.g. –65 °C) and quench- ing with DMF yields the corresponding aldehydes (K). In case of R = Me, aldehyde (K) can be transformed directly into nitrile (B) by using ammonia in combination with a suitable oxidant (e.g. iodine) at ambient temperature. In case of R = CF 3 , aldehyde (K) can be trans- formed into the amide (H) using a suitable reagent, e.g. phenyltrimethylammonium tribro- mide, at ambient temperature and further into the nitrile (B) as described above for R=H. EXAMPLES Preparation The compounds according to the invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the literature of organic synthesis for example using methods described in “Comprehensive Organic Transformations”, 2nd Edition, Richard C. Larock, John Wiley & Sons, 2010, and “March’s Advanced Organic Chemistry”, 7th Edition, Michael B. Smith, John Wiley & Sons, 2013. Preferably the compounds are obtained analogously to the methods of prepara- tion explained more fully hereinafter, in particular as described in the experimental section. In some cases the sequence adopted in carrying out the reaction schemes may be varied. Variants of these reactions that are known to the skilled artisan but are not described in de- tail herein may also be used. The general processes for preparing the compounds according to the invention will become apparent to the skilled man on studying the schemes that fol- low. Starting compounds are commercially available or may be prepared by methods that are described in the literature or herein, or may be prepared in an analogous or similar manner. Before the reaction is carried out, any corresponding functional groups in the starting compounds may be protected using conventional protecting groups. These protect- ing groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the skilled man and described in the literature for example in “Protect- ing Groups”, 3rd Edition, Philip J. Kocienski, Thieme, 2005, and “Protective Groups in Organic Synthesis”, 4th Edition, Peter G. M. Wuts, Theodora W. Greene, John Wiley & Sons, 2006. The terms "ambient temperature" and "room temperature" are used inter- changeably and designate a temperature of about 20 °C, e.g. between 19 and 24 °C. Abbreviations:

Preparation of intermediates Intermediate I.1 tert‐Butyl 4‐fluoro‐4‐(hydrazinecarbonyl)piperidine‐1‐carboxy late 1‐tert‐Butyl 4‐ethyl 4‐fluoropiperidine‐1,4‐dicarboxylate (160 g, 0.58 mol) is suspended in ethanol (640 mL) in a round-bottom flask. Hydrazine hydrate (70.6 mL, 1.16 mol) is added to the mixture at ambient temperature. The reaction mixture is heated to 50 °C and stirred for 12 h. After cooling to ambient temperature, the mixture is concentrated under reduced pressure to yield tert‐butyl 4‐fluoro‐4‐(hydrazinecarbonyl)piperidine‐1‐carboxy late in 80% purity. C11H20FN3O3 (M = 261.3 g/mol) ESI-MS: 284.2 [M+Na]+ Rt (HPLC): 0.62 min (method A) tert‐Butyl 4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4‐triazol‐3‐yl )piperidine‐1‐carboxylate tert‐Butyl 4‐fluoro‐4‐(hydrazinecarbonyl)piperidine‐1‐carboxy late (135 g, 0.413 mol, 80% purity) is mixed with dioxane (945 mL) in a round-bottom-flask. N,N-Dimethylformamid- dimethylacetal (137 mL, 1.03 mol) is added to the mixture at ambient temperature. The re- action mixture is heated to 50 °C and stirred for 1 h. A solution of methylamine (299 g, 30% in EtOH, 2.89 mol) and acetic acid (165 mL, 2.89 mol) are added into the mixture. The resulting reaction mixture is heated to 90 °C and stirred for 11 h. The mixture is concentrated under reduced pressure. The residue is purified by column chromatography (SiO2, PE/EtOAc gradient 20:1 to 0:1) to obtain tert‐butyl 4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4‐triazol‐3‐yl )piperi- dine‐1‐carboxylate. C13H21FN4O2 (M=284.3 g/mol) ESI-MS: 285.1 [M+H]+ Rt (HPLC): 0.77 min (method A) Intermediate I.1: 4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4‐triazol‐3‐yl )piperidine tert‐Butyl 4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4‐triazol‐3‐yl )piperidine‐1‐carboxylate (90 g, 0.32 mol) is combined with methanol (90 mL) in a round-bottom flask. A solution of HCl (4 M in MeOH, 450 mL, 1.8 mol) is added slowly at ambient temperature. The resulting reaction mixture is stirred at ambient temperature for 12 h. The desired product is collected by filtra- tion, washed with methanol, and dried to yield 4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4‐triazol‐3‐ yl)piperidine hydrochloride salt. The hydrochloride salt (13.5 g) is added to a solution of ammonia in methanol (7 M , 150 mL) and purified by column chromatography (Biotage SNAP Cartridge KP-NH 110 g, gradient DCM/MeOH 4:1 to 7:3) C8H13FN4 (M=184.2 g/mol) ESI-MS: 185 [M+H]+ Rt (HPLC): 0.20 min (method B) 4-(4-Methyl-4H-1,2,4-triazol-3-yl)piperidine (MFCD09055373, CAS: 297172-18-0), 4-(4- methyl-4H-1,2,4-triazol-3-yl)piperidine hydrochloride (MFCD19440843), 1-(4-methyl- 4H-1,2,4-triazol-3-yl)piperazine (MFCD27979337, CAS: 67869-95-8), and 4-methyl-4-(4- methyl-4H-1,2,4-triazol-3-yl)piperidine dihydrochloride (MFCD32875324) are obtained from commercial vendors. Hydrochloride salts are converted into the free piperidine or pi- perazine according to the procedure described for Int.I Intermediate II.1 2‐Bromo‐3‐fluoropyridine‐4‐carboxamide 2‐Bromo‐3‐fluoropyridine‐4‐carboxylic acid (25.0 g, 0.114 mol) is suspended in THF (250 mL), and 1,1’-carbonyldiimazole (22.1 g, 0.136 mol) is added in several smaller portions. The reaction mixture is stirred for 3 h at ambient temperature, before a solution of ammonia (32% in H 2 O, 64.8 mL, 0.938 mol) is added slowly. The resulting reaction mixture is stirred for additional 3h at ambient temperature. The mixture is concentrated, the residue loaded onto Extrelut® and purified by column chromatography (SiO2, DCM/MeOH 9:1) to yield 2‐ bromo‐3‐fluoropyridine‐4‐carboxamide. C6H4BrFN2O (M=219.0 g/mol) ESI-MS: 219 / 221 [M+H]+ Rt (HPLC): 0.28 min (method C) Intermediate II.1: 2‐bromo‐3‐fluoropyridine‐4‐carbonitrile 2‐Bromo‐3‐fluoropyridine‐4‐carboxamide (17.4 g, 79.5 mmol) is added to dichloromethane (250 mL). Burgess reagent (CAS: 29684-56-8, 22.0 g, 89.5 mmol) is added, and the resulting reaction mixture is stirred for 16 h at ambient temperature. The mixture is concentrated to half of the original volume and purified by column chromatography (SiO2, DCM) to yield 2‐bromo‐3‐fluoropyridine‐4‐carbonitrile. C6H2BrFN2 (M=201.0 g/mol) ESI-MS: no mass detected Rt (HPLC): 0.41 min (method F) 1H NMR (400 MHz, DMSO-d6) δ ppm 8.52 (d, J=4.9 Hz, 1 H), 8.05 (t, J=4.8 Hz, 1 H). Intermediate II.2 2‐Chloro-3‐fluoropyridine‐4‐carboxamide 2‐Chloro-3‐fluoropyridine‐4‐carboxylic acid (1.00 g, 5.41 mmol) is suspended in THF (25 mL), and 1,1’-carbonyldiimazole (1.05 g, 6.49 mmol) is added in several smaller portions. The reaction mixture is stirred for 2 h at ambient temperature, before a solution of ammonia (32% in H 2 O, 3.09 mL, 44.6 mmol) is added slowly. The resulting reaction mixture is stirred for additional 2 h at ambient temperature. The mixture is concentrated, the residue loaded onto Extrelut® and purified by column chromatography (SiO2, CyH/EtOAc gradient 1:0 to 1:1) to yield 2‐chloro‐3‐fluoropyridine‐4‐carboxamide. C 6 H 4 ClFN 2 O (M=174.6 g/mol) ESI-MS: 174 [M−H]− Rt (HPLC): 0.11 min (method B) Intermediate II.2: 2‐chloro‐3‐fluoropyridine‐4‐carbonitrile 2‐Chloro-3‐fluoropyridine‐4‐carboxamide (748 mg, 4.29 mmol) is added to DCM (20 mL). Burgess reagent (CAS: 29684-56-8, 1.26 g, 3.64 mmol) is added, and the resulting reaction mixture