Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED BICYCLES AS HSET INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2024/042007
Kind Code:
A1
Abstract:
The invention relates to substituted bicycles of the general formula I, and the use of the compounds of the present invention for the treatment and/or prevention of hyperproliferative diseases and disorders in mammals, especially humans, and pharmaceutical compositions containing such compound.

Inventors:
BUCHSTALLER HANS-PETER (DE)
KUHN DANIEL (DE)
COLLINS IAN (GB)
MATTHEWS THOMAS (GB)
LANZ JAN (GB)
SAINT-DIZIER FRANÇOIS (GB)
PREVET HUGUES (GB)
Application Number:
PCT/EP2023/072876
Publication Date:
February 29, 2024
Filing Date:
August 21, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK PATENT GMBH (DE)
International Classes:
C07D403/12; A61K31/4985; A61P35/00; C07D413/12; C07D413/14; C07D471/04; C07D487/04; C07D495/04
Domestic Patent References:
WO2007056469A22007-05-18
WO2006031348A22006-03-23
WO2023131690A12023-07-13
Download PDF:
Claims:
Claims

1. Compound of the formula I,

R1 denotes NO2, COOA, A, OA, NHA, NHCOA, CONHA, CONA2, COA or R4,

R2 denotes

R3 denotes H or A,

R4 denotes H, unsubsubstitued or with unbranched or branched alkyl with 1-

4 C-atoms substituted oxadiazolyl, tetrazolyl, pyrazolyl, oxazolyl or isoxazoyl, A denotes unbranched or branched alkyl or cycloalkyl with 1-10 C-atoms, wherein two adjacent CH- and/or CH2-groups may form a double bond and wherein one or two non-adjacent CH- and/or CH2-groups may be replaced by N-, O- and/or S-atoms and wherein 1-7 H-atoms may be replaced by F or Cl,

Hal denotes F, Cl, Br or I and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to claim 1 , wherein

W denotes and R1, R2, R3, R4 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to claim 1 or 2, wherein

W denotes and R1, R2, R3, R4 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to one or more of the preceding claims, wherein R1 denotes COCA, OA or R4, and W, R2, R3, R4 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to one or more of the preceding claims, wherein R1 denotes COCA and W, R2, R3, R4 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to one or more of the preceding claims, wherein R2 denotes and W, R1, R3, R4 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to one or more of the preceding claims, wherein R2 denotes and W, R1, R3, R4 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to one or more of the preceding claims, wherein R4 denotes oxadiazolyl, oxazolyl or tetrazolyl and W, R1, R2, R3 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound according to one or more of the preceding claims, wherein R4 denotes oxadiazolyl and W, R1, R2, R3 and A have the meanings as in Claim 1 , and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Compound selected from the group consisting of: and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Pharmaceutical preparation comprising at least one compound according to one or more of claims 1 to 10 and/or physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. Pharmaceutical preparation according to claim 11 comprising further excipients and/or adjuvants. Pharmaceutical preparation comprising at least one compound according to one or more of claims 1 to 12 and/or physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active compound. Process for the preparation of a pharmaceutical preparation, characterised in that a compound according to one or more of claims 1 to 10 and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, is brought into a suitable dosage form together with a solid, liquid or semi-liquid excipient or adjuvant. Medicament comprising at least one compound according to one or more of claims 1 to 10 and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis of physiological and/or pathophysiological states. Medicament comprising at least one compound according to one or more of claims 1 to 10 and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis of physiological and/or pathophysiological states, selected from the group consisting of hyperproliferative diseases and disorders. Medicament for use according to claim 16, wherein the hyperproliferative disease or disorder is cancer. Medicament for use according to claim 17, wherein the cancer is selected from the group consisting of acute and chronic lymphocytic leukemia, acute granulocytic leukemia, adrenal cortex cancer, bladder cancer, brain cancer, breast cancer, cervical hyperplasia, cervical cancer, chorio cancer, chronic granulocytic leukemia, chronic lymphocytic leukemia, colon cancer, endometrial ccancer, esophageal cancer, essential thrombocytosis, genitourinary carcinoma, glioma, glioblastoma, hairy cell leukemia, head and neck carcinoma, Hodgkin's disease, Kaposi's sarcoma, lung carcinoma, lymphoma, malignant carcinoid carcinoma, malignant hypercalcemia, malignant melanoma, malignant pancreatic insulinoma, medullary thyroid carcinoma, melanoma, multiple myeloma, mycosis fungoides, myeloid and lymphocytic leukemia, neuroblastoma, non-Hodgkin's lymphoma, non-small cell lung cancer, osteogenic sarcoma, ovarian carcinoma, pancreatic carcinoma, polycythemia vera, primary brain carcinoma, primary macroglobulinemia, prostatic cancer, renal cell cancer, rhabdomyosarcoma, skin cancer, small-cell lung cancer, soft-tissue sarcoma, squamous cell cancer, stomach cancer, testicular cancer, thyroid cancer and Wilms' tumor. Set (kit) consisting of separate packs of a) an effective amount of a compound according to one or more of claims 1 to 10 and/or physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios, and b) an effective amount of a further medicament active compound.

Description:
Substituted bicycles as HSET Inhibitors

The invention relates to substituted bicycles of the general formula I,

R 1

\ , - R2 | and the use of the compounds of the present invention for the treatment and/or prevention of hyperproliferative diseases and disorders such as cancer in mammals, especially humans, and pharmaceutical compositions containing such compounds.

Background of the invention

DNA replication, followed by equal chromosome segregation, ensures the accurate transmission of the genetic information to daughter cells (Hall et al., 2003; Nigg, 2002; Zyss and Gergely, 2009). In most normal and malignant cells, centrosomes act as the dominant sites for spindle pole formation (Meunier and Vernos, 2012). Centrosome duplication is also tightly controlled and occurs simultaneously with DNA replication, thereby ensuring the generation of two functional centrosomes that form the poles of the mitotic spindle (Sharp et al., 2000). In the assembly of a functional mitotic spindle, microtubule (MT) motor proteins play a central role (Cai et al., 2010; Ganem and Compton, 2004). One such protein, HSET (encoded by KIFC1 in humans and Kifc5a in mice), a minus-end MT motor, is of interest in cancer due to its impact on cell division (Cai et al., 2010; Goshima et al., 2005).