is stirred for 16 h at ambient temperature. The mixture is concentrated to half of the original volume and purified by column chromatography (SiO2, CyH/EtOAc gradient 1:0 to 7:3) to yield 2‐chloro‐3‐fluoropyridine‐4‐carbonitrile. C 6 H 2 ClFN 2 (M=156.5 g/mol) ESI-MS: no mass detected Rt (HPLC): 0.41 min (method C) 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.53 (d, J=4.9 Hz, 1 H), 8.06 (t, J=4.7 Hz, 1 H) Intermediate II.3 2‐Bromo‐3‐fluoro‐6‐methylpyridine‐4‐carbaldehy de Under an argon atmosphere, 2‐bromo‐3‐fluoro‐6‐methylpyridine (1.00 g, 5.16 mmol) is added to THF (50 mL), and the resulting mixture is cooled to −65 °C. A solution of lithium diisopropylamide (1 M in THF, 5.42 mL, 5.42 mmol) is added dropwise and the mixture stirred for another 30 min at -65 °C. A solution of DMF (476 µL DMF in 3 mL THF, 6.19 mmol) is added dropwise. The mixture is stirred for 20 min at -65 °C and subsequently al- lowed to warm to ambient temperature. The reaction is quenched by addition of an aqueous saturated ammonium chloride solution and Me-THF. The phases are separated, the organic phase is washed with water, dried over MgSO 4 and concentrated to yield 2‐bromo‐3‐fluoro‐ 6‐methylpyridine‐4‐carbaldehyde. C7H5BrFNO (M=218.0 g/mol) ESI-MS: no mass detected Rt (HPLC): 0.41 min (method C) 1H NMR (400 MHz, DMSO-d6) δ ppm 10.16 (s, 1 H), 7.64 (d, J=4.6 Hz, 1 H), 2.53 (d, J=1.0 Hz, 3 H) Intermediate II.3: 2‐bromo‐3‐fluoro‐6‐methylpyridine‐4‐carbonitri le 2‐Bromo‐3‐fluoro‐6‐methylpyridine‐4‐carbaldehy de (460 mg, 2.11 mmol) is added to THF (4.6 mL) and a solution of ammonia (28% in H 2 O, 4.6 mL) and iodine (580 mg, 2.28 mmol) are added. The resulting reaction mixture is stirred for 5 h at ambient temperature. The reac- tion mixture is diluted with ethyl acetate and washed twice with saturated aqueous NH4Cl solution. The organic phase is concentrated, loaded onto Extrelut® and purified by column chromatography (SiO2, CyH/EtOAc gradient 1:0 to 4:1) to yield 2‐bromo‐3‐fluoro‐6‐ methylpyridine‐4‐carbonitrile. C 7 H 4 BrFN 2 (M=215.0 g/mol) ESI-MS: 215 / 217 [M+H]+ Rt (HPLC): 0.40 min (method C) Intermediate II.4 2‐Bromo‐3‐fluoro‐6‐(trifluoromethyl)pyridine‐4 carbaldehyde Under an argon atmosphere, 2‐bromo‐3‐fluoro‐6‐(trifluoromethyl)pyridine (1.00 g, 4.1 mmol) is added to THF (25 mL), and the resulting mixture is cooled to −70 °C. A solution of lithium diisopropylamide (1 M in THF, 4.51 mL, 4.51 mmol) is added dropwise, and the mixture is stirred for 45 min at −70 °C. DMF (0.378 mL, 4.92 mmol) is added dropwise. The mixture is stirred for additional 30 min at −70 °C. The reaction is quenched by addition of acetic acid (800 µL) and diluted with water and ethyl acetate. The organic phase is separated, dried over MgSO4, and concentrated. The residue is purified by column chromatography (SiO 2 , CyH/EtOAc gradient 1:0 to 4:1) to yield 2‐bromo‐3‐fluoro‐6‐(trifluoromethyl)pyri- dine‐4‐carbaldehyde. C7H2BrF4NO (M=272.0 g/mol) ESI-MS: no mass detected Rt (HPLC): 0.48 min (method E) 1H NMR (400 MHz, DMSO-d6) δ ppm 10.19 (s, 1 H), 8.26 (d, J=4.3 Hz, 1 H). 2‐Bromo‐3‐fluoro‐6‐(trifluoromethyl)pyridine‐4 carboxamide Ammonium acetate (5.53 g, 71.8 mmol) and 2‐bromo‐3‐fluoro‐6‐(trifluoromethyl)pyridine‐ 4‐carbaldehyde (2.17 g, 7.18 mmol) are mixed, and acetonitrile (44 mL) is added. Phenyl- trimethylammonium tribromide (5.57 g, 14.4 mmol) is added in small portions, and the re- sulting reaction mixture is stirred for 72 h at ambient temperature. The mixture is filtered, and the residue is washed with acetonitrile, purified by column chromatography (dry load with Celite®, SiO 2 , CyH/EtOAc gradient 1:0 to 7:3) to yield the desired product. C 7 H 3 BrF 4 N 2 O (M=287.0 g/mol) ESI-MS: 285 / 287 [M−H]− Rt (HPLC): 0.50 min (method