In recent years, the importance of centrosomes, and in particular HSET, for bipolar spindle formation has attracted much attention, although the precise role of HSET in this process remains a topic for debate (Mahoney et al., 2006; Tillement et al., 2009). Recent reports have linked centrosome amplification and high HSET expression to chromosome missegregation and aneuploidy, which are hallmarks of human cancer (Marx et al., 2009). Centrosome amplification disrupts asymmetric cell division in neuroblastoma cells and causes tumorigenesis in a fly model (Basto et al., 2008), and supernumerary centrosomes are also found in most solid tumor types, forming markers for aggressiveness in breast, brain, prostate, cervix, kidney, and bladder cancers (Chan, 2011). Hence, it is increasingly apparent that supernumerary centrosomes are not only indicative of malignancy but may also drive malignant transformation (Ogden et al., 2013). However, not all cells with centrosome amplification undergo multipolar mitosis, and a key mechanism by which cells with extra centrosomes achieve a pseudo-bipolar spindle is centrosome clustering (Basto et al., 2008; Ganem et al., 2009).

Although centrosome clustering prevents multipolar mitosis and cell death, it prolongs mitosis and increases the frequency of chromosome missegregation as a result of merotelic kinetochore attachments (Ganem et al., 2009; Kwon et al., 2008; Yang et al., 2008). Based on previous studies, centrosome clustering may prove to be the Achilles heel of cancer cells with supernumerary centrosomes (Basto et al., 2008), and a growing body of evidence suggests that inhibition of centrosome clustering could provide a new therapeutic strategy for tumors with a high incidence of centrosome amplification (Jordan and Wilson, 2004; Ogden et al., 2012).

A key protein that is known to be crucial for centrosome clustering is HSET (Ned in flies). HSET is required by tumour cells to cluster supernumerary centrosomes (Basto et al., 2008; Kwon et al., 2008). HSET is a member of the Kinesin 14 family of MT motor proteins, which are force-generating enzymes that facilitate movement along MTs within the cell (Mountain et al., 1999) and which transport organelles, protein complexes and mRNAs along microtubules in an ATP-dependent fashion. HSET is a minus-end directed motor kinesin, that cross-links and slides microtubules exerting inward forces (Walczak et al., 1997; Cai et al., 2009; Rath et al., 2012). Although the precise role of HSET in cell division is not clear, previous evidence suggests that it is essential for the survival of cancer, but not normal, cells (Ganem et al., 2009; Kwon et al., 2008). High HSET expression levels are strongly correlated with metastasis of non-small cell lung cancer to the brain, pointing to an association between HSET, centrosome amplification, and tumorigenesis (Cai et al., 2010; Gordon et al., 2001 ; Grinberg-Rashi et al., 2009). Knockdown of HSET in normal retinal pigment epithelial 1 (RPE-1) cells or the breast cancer cell line MCF-7 (which does not have a high incidence of centrosome amplification) does not inhibit bipolar spindle formation, and cells undergo normal division (Kleylein-Sohn et al., 2012; Kwon et al., 2008). In contrast, knockdown of HSET in the supernumerary centrosome-containing breast cancer and neuroblastoma cell lines MDA-MB-231 and N1 E-115, respectively, prevents centrosome clustering and induces cell death by multipolar anaphases (Kwon et al., 2008). Hence, the above findings point to HSET as a target of interest in cancer treatment (Basto et al., 2008; Kraljevic Pavelic et al., 2011; Kramer et al., 2011; Kwon et al., 2008).

A number of studies have shown that HSET depletion increases cell death and the frequency of multipolarity in cells with supernumerary centrosomes, but not in cells with a normal number of centrosomes. For example, HSET depletion induces spindle multi-polarity and selectively sensitizes centrosome amplified ER- breast cancer cell lines, including triple negative breast cancer (TNBC), to cell death (Patel et al., 2018). Depletion of HSET was identified as inducing selective cytotoxicity in centrosome amplified cancer cells (Drosopoulos et al., 2014). In addition, HSET overexpression has been correlated with poor prognosis and resistance to docetaxel in breast cancer (De et al., 2009; Li et al., 2015), is observed in ovarian adenocarcinoma patients (Pawar et al., 2014) and in numerous other cancer types (Pannu et al., 2015). Furthermore, in non-small cell lung carcinoma (NSCLC) HSET expression was found to be highly predictive of the presence of brain metastasis in both early and advanced disease (Grinberg-Rashi et al., 2009).

A wide range of tumours, including centrosome amplified tumours, are treated by cytotoxic microtubule-targeted drugs (e.g. taxol, eribulin). Although inducing temporary remission, these drugs typically show severe side effects and the emergence of drug resistance leading to early relapse. More recently, agents targeting kinesin motor proteins, e.g. Eg5 inhibitors, have been explored to treat a variety of human tumours, which induce mono-polar spindles (the opposite phenotype to HSET inhibition), and target all rapidly dividing cells, including bone marrow cells. Consequently, they share dose-limiting toxicities with other antimitotic therapies. In contrast, an HSET inhibitor is anticipated to show reduced toxicity by selectively killing cells with centrosome amplification whereas cells with the normal number of centrosomes will remain unaffected (Ganem et al., 2009; Patel et al., 2015). These data together provide support for developing agents that selectively inhibit HSET to target centrosome-amplified tumours (Myers and Collins, 2016). Examples of small molecule HSET inhibitors have been described in the literature. AZ82 is an ADP/ATP competitive inhibitor shown to be selective against a panel of nine other kinesins including Eg5 (Wu et al., 2013). AZ82 inhibited microtubule- stimulated HSET ATPase activity (IC50 = 0.3 pM) in a biochemical assay and induced multipolar spindle formation and mitotic catastrophe in cells with amplified centrosomes. CW069 was an inhibitor of HSET (IC50 = 75 pM) in biochemical assays (Watts et al., 2013). SR31527, also showed biochemical inhibition of HSET (IC50 = 6.6 pM) (Zhang et al., 2016). More information can be found in W009155025 and W015085088.

Thus, there remains a need for therapies for the treatment and prevention of hyperproliferative diseases and disorders such as cancer. Therefore, the aim was to find HSET inhibitors that serve as potential therapeutics for the treatment of cancer diseases.

Summary of the invention

Surprisingly, it has been found that the compounds according to the invention are highly selective and effective inhibitors of HSET and thus the compounds of the present invention can be used for the treament of hyperproliferative diseases and disorders such as cancer.