B) Intermediate II.4: 2‐bromo‐3‐fluoro‐6‐(trifluoromethyl)pyridine‐4 carbonitrile Product of the previous step, 2‐bromo‐3‐fluoro‐6‐(trifluoromethyl)pyridine‐4 carboxamide (975 mg, 3.40 mmol) is suspended in dichloromethane (80 mL), and Burgess reagent (CAS: 29684-56-8, 1.25 g, 5.10 mmol) is added at ambient temperature. The resulting re- action mixture is stirred for 40 h and then directly purified by column chromatography (dry load with Celite®, SiO 2 , CyH/EtOAc gradient 1:0 to 9:1) to yield the desired product. C 7 HBrF 4 N 2 (M=268.9 g/mol) ESI-MS: 268 / 270 [M+H]+ Rt (HPLC): 0.59 min (method C) General procedure for the synthesis of Intermediates III 2‐Bromo‐3‐[4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4 triazol‐3‐yl)piperidin‐1‐yl]pyridine‐4‐car- bonitrile Int. II.1 (500 mg, 2.49 mmol) and Int. I.1 (687 mg, 3.73 mmol) are suspended in DMSO (5.0 mL), and DIPEA (861 µL, 4.98 mmol) is added at ambient temperature. The resulting mix- ture is stirred for 72 h at ambient temperature. The mixture is diluted with water (0.1 mL), and the precipitate is collected by filtration, washed with water, and dried to yield the desired product 2‐bromo‐3‐[4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4 triazol‐3‐yl)piperidin‐1‐yl]pyridine‐4‐ carbonitrile, which is used in the next step without further purification. C 14 H 14 BrFN 6 (M=365.2 g/mol) ESI-MS: 365 / 367 [M+H]+ Rt (HPLC): 0.42 min (method C) Intermediate IV.1 4-(4-Methyl-4H-1,2,4-triazol-3-yl)piperidine hydrochloride (180 mg, 0.89 mmol), 3-fluoro- 4-iodoopicolinonitrile (200 mg, 0.81 mmol) and potassium carbonate (334 mg, 2.42 mmol) are suspended in DMF (4.0 mL), and the resulting reaction mixture is heated to 90°C and stirred for 18 h. After cooling to ambient temperature, the mixture is concentrated, redis- solved in a mixture of acetonitrile, water, and methanol, filtered, and purified by preparative HPLC (XBridge C18, acetonitrile/water gradient containing 0.1% NH 3 ) to yield the desired product along with a by-product resulting from iodine replacement. C14H15IN6 (M=394.2 g/mol) ESI-MS: 395 [M+H]+ Rt (HPLC): 0.38 min (method C) Intermediate V.1 5-Bromo-2-tert-butyl-2H-pyrazolo[3,4-b]pyridine 5-Bromo-1H-pyrazolo[3,4-b]pyridine (4.0 g, 19.8 mmol) is suspended in toluene (23 mL), and tert-butyl acetate (26.6 mL, 198 mmol) is added. Methanesulfonic acid (1.3 mL, 19.8 mmol) is added slowly. The resulting reaction mixture is heated to 80 °C and stirred for 1 h. After cooling to ambient temperature, additional methanesulfonic acid (1.3 mL, 19.8 mmol) is added, and the reaction mixture is heated to 80 °C and stirred for 1 h. After cooling to ambient temperature, the reaction mixture is concentrated and purified by preparative HPLC (XBridge C18, ACN/water gradient containing 0.1% TFA) to yield the 5-bromo-2-tert-bu- tyl-2H-pyrazolo[3,4-b]pyridine. C10H12BrN3 (M=254.1 g/mol) ESI-MS: 254 / 256 [M+H]+ Rt (HPLC): 0.50 min (method C) Intermediate V.1 5-Bromo-2-tert-butyl-2H-pyrazolo[3,4-b]pyridine (1.50 g, 3.87 mmol), bis(pinacoloto)di- boron (1.21 g, 4.78 mmol), and potassium acetate (763 mg, 7.77 mmol) are added to 1,4- dioxane (15 mL), and the resulting mixture is degassed by passing an argon stream through the mixture for 10 min. [1,1′-Bis-(diphenylphosphino)-ferrocen]-dichloro-palladium (II) di- chloromethane complex (Pd(dppf)Cl2*CH2Cl2, CAS: 95464-05-4) (190 mg, 0.232 mmol) is added, and the mixture is degassed for additional 3 min. The mixture is then heated to 110 °C and stirred at this temperature for 4 h. After cooling to ambient temperature, the mixture is concentrated, and the residue is dissolved in a ACN/water mixture, filtered, and purified by preparative HPLC (XBridge C18, ACN/water gradient containing 0.1% TFA) to yield {2‐tert‐butyl‐2H‐pyrazolo[3,4‐b]pyridin‐5‐yl}b oronic acid. C 10 H 14 BN 3 O 2 (M=219.1 g/mol) ESI-MS: 220 [M+H]+ Rt (HPLC): 0.27 min (method C) Intermediate V.2 6‐Bromo‐3‐tert‐butyl‐[1,2,4]triazolo[4,3‐a]pyrim idine 5-Bromo-2-hydrazinopyrimidine (200 mg, 1.01 mmol) and pivaloyl chloride (985 mg, 8.09 mmol) are added into a microwave vial, which is subsequently sealed. The mixture is heated to 120 °C and stirred at this temperature for 22 h. Water and ACN are added, and the mixture is concentrated. The residue is taken up with ACN and purified by preparative HPLC (XBridge C18, ACN/water gradient containing 0.1% TFA) to yield the desired compound. C9H11BrN4 (M=255.1 g/mol) ESI-MS: 255 / 257 [M+H]+ Rt (HPLC): 0.46 min (method C) Intermediate V.2 6‐Bromo‐3‐tert‐butyl‐[1,2,4]triazolo[4,3‐a]pyrim idine (75.0 mg, 0.294 mmol), bis-(pinaco- lato)-diboron (100 mg, 0.394 mmol), and potassium acetate (90.0 mg, 0.917 mmol) are added to 1,4-dioxane (1.5 mL). The mixture is degassed by passing an argon stream through the mixture for 15 min. Bis(triphenylphosphine)palladium(II) dichloride (CAS: 13965-03- 2; 20.4 mg, 0.029 mmol) is added, and the mixture is further degassed for 3 min. The result- ing reaction mixture is heated to 60 °C and stirred at this temperature for 20 h. After cooling to ambient temperature, the mixture is diluted with EtOAc and filtered. The filtrate is con- centrated and taken up with a mixture of ACN, water, and TFA. It is then purified by pre- parative HPLC (Sunfire C18, ACN/water gradient containing 0.1% TFA) to yield the desired compound. C9H13BN4O2 (M=220.0 g/mol) ESI-MS: 221 [M+H]+ Rt (HPLC): 0.34 min (method C) Intermediate V.3 6‐Bromo‐2‐tert‐butylimidazo[1,2‐a]pyrimidine 2-Amino-5-bromopyrimidine (250 mg, 1.41 mmol) and 1-chloropinacolone (285 µL, 2.11 mmol) are added to ethanol (2.0 mL), and the resulting reaction mixture is stirred for 96 h at 90 °C. After cooling to ambient temperature, the mixture is diluted with ACN and purified by column chromatography (SiO 2 , DCM/MeOH gradient 1:0 to 9:1) to yield the desired product. C10H12BrN3 (M=254.1 g/mol) ESI-MS: 254 / 256 [M+H]+ Rt (HPLC): 0.28 min (method C) Intermediate V.3 6‐Bromo‐2‐tert‐butylimidazo[1,2‐a]pyrimidine (144 mg, 0.567 mmol), bis-(pinacolato)- diboron (215 mg, 0.850 mmol), and potassium acetate (167 mg, 1.70 mmol) are added to 1,4-dioxane (1.0 mL). The mixture is degassed by passing an argon stream through the mixture for 10 min. Bis(triphenylphosphine)palladium(II) dichloride (CAS: 13965-03-2; 39.8 mg, 56.7 µmol) is added, and the mixture is further degassed for 3 min. The resulting reaction mixture is heated to 90 °C and stirred at this temperature for 5 h. After cooling to ambient temperature, the mixture is concentrated. The residue is taken up with a mixture of water and ACN and then purified by preparative HPLC (XBridge C18, ACN/water gradi- ent containing 0.1% TFA) to yield the desired compound. C10H14BN3O2 (M=219.1 g/mol) ESI-MS: 220 [M+H]+ Rt (HPLC): 0.25 min (method C) Intermediate V.4 6‐Bromo‐2‐(trifluoromethyl)imidazo[1,2‐a]pyrimidine 2-Amino-5-bromopyrimidine (1.0 g, 5.6 mmol) and 1-chloro-3,3,3-trifluoroacetone (889 µL, 8.45 mmol) are added to ethanol (8 mL), and the resulting reaction mixture is stirred for 120 h at 90 °C. After cooling to ambient temperature, the mixture is concentrated and loaded onto Extrelut®. It is then purified by column chromatography (SiO 2 , DCM/MeOH gradient 1:0 to 9:1) to yield the desired product. C7H3BrF3N3 (M=266.0 g/mol) ESI-MS: 266 / 268 [M+H]+ Rt (HPLC): 0.38 min (method C) Intermediate V.4 6‐Bromo‐2‐(trifluoromethyl)imidazo[1,2‐a]pyrimidine (373 mg, 1.40 mmol), bis-(pinaco- lato)-diboron (215 mg, 1.54 mmol), and potassium acetate (412 mg, 4.21 mmol) are added to 1,4-dioxane (1.0 mL). The mixture