The invention relates to the compounds of the general formula I,

R 1

\ , - R2 | wherein

W denotes

30

R 1 denotes NO 2 , COCA, A, OA, NHA, NHCOA, CONHA, CONA 2 , COA or R 4 ,

R 2 denotes

R 3 denotes H or A,

R 4 denotes H, unsubsubstitued or with unbranched or branched alkyl with 1-4 C- atoms substituted oxadiazolyl, tetrazolyl, pyrazolyl, oxazolyl or isoxazoyl,

A denotes unbranched or branched alkyl or cycloalkyl with 1-10 C-atoms, wherein two adjacent CH- and/or CH2-groups may form a double bond and wherein one or two non-adjacent CH- and/or CH2-groups may be replaced by N-, O- and/or S-atoms and wherein 1-7 H-atoms may be replaced by F or Cl, Hal denotes F, Cl, Br or I and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein

W denotes and R 1 , R 2 , R 3 , R 4 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein

W denotes and R 1 , R 2 , R 3 , R 4 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein

R 1 denotes COOA, OA or R 4 , and W, R 2 , R 3 , R 4 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein

R 1 denotes COOA and W, R 2 , R 3 , R 4 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein

R 2 denotes and W, R 1 , R 3 , R 4 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein

R 2 denotes and W, R 1 , R 3 , R 4 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios. A preferred embodiment of the present invention are compounds according to formula I, wherein R 4 denotes oxadiazolyl, oxazolyl or tetrazolyl and W, R 1 , R 2 , R 3 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

A preferred embodiment of the present invention are compounds according to formula I, wherein R 4 denotes oxadiazolyl and W, R 1 , R 2 , R 3 and A have the meanings as disclosed above, and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

The invention preferably relates to a compound selected from the group consisting of:

and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

Furthermore, the abbreviations below have the following meanings:

Boc ter-butoxycarbonyl

CBZ benzyloxycarbonyl

DNP 2,4-dinitrophenyl

FMOC 9-fluorenylmethoxycarbonyl imi-DNP 2,4-dinitrophenyl in the 1-position of the imidazole ring

OMe methyl ester

POA phenoxyacetyl

DCCI dicyclohexylcarbodiimide

HOBt 1-hydroxybenzotriazole

The invention further relates to a pharmaceutical preparation comprising one or more compounds according to the present invention and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios.

The invention also relates to a pharmaceutical preparation according to the invention of this type, comprising further excipients and/or adjuvants.

In addition, the invention relates to an above pharmaceutical preparation according to the invention, comprising at least one further medicament active compound.

Pharmaceutically or physiologically acceptable derivatives are taken to mean, for example, salts of the compounds of the present invention, and also so-called pro- drug compounds. Prodrug compounds are taken to mean derivatives of the compounds of the present invention which have been modified by means of, for example, alkyl or acyl groups (see also amino- and hydroxyl-protecting groups below), sugars or oligopeptides and which are rapidly cleaved or liberated in the organism to form the effective molecules. These also include biodegradable polymer derivatives of the compound of the present invention, as described, for example, in Int. J. Pharm. 115 (1995), 61-67.

The compound of the present invention can be used in its final non-salt form. On the other hand, the present invention also encompasses the use of the compound of the present invention in the form of its pharmaceutically acceptable salts, which can be derived from various organic and inorganic bases by procedures known in the art. Pharmaceutically acceptable salt forms of the compound of the present invention are for the most part prepared by conventional methods. If the compound of the present invention contains a carboxyl group, one of its suitable salts can be formed by reacting the compound of the present invention ith a suitable base to give the corresponding base-addition salt. Such bases are, for example, alkali metal hydroxides, including potassium hydroxide, sodium hydroxide and lithium hydroxide; alkaline-earth metal hydroxides, such as barium hydroxide and calcium hydroxide; alkali metal alkoxides, for example potassium ethoxide and sodium propoxide; and various organic bases, such as piperidine, diethanolamine and N-methylglutamine. The aluminium salts of the compound of the present invetion are likewise included.

Furthermore, the base salts of the compounds of the present invention include aluminium, ammonium, calcium, copper, iron(lll), iron(ll), lithium, magnesium, man- ganese(lll), manganese(ll), potassium, sodium and zinc salts, but this is not intended to represent a restriction.

Of the above-mentioned salts, preference is given to ammonium; the alkali metal salts sodium and potassium, and the alkaline-earth metal salts calcium and magnesium. Salts of the compounds of the present invention which are derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines, also including naturally occurring substituted amines, cyclic amines, and basic ion exchanger resins, for example arginine, betaine, caffeine, chloroprocaine, choline, N,N'-dibenzylethylenediamine (benzathine), dicyclohexylamine, diethanolamine, diethylamine, 2-diethylamino- ethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triethylamine, trimethylamine, tripropylamine and tris- (hydroxymethyl)methylamine (tromethamine), but this is not intended to represent a restriction.

As mentioned, the pharmaceutically acceptable base-addition salts of the compound of the present invention are formed with metals or amines, such as alkali metals and alkaline-earth metals or organic amines. Preferred metals are sodium, potassium, magnesium and calcium. Preferred organic amines are N,N’-dibenzylethylene- diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methyl-D- glucamine and procaine.

The base-addition salts of the compounds of the present invention are prepared by bringing the free acid form into contact with a sufficient amount of the desired base, causing the formation of the salt in a conventional manner. The free acid can be regenerated by bringing the salt form into contact with an acid and isolating the free acid in a conventional manner. The free acid forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free acid forms thereof.

In view of that stated above, it can be seen that the term “pharmaceutically acceptable salt” in the present connection is taken to mean an active compound which comprises the compound of the present invention in the form of one of its salts, in particular if this salt form imparts improved pharmacokinetic properties on the active compound compared with the free form of the active compound or any other salt form of the active compound used earlier. The pharmaceutically acceptable salt form of the active compound can also provide this active compound for the first time with a desired pharmacokinetic property which it did not have earlier and can even have a positive influence on the pharmacodynamics of this active compound with respect to its therapeutic efficacy in the body. Solvates of the compound of the present invention are taken to mean adductions of inert solvent molecules of the compound of the present invention which form owing to their mutual attractive force. Solvates are, for example, hydrates, such as monohydrates or dihydrates, or alcoholates, i.e. addition compounds with alcohols, such as, for example, with methanol or ethanol.

All physiologically acceptable salts, derivatives, solvates and stereoisomers of these compounds, including mixtures thereof in all ratios, are also in accordance with the invention.