is degassed by passing an argon stream through the mixture for 10 min. Bis(triphenylphosphine)palladium(II) dichloride (98.4 mg, 0.140 mmol) is added, and the mixture is further degassed for 3 min. The resulting reaction mixture is heated to 90 °C and stirred at this temperature for 5 h. After cooling to ambient temperature, the mixture is concentrated. The residue is taken up with a mixture of water and ACN and purified by preparative HPLC (XBridge C18, ACN/water gradient containing 0.1% TFA) to yield the desired compound. C7H5BF3N3O2 (M=230.9 g/mol) ESI-MS: 232 [M+H]+ Rt (HPLC): 0.29 min (method C) Intermediate VI.1 5‐Bromo‐2‐fluoro‐3‐{[2‐(trimethylsilyl)ethoxy]me thoxy}pyridine 5-Bromo-2-fluoropyridin-3-ol (3.00 g, 15.3 mmol) is added to DCM (40 mL), and DIPEA (5.3 mL, 31 mmol) is added.2-(Trimethylsilyl)ethoxymethyl chloride (3.15 mL, 16.8 mmol) is added dropwise, and the resulting reaction mixture is stirred for 90 min at ambient tem- perature. The mixture is concentrated, loaded onto Extrelut®, and purified by column chro- matography (SiO2, CyH/EtOAc 1:0 to 3:1) to yield the desired product. C11H17BrFNO2Si (M=322.2 g/mol) ESI-MS: 322 / 324 [M+H]+ Rt (HPLC): 0.84 min (method C) (6‐Fluoro‐5‐{[2‐(trimethylsilyl)ethoxy]methoxy}pyrid in‐3‐yl)boronic acid Under an argon atmosphere, 1,4-dioxane (30 mL) is added to a mixture of 5‐bromo‐2‐fluoro‐ 3‐{[2‐(trimethylsilyl)ethoxy]methoxy}pyridine (4.80 g, 14.9 mmol), bis(pinacolato)diboron (3.50 g, 13.8 mmol), and potassium acetate (4.39 g, 44.7 mmol). The suspension is degassed by passing an Argon stream through the mixture for 10 min. [1,1′-Bis-(diphenylphosphino)- ferrocen]-dichloro-palladium(II) dichloromethane complex (Pd(dppf)Cl2*CH2Cl2, CAS: 95464-05-4) (545 mg, 0.745 mmol) is added, and the mixture is further degassed for 3 min. The mixture is then heated to 80 °C and stirred at this temperature for 18 h. After cooling to ambient temperature, the mixture is diluted with EtOAc and water. The organic phase is separated, dried over MgSO4, and concentrated. The residue is purified by preparative HPLC (XBridge C18, ACN/water gradient) to yield the desired product in 80% purity. C 11 H 19 BFNO 4 Si (M=287.2 g/mol) ESI-MS: 288 [M+H]+ Rt (HPLC): 0.60 min (method C) 6'‐Fluoro‐3‐[4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4 ‐triazol‐3‐yl)piperidin‐1‐yl]‐5'‐{[2‐(trime- thylsilyl)ethoxy]methoxy}‐[2,3'‐bipyridine]‐4‐carbon itrile Under an argon atmosphere, 1,4-dioxane (3.0 mL) is added to a mixture of intermediate III.1 (211 mg, 0.578 mmol), (6‐fluoro‐5‐{[2‐(trimethylsilyl)ethoxy]methoxy}pyrid in‐3‐yl)bo- ronic acid (80% purity, 519 mg, 1.44 mmol), and tetrakis(triphenylphosphine)palladium(0) (66.8 mg, 0.058 mmol). The suspension is degassed by passing an Argon stream through the mixture for 10 min. A solution of cesium carbonate (2 M in H2O, 867 µL, 1.73 mmol) is added, and the mixture is again degassed for 3 min. The reaction mixture is then heated to 80 °C and stirred at this temperature for 8 h. After cooling to ambient temperature, the mix- ture is concentrated. The residue is taken up in ACN and purified by preparative HPLC (XBridge C18, ACN/water gradient containing 0.1% TFA) to yield the desired product. C 25 H 31 F 2 N 7 O 2 Si (M=527.6 g/mol) ESI-MS: 528 [M+H]+ Rt (HPLC): 0.67 min (method C) Intermediate VI.1 6'‐Fluoro‐3‐[4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4 ‐triazol‐3‐yl)piperidin‐1‐yl]‐5'‐{[2‐(trimet hylsi- lyl)ethoxy]methoxy}‐[2,3'‐bipyridine]‐4‐carbonitrile (290 mg, 0.45 mmol) is added to di- chloromethane and a solution of hydrochlorid acid (4 M in dioxane, 563 µL, 2.25 mmol) is added. The resulting reaction mixture is stirred for 2 h at ambient temperature. The mixture is diluted with MTBE, and the precipitated solid is