Compounds of the present invention may contain one or more centres of chirality, so that all stereoisomers, enantiomers, diastereomers, etc., of the compounds of the present inventionare also claimed in the present invention.

The invention also relates to the optically active forms (stereoisomers), the enantiomers, the racemates, the diastereomers and hydrates and solvates of these compounds.

Compounds of the present invention according to the invention may be chiral owing to their molecular structure and may accordingly occur in various enantiomeric forms. They may therefore be in racemic or optically active form. Since the pharmaceutical efficacy of the racemates or stereoisomers of the compounds according to the invention may differ, it may be desirable to use the enantiomers. In these cases, the end product, but also even the intermediates, may be separated into enantiomeric compounds by chemical or physical measures known to the person skilled in the art or already employed as such in the synthesis.

Pharmaceutically or physiologically acceptable derivatives are taken to mean, for example, salts of the compounds according to the invention and also so-called prodrug compounds. Prodrug compounds are taken to mean compounds of the present invention which have been modified with, for example, alkyl or acyl groups (see also amino- and hydroxyl-protecting groups below), sugars or oligopeptides and which are rapidly cleaved or liberated in the organism to form the effective compounds according to the invention. These also include biodegradable polymer derivatives of the compounds according to the invention, as described, for example, in Int. J. Pharm. 115 (1995), 61-67.

Suitable acid-addition salts are inorganic or organic salts of all physiologically or pharmacologically acceptable acids, for example halides, in particular hydrochlorides or hydrobromides, lactates, sulfates, citrates, tartrates, maleates, fumarates, oxalates, acetates, phosphates, methylsulfonates or p-toluenesulfonates.

Very particular preference is given to the hydrochlorides, the trifluoroacetates or the bistrifluoroacetates of the compounds according to the invention.

Solvates of the compounds of the present invention are taken to mean adductions of inert solvent molecules onto the compounds of the present invention which form owing to their mutual attractive force. Solvates are, for example, hydrates, such as monohydrates or dihydrates, or alcoholates, i.e. addition compounds with alcohols, such as, for example, with methanol or ethanol.

It is furthermore intended that a compound of the present invention includes isotopelabelled forms thereof. An isotope-labelled form of a compound of the present inventionis identical to this compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally. Examples of isotopes which are readily commercially available, and which can be incorporated into a compound of the present invention by well-known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 0, 31 P, 32 P, 35 S, 18 F and 36 CI, respectively. A compound of the present invention, a prodrug thereof or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other isotopes of other atoms is intended to be part of the present invention. An isotope-labelled compound of the present invention can be used in a number of beneficial ways. For example, an isotope-labelled compound of the present invention into which, for example, a radioisotope, such as 3 H or 14 C, has been incorporated is suitable for medicament and/or substrate tissue distribution assays. These radioisotopes, i.e. tritium ( 3 H) and carbon-14 ( 14 C), are particularly preferred owing to their simple preparation and excellent detectability. Incorporation of heavier isotopes, for example deuterium ( 2 H), into a compound of the present invention has therapeutic advantages owing to the higher metabolic stability of this isotope-labelled compound. Higher metabolic stability translates directly into an increased in-vivo half-life or lower dosages, which under most circumstances would represent a preferred embodiment of the present invention. An isotope-labelled compound of the present invention can usually be prepared by carrying out the procedures disclosed in the synthesis schemes and the related description, in the example part and in the preparation part in the present text, replacing a non-isotope-labelled reactant with a readily available isotope-labelled reactant.

In order to manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect, deuterium ( 2 H) can also be incorporated into a compound of the present invention. The primary kinetic isotope effect is a change in the rate of a chemical reaction that results from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange. Exchange of a heavier isotope usually results in a lowering of the ground state energy for a chemical bond and thus causes a reduction in the rate in rate-limiting bond breakage. If the bond breakage occurs in or in the vicinity of a saddle-point region along the coordinate of a multi-product reaction, the product distribution ratios can be altered substantially. For explanation: if deuterium is bonded to a carbon atom in a non-exchangeable position, rate differences of kM/ko = 2-7 are typical. If this rate difference is successfully applied to a compound of the present invention that is susceptible to oxidation, the profile of this compound in vivo can thereby be drastically modified and result in improved pharmacokinetic properties.

When discovering and developing therapeutic agents, the person skilled in the art attempts to optimise pharmacokinetic parameters while retaining desirable in-vitro properties. It is reasonable to assume that many compounds with poor pharmacokinetic profiles are susceptible to oxidative metabolism. In-vitro liver microsomal assays currently available provide valuable information on the course of oxidative metabolism of this type, which in turn permits the rational design of deuterated compounds of the present invention with improved stability through resistance to such oxidative metabolism. Significant improvements in the pharmacokinetic profiles of the compounds of the present invention are thereby obtained and can be expressed quantitatively in terms of increases in the in-vivo half-life (T1/2), concentration at maximum therapeutic effect (Cmax), area under the dose response curve (AUC), and F; and in terms of reduced clearance, dose and costs of materials.

The following is intended to illustrate the above: a compound of the present invention which has multiple potential sites of attack for oxidative metabolism, for example benzylic hydrogen atoms and hydrogen atoms bonded to a nitrogen atom, is prepared as a series of analogues in which various combinations of hydrogen atoms are replaced by deuterium atoms, so that some, most or all of these hydrogen atoms have been replaced by deuterium atoms. Half-life determinations enable favourable and accurate determination of the extent to which the improvement in resistance to oxidative metabolism has improved. In this way, it is determined that the half-life of the parent compound can be extended by up to 100% as the result of deuterium-hydrogen exchange of this type.

The replacement of hydrogen by deuterium in a compound of the present inventioncan also be used to achieve a favourable modification of the metabolite spectrum of the starting compound in order to diminish or eliminate undesired toxic metabolites. For example, if a toxic metabolite arises through oxidative carbonhydrogen (C-H) bond cleavage, it can reasonably be assumed that the deuterated analogue will greatly diminish or eliminate production of the undesired metabolite, even if the particular oxidation is not a rate-determining step. Further information on the state of the art with respect to deuterium-hydrogen exchange is given, for example in Hanzlik et al., J. Org. Chem. 55, 3992-3997, 1990, Reider et al., J. Org. Chem. 52, 3326-3334, 1987, Foster, Adv. Drug Res. 14, 1-40, 1985, Gillette et al., Biochemistry 33(10), 2927-2937, 1994, and Jarman et al., Carcinogenesis 16(4), 683-688, 1993.