collected by filtration, washed with MTBE, and dried to yield the desired product. C 19 H 17 F 2 N 7 O (M=397.4 g/mol) ESI-MS: 398 [M+H]+ Rt (HPLC): 0.39 min (method C) Intermediate VI.2 6'‐Fluoro‐3‐[4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4 ‐triazol‐3‐yl)piperidin‐1‐yl]‐5'‐formyl‐ [2,3'‐bipyridine]‐4‐carbonitrile Under an argon atmosphere, intermediate III.1 (1.0 g, 2.74 mmol), 2-fluoro-5-(4,4,5,5-tetra- methyl-1,3,2-dioxaborolan-2-yl)nicotinaldehyde (CAS: 1333319-63-3; 796 mg, 3.01 mmol) , and a solution of potassium carbonate (2 M in water, 2.74 mL, 5.48 mmol) are added to 1,4- dioxane (30 mL). The mixture is degassed by passing an argon stream through the mixture for 10 min. Then, [1,1′-Bis-(diphenylphosphino)-ferrocen]-dichloro-palladium (II) (Pd(dppf)Cl2, CAS: 72287-26-4) (200 mg, 0.274 mmol) is added, and the mixture is further degassed for 3 min. The reaction mixture is then heated to 100 °C and stirred at this temper- ature for 1 h. After cooling to ambient temperature, the mixture is diluted with EtOAc and water. The organic phase is separated, dried over sodium sulfate, and concentrated. The res- idue is taken up with DMF and purified by preparative HPLC (ZORBAX StableBond C18, ACN/water gradient containing 0.1% TFA) to yield the desired product. C20H17F2N7O (M=409.4 g/mol) ESI-MS: 410 [M+H]+ Rt (HPLC): 0.73 min (method G) Intermediate VI.2 6'‐Fluoro‐3‐[4‐fluoro‐4‐(4‐methyl‐4H‐1,2,4 ‐triazol‐3‐yl)piperidin‐1‐yl]‐5'‐formyl‐[2,3 '‐bi- pyridine]‐4‐carbonitrile (660 mg, 1.61 mmol) and tetrabutylammonium iodide (59.5 mg, 0.161 mmol) are added to DMSO (20 mL). Sodium azide (212 mg, 3.22 mmol) is then added in small portions at ambient temperature. The resulting reaction mixture is stirred for 1 h at ambient temperature before it is diluted with an aqueous half saturated sodium chloride so- lution. The organic phase is extracted with EtOAc (3x), and the combined organic layers are dried over sodium sulfate and concentrated to yield the desired product that was used in the next step without further purification. C 20 H 17 FN 10 O (M=432.4 g/mol) ESI-MS: 433 [M+H]+ Rt (HPLC): 0.66 min (method G) Preparation of Final Compounds Example 1

Under an argon atmosphere, intermediate III.1 (1.43 g, 3.92 mmol) and 2-fluoropyridine-5- boronic acid pinacol ester (873 mg, 3.92 mmol) are suspended in 1,4-dioxane (15 mL), and a solution of potassium carbonate (2 M in water, 3.9 mL, 7.8 mmol) is added. The resulting mixture is degassed by passing an argon stream through the solution for 15 min. [1,1′-Bis- (diphenylphosphino)-ferrocen]-dichloro-palladium(II) (Pd(dppf)Cl2, CAS: 72287-26-4) (286 mg, 0.392 mmol) is added, and the mixture is further degassed for 3 min. The reaction mixture is heated to 100 °C and stirred for 5 h. After cooling to ambient temperature, it is diluted with ethyl acetate and filtered over Celite®. The filtrate is concentrated and purified via preparative HPLC (XBridge C18 column, ACN/water gradient containing 0.1% NH 3 ) to yield the desired compound. C19H17F2N7 (M=381.4 g/mol) ESI-MS: 382 [M+H]+ Rt (HPLC): 0.40 min (method C) 1H NMR (400 MHz, DMSO-d6) δ ppm: 8.54 (d, J=4.9 Hz, 1 H), 8.50 (d, J=2.3 Hz, 1 H), 8.48 (s, 1 H), 8.25 (td, J=8.2, 2.5 Hz, 1 H), 7.82 (d, J=4.8 Hz, 1 H), 7.34 (dd, J=8.4, 2.6 Hz, 1 H), 3.73 (d, J=1.6 Hz, 3 H), 3.15 - 3.28 (m, 4 H), 2.09 - 2.27 (m, 4 H) Examples synthesized analogous to the procedure described for example 1 Analytical data of synthesized examples , d, , H), Hz, 01 - - , 3 H), .93 4.9 , (s, 2 H), , , H) d, 7 - d, (s, 31 H), , , , ) H), s, 7, d, (d, (br ), ) .54 1.6 m, Hz, 74 17 - H), d, (s, Hz, Hz, Hz, 4 H), H), H), d, , H), Hz, 18 - - - ), ), , z, br 4 d, d, ), z, 6 , ), ), ), d, - 9 , , , , , , J 9 , = 6 , ) , , , , , - ), 15 ), -,d, - ),),), ,d,d, ),), ),), ,d, , , , Example 26 The intermediate VI.1 (90.0 mg, 0.207 mmol) and cesium carbonate (135.2 mg, 0.414 mmol) are added to DMF (2.0 mL) and (iodomethyl)cyclopropane (56.6 mg, 0.311 mmol) is added. The resulting reaction mixture is heated to 80 °C and stirred at this temperature for 8 h. After cooling to ambient temperature, it is diluted with a mixture of ACN/water/TFA and purified by preparative HPLC (XBridge C18, ACN/water gradient containing 0.1% TFA) to yield the desired product. C23H23F2N7O (M=451.5 g/mol) ESI-MS: 452 [M+H]+ Rt (HPLC): 0.52 min (method C) 1H NMR (400 MHz, DMSO-d6) δ ppm 8.53 (s, 1 H), 8.51 - 8.53 (m, 1 H), 8.52 (d, J=4.8 Hz, 1 H), 7.94 (t, J=1.6 Hz, 1 H), 7.88 (dd, J=10.1, 1.8 Hz, 1 H), 7.82 (d, J=4.9 Hz, 1 H), 3.99 (d, J=7.1 Hz, 2 H), 3.74 (d, J=1.6 Hz, 3 H), 3.15 - 3.32 (m, 4 H), 2.09 - 2.30 (m, 4 H), 1.20 - 1.33 (m, 1 H), 0.52 - 0.65 (m, 1 H), 0.30 - 0.42 (m, 2 H). Examples synthesized analogous to the procedure described for example 26 Analytical data of synthesized examples Example 29 Under an argon atmosphere, intermediate VI.2 (20.0 mg, 0.046 mmol) and iron(II) bromide (2.0 mg, 9.5 µmol) are suspended in DMSO (1.0 mL). 1,1,1-Trifluoro-2-methyl-propan-2- amine hydrochloride (11.3 mg, 0.069 mmol) is added, and the resulting reaction mixture is heated to 120 °C and stirred at this temperature for 2 h. After cooling to ambient temperature, the mixture is diluted with water and purified by preparative HPLC (ZORBAX StableBond C18, water/ACN gradient containing 0.1% TFA) to yield the desired product. C 24 H 23 F 4 N 9 (M=513.5 g/mol) ESI-MS: 514 [M+H]+ Rt (HPLC): 0.65 min (method L) 1H NMR (400 MHz, DMSO-d6) δ ppm 8.95 (s, 1 H), 8.92 (d, J=2.2 Hz, 1 H), 8.56 (d, J=4.8 Hz, 1 H), 8.49 (s, 1 H), 8.41 (d, J=2.2 Hz, 1 H), 7.81 (d, J=4.8 Hz, 1 H), 3.71 (d, J=1.3 Hz, 3 H), 3.23 - 3.28 (m, 4 H), 2.15 - 2.30 (m, 4 H), 2.03 (s, 6 H). Examples synthesized analogous to the procedure described for example 29 Analytical data of synthesized examples m , 4.8 1 H), , , 3 - 0 2 , 8 2 8 4 2 , 3 - H) , 4.9 38 (d, 22 4 4 , 2 Analytical HPLC methods Method A Analytical column: Kinetex EVO C18_2.1 x 30 mm_5 μm; column temperature: 40°C Method B Device description: Waters Acquity; Analytical column: XBridge (Waters) BEH C18_2.1 x 30 mm_2.5µm; column temperature: 60°C Method C Device description: Waters Acquity; Analytical column: Xbridge (Waters) BEH C18_2.1 x 30 mm_1.7µm; column temperature: 60°C Method D Device description: Waters Acquity; Analytical column: Xbridge (Waters) C18_3.0 x 30 mm_2.5 µm; column temperature: 60°C Method E Device description: Waters Acquity; Analytical column: Xbridge (Waters) BEH Phe- nyl_2.1 x 30 mm_1.7µm; column temperature: 60°C Method F Device description: Agilent 1200; Analytical column: XBridge (Waters) C18_3.0 x 30 mm_2.5μm; column temperature: 60°C Method G Device description: Agilent 1200; Analytical column: Zorbax (Agilent) StableBond C18_3.0 x 30 mm_1.8 µm; column temperature: 60 °C Method H Device description: Waters Acquity; Analytical column: Sunfire (Waters) C18_3.0 x 30 mm_2.5 µm; column temperature: 60°C Method I Device description: Waters Acquity; Analytical column: XBridge (Waters) C18_3.0 x 30 mm_2.5 µm; column temperature: 60 °C Method J Device description: Waters Acquity; Analytical column: Sunfire (Waters) C18_3.0 x 30 mm_2.5 µm; column temperature: 60°C Method K Device description: Agilent 1200; Analytical column: Xbridge (Waters) C18_3.0 x 30 mm_2.5 µm; column temperature: 60°C Method L Device description: Waters Acquity; Analytical column: Sunfire (Waters) C18_3.0 x 30 mm_2.5 μm; column temperature: 60°C Method M Device description: Agilent 1260 Infinity II SFC; Column: CHIRAL ART (YMC) Cellu- lose SC_4.6 x 250 mm_1.7μm; column temperature: 40°C