The invention also relates to mixtures of the compounds of the present invention according to the invention, for example mixtures of two diastereomers, for example in the ratio 1:1, 1:2, 1 :3, 1 :4, 1:5, 1:10, 1:100 or 1:1000. These are particularly preferably mixtures of two stereoisomeric compounds. However, preference is also given to mixtures of two or more compounds of the present invention. In addition, the invention relates to a process for the preparation of the compounds of the present invention, characterized in that a) the base of a compound of the present invention is converted into one of its salts by treatment with an acid, or b) an acid of a compound of the present invention is converted into one of its salts by treatment with a base.

It is also possible to carry out the reactions stepwise in each case and to modify the sequence of the linking reactions of the building blocks with adaptation of the protecting-group concept.

The starting materials or starting compounds are generally known. If they are novel, they can be prepared by methods known per se.

If desired, the starting materials can also be formed in situ by not isolating them from the reaction mixture, but instead immediately converting them further into the compounds of the present invention.

The compounds of the present invention are preferably obtained by liberating them from their functional derivatives by solvolysis, in particular by hydrolysis, or by hydrogenolysis. Preferred starting materials for the solvolysis or hydrogenolysis are those which contain correspondingly protected amino, carboxyl and/or hydroxyl groups instead of one or more free amino, carboxyl and/or hydroxyl groups, preferably those which carry an amino-protecting group instead of an H atom which is connected to an N atom. Preference is furthermore given to starting materials which carry a hydroxyl-protecting group instead of the H atom of a hydroxyl group. Preference is also given to starting materials which carry a protected carboxyl group instead of a free carboxyl group. It is also possible for a plurality of identical or different protected amino, carboxyl and/or hydroxyl groups to be present in the molecule of the starting material. If the protecting groups present are different from one another, they can in many cases be cleaved off selectively.

The term “amino-protecting group” is generally known and relates to groups which are suitable for protecting (blocking) an amino group against chemical reactions, but which can easily be removed after the desired chemical reaction has been carried out elsewhere in the molecule. Typical of such groups are, in particular, unsubstituted or substituted acyl groups, furthermore unsubstituted or substituted aryl (for example 2,4-dinitophenyl) or aralkyl groups (for example benzyl, 4- nitrobenzyl, triphenylmethyl). Since the amino-protecting groups are removed after the desired reaction or reaction sequence, their type and size are, in addition, not crucial, but preference is given to those having 1-20, in particular 1-8, C atoms. The term “acyl group” is to be understood in the broadest sense in connection with the present process. It encompasses acyl groups derived from aliphatic, araliphatic, aromatic or heterocyclic carboxylic acids or sulfonic acids and, in particular, alkoxycarbonyl, aryloxycarbonyl and especially aralkoxycarbonyl groups. Examples of such acyl groups are alkanoyl, such as acteyl, propionyl, buturyl, aralkanoyl, such as phenylacetyl, aroyl, such as benzoyl or toluyl, aryoxyaklkanoyl, such as phenoxyacetyl, alkyoxycarbonyyl, such as methoxycarbonyl, ethoxycarbonyl, 2,2,2- trichloroethoxycarbonyl, BOC, 2-iodoethoxycaronyl, aralkoxycarbonyl, such as CBZ, 4-methoxybenzyloxycarbonyl or FMOC. Preferred acyl groups are CBZ, FMOC, benzyl and acetyl.

The term “acid-protecting group” or “carboxyl-protecting group” is likewise generally known and relates to groups which are suitable for protecting a -COOH group against chemical reactions, but which can easily be removed after the desired chemical reaction has been carried out elsewhere in the molecule. The use of esters instead of the free acids, for example of substituted and unsubstituted alkyl esters (such as methyl, ethyl, tert-butyl and substituted derivatives thereof), of substituted and unsubstituted benzyl esters or silyl esters, is typical. The type and size of the acid-protecting groups is not crucial, but preference is given to those having 1-20, in particular 1-10, C atoms.

The term “hydroxyl-protecting group” is likewise generally known and relates to groups which are suitable for protecting a hydroxyl group against chemical reactions, but which can easily be removed after the desired chemical reaction has been carried out elsewhere in the molecule. Typical of such groups are the above- mentioned unsubstituted or substituted aryl, aralkyl or acyl groups, furthermore also alkyl groups. Their type and size of the hydroxyl-protecting groups is not crucial, but preference is given to those having 1-20, in particular 1-10, C atoms. Examples of hyrdoxyl-protecting groups are, inter alia, benzyl, p-nitrobenzoyl, p-toluenesulfonyl and acetyl, where benzyl and acetyl are preferred.

Further typical examples of amino-, acid- and hydroxyl-protecting groups are found, for example, in “Greene’s Protective Groups in Organic Synthesis”, fourth edition, Wiley-lnterscience, 2007.

The functional derivatives of the compounds of the present invention to be used as starting materials can be prepared by known methods of amino-acid and peptide synthesis, as described, for example, in the said standard works and patent applications.

The compounds of the present invention are liberated from their functional derivatives, depending on the protecting group used, for example, with the aid of strong acids, advantageously using trifluoroacetic acid or perchloric acid, but also using other strong inorganic acids, such as hydrochloric acid or sulfuric acid, strong organic acids, such as trichloroacetic acid, or sulfonic acids, such as benzoyl- or p- toluenesulfonic acid. The presence of an additional inert solvent and/or a catalyst is possible but is not always necessary.

Depending on the respective synthetic route, the starting materials can optionally be reacted in the presence of an inert solvent.

Suitable inert solvents are, for example, heptane, hexane, petroleum ether, DMSO, benzene, toluene, xylene, trichloroethylene-, 1 ,2-dichloroethane, carbon tetrachloride, chloroform or dichloromethane; alcohols, such as methanol, ethanol, isopropanol, n-propanol, n-butanol or tert-butanol; ethers, such as diethyl ether, diisopropyl ether (preferably for substitution on the indole nitrogen), tetrahydrofuran (THF) or dioxane; glycol ethers, such as ethylene glycol monomethyl or monoethyl ether, ethylene glycol dimethyl ether (diglyme); ketones, such as acetone or butanone; amides, such as acetamide, dimethylacetamide, N-methylpyrrolidone (NMP) or dimethylformamide (DMF); nitriles, such as acetonitrile; esters, such as ethyl acetate, carboxylic acids or acid anhydrides, such as, for example, such as acetic acid or acetic anhydride, nitro compounds, such as nitromethane or nitro- benzene, optionally also mixtures of the said solvents with one another or mixtures with water.

The amount of solvent is not crucial; 10 g to 500 g of solvent can preferably be added per g of the compound of the present invention to be reacted.

It may be advantageous to add an acid-binding agent, for example an alkali metal or alkaline-earth metal hydroxide, carbonate or bicarbonate or other alkali or alkaline- earth metal salts of weak acids, preferably a potassium, sodium or calcium salt, or to add an organic base, such as, for example, triethylamine, dimethylamine, pyridine or quinoline, or an excess of the amine component.

The resultant compounds according to the invention can be separated from the corresponding solution in which they are prepared (for example by centrifugation and washing) and can be stored in another composition after separation, or they can remain directly in the preparation solution. The resultant compounds according to the invention can also be taken up in desired solvents for the particular use.

The reaction duration depends on the reaction conditions selected. In general, the reaction duration is 0.5 hour to 10 days, preferably 1 to 24 hours. On use of a microwave, the reaction time can be reduced to values of 1 to 60 minutes.

The compounds of the present invention and also the starting materials for their preparation are, in addition, prepared by known methods, as described in the literature (for example in standard works, such as Houben-Weyl, Methoden der organischen Chemie [Methods of Organic Chemistry], Georg-Thieme-Verlag, Stuttgart), for example under reaction conditions which are known and suitable for the said reactions. Use can also be made here of variants known per se, which are not described here in greater detail.

Conventional work-up steps, such as, for example, addition of water to the reaction mixture and extraction, enable the compounds to be obtained after removal of the solvent. It may be advantageous, for further purification of the product, to follow this with a distillation or crystallisation or to carry out a chromatographic purification. An acid of the present invention can be converted into the associated addition salt using a base, for example by reaction of equivalent amounts of the acid and base in an inert solvent, such as ethanol, and inclusive evaporation. Suitable bases for this reaction are, in particular, those which give physiologically acceptable salts. Thus, the acid of the present inventioncan be converted into the corresponding metal salt, in particular alkali or alkaline-earth metal salt, using a base (for example sodium hydroxide, potassium hydroxide, sodium carbonate or potassium carbonate) or into the corresponding ammonium salt. Organic bases which give physiologically acceptable salts, such as, for example, ethanolamine, are also suitable for this reaction.

On the other hand, a base of the present invention can be converted into the associated acid-addition salt using an acid, for example by reaction of equivalent amounts of the base and acid in an inert solvent, such as ethanol, with subsequent evaporation. Suitable acids for this reaction are, in particular, those which give physiologically acceptable salts. Thus, it is possible to use inorganic acids, for example sulfuric acid, nitric acid, hydrohalic acids, such as hydrochloric acid or hydrobromic acid, phosphoric acids, such as orthophosphoric acid, sulfamic acid, furthermore organic acids, in particular aliphatic, alicyclic, araliphatic, aromatic or heterocyclic, mono- or polybasic carboxylic, sulfonic or sulfuric acids, for example formic acid, acetic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, lactic acid, tartaric acid, malic acid, citric acid, gluconic acid, ascorbic acid, nicotinic acid, isonicotinic acid, methane- or ethanesulfonic acid, ethanedisulfonic acid, 2-hydroxysulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, naphthalenemom- and disulfonic acids or laurylsulfuric acid. Salts with physiologically unacceptable acids, for example picrates, can be used for the isolation and/or purification of the compounds of the present invention.

It has been found that the compounds of the present invention are well tolerated and have valuable pharmacological properties.

The invention therefore furthermore relates to the use of compounds according to the invention for the preparation of a medicament for the treatment and/or prophylaxis of diseases which are caused, promoted and/or propagated by HSET. The invention thus also relates, in particular, to a medicament comprising at least one compound according to the invention and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis of physiological and/or pathophysiological states.

Particular preference is given to physiological and/or pathophysiological states which are connected to HSET.

Physiological and/or pathophysiological states are taken to mean physiological and/or pathophysiological states which are medically relevant, such as, for example, diseases or illnesses and medical disorders, complaints, symptoms or complications and the like, in particular diseases.

The invention furthermore relates to a medicament comprising at least one compound according to the invention and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis of physiological and/or pathophysiological states selected from the group consisting of hyperproliferative diseases and disorders.

The invention further relates to a medicament comprising at least one compound according to the invention and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis of physiological and/or pathophysiological states selected from the group consisting of hyperproliferative and infectious diseases and disorders, wherein the hyperproliferative disease or disorder is cancer.

The invention thus particularly preferably relates to a medicament comprising at least one compound according to the invention and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, wherein the cancer is selected from the group consisting of acute and chronic lymphocytic leukemia, acute granulocytic leukemia, adrenal cortex cancer, bladder cancer, brain cancer, breast cancer, cervical hyperplasia, cervical cancer, chorio cancer, chronic granulocytic leukemia, chronic lymphocytic leukemia, colon cancer, endometrial ccancer, esophageal cancer, essential thrombocytosis, genitourinary carcinoma, glioma, glioblastoma, hairy cell leukemia, head and neck carcinoma, Hodgkin's disease, Kaposi's sarcoma, lung carcinoma, lymphoma, malignant carcinoid carcinoma, malignant hypercalcemia, malignant melanoma, malignant pancreatic insulinoma, medullary thyroid carcinoma, melanoma, multiple myeloma, mycosis fungoides, myeloid and lymphocytic leukemia, neuroblastoma, non-Hodgkin's lymphoma, non-small cell lung cancer, osteogenic sarcoma, ovarian carcinoma, pancreatic carcinoma, polycythemia vera, primary brain carcinoma, primary macroglobulinemia, prostatic cancer, renal cell cancer, rhabdomyosarcoma, skin cancer, small-cell lung cancer, soft-tissue sarcoma, squamous cell cancer, stomach cancer, testicular cancer, thyroid cancer and Wilms' tumor.

The invention further preferably relates to a medicament comprising at least one compound according to the invention and/or one of its physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers, including mixtures thereof in all ratios, for use in the treatment and/or prophylaxis of physiological and/or pathophysiological states selected from the group consisting of hyperproliferative and infectious diseases and disorders, wherein the hyperproliferative disease or disorder is selected from the group consisting of age-related macular degeneration, Crohn's disease, cirrhosis, chronic inflammatory-related disorders, proliferative diabetic retinopathy, proliferative vitreoretinopathy, retinopathy of prematurity, granulomatosis, immune hyperproliferation associated with organ or tissue transplantation and an immunoproliferative disease or disorder selected from the group comnsisting of inflammatory bowel disease, psoriasis, rheumatoid arthritis, systemic lupus erythematosus (SLE), vascular hyperproliferation secondary to retinal hypoxia and vasculitis.

It is intended that the medicaments disclosed above include a corresponding use of the compounds according to the invention for the preparation of a medicament for the treatment and/or prophylaxis of the above physiological and/or pathophysiological states. It is additionally intended that the medicaments disclosed above include a corresponding method for the treatment and/or prophylaxis of the above physiological and/or pathophysiological states in which at least one compound according to the invention is administered to a patient in need of such a treatment.

The compounds according to the invention preferably exhibit an advantageous biological activity which can easily be demonstrated in enzyme assays and animal experiments, as described in the examples. In such enzyme-based assays, the compounds according to the invention preferably exhibit and cause an inhibiting effect, which is usually documented by IC50 values in a suitable range, preferably in the micromolar range and more preferably in the nanomolar range.

The compounds according to the invention can be administered to humans or animals, in particular mammals, such as apes, dogs, cats, rats or mice, and can be used in the therapeutic treatment of the human or animal body and in the combating of the above-mentioned diseases. They can furthermore be used as diagnostic agents or as reagents.

Furthermore, compounds according to the invention can be used for the isolation and investigation of the activity or expression of HSET. In addition, they are particularly suitable for use in diagnostic methods for diseases in connection with disturbed HSET activity. The invention therefore furthermore relates to the use of the compounds according to the invention for the isolation and investigation of the activity or expression of HSET or as binders and inhibitors of HSET.

For diagnostic purposes, the compounds according to the invention can, for example, be radioactively labelled. Examples of radioactive labels are 3 H, 14 C, 231 l and 125 l. A preferred labelling method is the iodogen method (Fraker et al., 1978). In addition, the compounds according to the invention can be labelled by enzymes, fluorophores and chemophores. Examples of enzymes are alkaline phosphatase, - galactosidase and glucose oxidase, an example of a fluorophore is fluorescein, an example of a chemophore is luminol, and automated detection systems, for example for fluorescent colorations, are described, for example, in US 4,125,828 and US 4,207,554. The present invention further relates to pharmaceutical compositions containing the compounds of the present invention and their use for the treatment and/or prophylaxis of diseases and disorders where the partial or total inactivation of HSET could be beneficial.

The compounds of the present invention can be used for the preparation of pharmaceutical preparations, in particular by non-chemical methods. In this case, they are brought into a suitable dosage form together with at least one solid, liquid and/or semi-liquid excipient or adjuvant and optionally in combination with one or more further active compound(s).

The invention therefore furthermore relates to pharmaceutical preparations comprising at least one compound of the present invention and/or physiologically acceptable salts, derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios. In particular, the invention also relates to pharmaceutical preparations which comprise further excipients and/or adjuvants, and also to pharmaceutical preparations which comprise at least one further medicament active compound.

In particular, the invention also relates to a process for the preparation of a pharmaceutical preparation, characterised in that a compound of the present inventionand/or one of its physiologically acceptable salts, derivatives, solvates and stereoisomers, including mixtures thereof in all ratios, is brought into a suitable dosage form together with a solid, liquid or semi-liquid excipient or adjuvant and optionally with a further medicament active compound.

The pharmaceutical preparations according to the invention can be used as medicaments in human or veterinary medicine. The patient or host can belong to any mammal species, for example a primate species, particularly humans; rodents, including mice, rats and hamsters; rabbits; horses, cattle, dogs, cats, etc. Animal models are of interest for experimental investigations, where they provide a model for the treatment of a human disease.

Suitable carrier substances are organic or inorganic substances which are suitable for enteral (for example oral), parenteral or topical administration and do not react with the novel compounds, for example water, vegetable oils (such as sunflower oil or cod-liver oil), benzyl alcohols, polyethylene glycols, gelatine, carbohydrates, such as lactose or starch, magnesium stearate, talc, lanolin or vaseline. Owing to his expert knowledge, the person skilled in the art is familiar which adjuvants are suitable for the desired medicament formulation. Besides solvents, for example water, physiological saline solution or alcohols, such as, for example, ethanol, propanol or glycerol, sugar solutions, such as glucose or mannitol solutions, or a mixture of the said solvents, gel formers, tablet assistants and other activeingredient carriers, it is also possible to use, for example, lubricants, stabilisers and/or wetting agents, emulsifiers, salts for influencing the osmotic pressure, antioxidants, dispersants, antifoams, buffer substances, flavours and/or aromas or flavour correctants, preservatives, solubilisers or dyes. If desired, preparations or medicaments according to the invention may comprise one or more further active compounds, for example one or more vitamins.

If desired, preparations or medicaments according to the invention may comprise one or more further active compounds and/or one or more action enhancers (adjuvants).

The terms “pharmaceutical formulation” and “pharmaceutical preparation” are used as synonyms for the purposes of the present invention.

As used here, “pharmaceutically tolerated” relates to medicaments, precipitation reagents, excipients, adjuvants, stabilisers, solvents and other agents which facilitate the administration of the pharmaceutical preparations obtained therefrom to a mammal without undesired physiological side effects, such as, for example, nausea, dizziness, digestion problems or the like.

In pharmaceutical preparations for parenteral administration, there is a requirement for isotonicity, euhydration and tolerability and safety of the formulation (low toxicity), of the adjuvants employed and of the primary packaging. Surprisingly, the compounds according to the invention preferably have the advantage that direct use is possible and further purification steps for the removal of toxicologically unacceptable agents, such as, for example, high concentrations of organic solvents or other toxicologically unacceptable adjuvants, are thus unnecessary before use of the compounds according to the invention in pharmaceutical formulations.

The invention particularly preferably also relates to pharmaceutical preparations comprising at least one compound according to the invention in precipitated noncrystalline, precipitated crystalline or in dissolved or suspended form, and optionally excipients and/or adjuvants and/or further pharmaceutical active compounds.

The compounds according to the invention preferably enable the preparation of highly concentrated formulations without unfavourable, undesired aggregation of the compounds according to the invention occurring. Thus, ready-to-use solutions having a high active-ingredient content can be prepared with the aid of compounds according to the invention with aqueous solvents or in aqueous media.

The compounds and/or physiologically acceptable salts and solvates thereof can also be lyophilised and the resultant lyophilisates used, for example, for the preparation of injection preparations.

Aqueous preparations can be prepared by dissolving or suspending compounds according to the invention in an aqueous solution and optionally adding adjuvants. To this end, defined volumes of stock solutions comprising the said further adjuvants in defined concentration are advantageously added to a solution or suspension having a defined concentration of compounds according to the invention, and the mixture is optionally diluted with water to the pre-calculated concentration. Alternatively, the adjuvants can be added in solid form. The amounts of stock solutions and/or water which are necessary in each case can subsequently be added to the aqueous solution or suspension obtained. Compounds according to the invention can also advantageously be dissolved or suspended directly in a solution comprising all further adjuvants.

The solutions or suspensions comprising compounds according to the invention and having a pH of 4 to 10, preferably having a pH of 5 to 9, and an osmolality of 250 to 350 mosmol/kg can advantageously be prepared. The pharmaceutical preparation can thus be administered directly substantially without pain intravenously, intraarterially, intraarticularly, subcutaneously or percutaneously. In addition, the preparation may also be added to infusion solutions, such as, for example, glucose solution, isotonic saline solution or Ringer's solution, which may also contain further active compounds, thus also enabling relatively large amounts of active compound to be administered.

Pharmaceutical preparations according to the invention may also comprise mixtures of a plurality of compounds according to the invention.

The preparations according to the invention are physiologically well tolerated, easy to prepare, can be dispensed precisely and are preferably stable with respect to assay, decomposition products and aggregates throughout storage and transport and during multiple freezing and thawing processes. They can preferably be stored in a stable manner over a period of at least three months to two years at refrigerator temperature (2-8°C) and at rt (23-27 °C) and 60% relative atmospheric humidity (R.H.).

For example, the compounds according to the invention can be stored in a stable manner by drying and when necessary converted into a ready-to-use pharmaceutical preparation by dissolution or suspension. Possible drying methods are, for example, without being restricted to these examples, nitrogen-gas drying, vacuum-oven drying, lyophilisation, washing with organic solvents and subsequent air drying, liquid-bed drying, fluidised-bed drying, spray drying, roller drying, layer drying, air drying at rt and further methods.

The term “effective amount” denotes the amount of a medicament or of a pharmaceutical active compound which causes in a tissue, system, animal or human a biological or medical response which is sought or desired, for example, by a researcher or physician.

In addition, the term “therapeutically effective amount” denotes an amount which, compared with a corresponding subject who has not received this amount, has the following consequence: improved treatment, healing, prevention or elimination of a disease, syndrome, disease state, complaint, disorder or prevention of side effects or also a reduction in the progress of a disease, complaint or disorder. The term “therapeutically effective amount” also encompasses the amounts which are effective for increasing normal physiological function.

On use of preparations or medicaments according to the invention, the compounds according to the invention and/or physiologically acceptable salts and solvates thereof are generally used analogously to known, commercially available preparations or preparations, preferably in dosages of between 0.1 and 500 mg, in particular 5 and 300 mg, per use unit. The daily dose is preferably between 0.001 and 250 mg/kg, in particular 0.01 and 100 mg/kg, of body weight. The preparation can be administered one or more times per day, for example two, three or four times per day. However, the individual dose for a patient depends on a large number of individual factors, such as, for example, on the efficacy of the particular compound used, on the age, body weight, general state of health, sex, nutrition, on the time and method of administration, on the excretion rate, on the combination with other medicaments and on the severity and duration of the particular disease.

A measure of the uptake of a medicament active compound in an organism is its bioavailability. If the medicament active compound is delivered to the organism intravenously in the form of an injection solution, its absolute bioavailability, i.e. the proportion of the pharmaceutical which reaches the systemic blood, i.e. the major circulation, in unchanged form, is 100%. In the case of oral administration of a therapeutic active compound, the active compound is generally in the form of a solid in the formulation and must therefore first be dissolved in order that it is able to overcome the entry barriers, for example the gastrointestinal tract, the oral mucous membrane, nasal membranes or the skin, in particular the stratum corneum, or can be absorbed by the body. Data on the pharmacokinetics, i.e. on the bioavailability, can be obtained analogously to the method of J. Shaffer et al., J. Pharm. Sciences, 88 (1999), 313-318.

Furthermore, medicaments of this type can be prepared by means of one of the processes generally known in the pharmaceutical art.

Medicaments can be adapted for administration via any desired suitable route, for example by the oral (including buccal or sublingual), rectal, pulmonary, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal and in particular intraarticular) routes. Medicaments of this type can be prepared by means of all processes known in the pharmaceutical art by, for example, combining the active compound with the excipient(s) or adjuvant(s).

Parenteral administration is preferably suitable for administration of the medicaments according to the invention. In the case of parenteral administration, intra-articular administration is particularly preferred.

The compounds according to the invention are also suitable for the preparation of medicaments to be administered parenterally having slow, sustained and/or controlled release of active compound. They are thus also suitable for the preparation of delayed-release formulations, which are advantageous for the patient since administration is only necessary at relatively large time intervals.

The medicaments adapted to parenteral administration include aqueous and nonaqueous sterile injection solutions comprising antioxidants, buffers, bacteriostatics and solutes, by means of which the formulation is rendered isotonic with the blood or synovial fluid of the recipient to be treated; as well as aqueous and non-aqueous sterile suspensions, which can comprise suspension media and thickeners. The formulations can be delivered in single-dose or multi-dose containers, for example sealed ampoules and vials, and stored in the freeze-dried (lyophilised) state, so that only the addition of the sterile carrier liquid, for example water for injection purposes, immediately before use is necessary. Injection solutions and suspensions prepared in accordance with the formulation can be prepared from sterile powders, granules and tablets.

The compounds according to the invention can also be administered in the form of liposome delivery systems, such as, for example, small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from various phospholipids, such as, for example, cholesterol, stearylamine or phosphatidylcholines.

The compounds according to the invention can also be coupled to soluble polymers as targeted medicament excipients. Such polymers can encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidophenol, polyhydroxyethylaspartamidophenol or polyethylene oxide polylysine, substituted by palmitoyl radicals. The compounds according to the invention can furthermore be coupled to a class of biodegradable polymers which are suitable for achieving slow release of a medicament, for example polylactic acid, poly-epsilon-caprolactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydroxypyrans, polycyanoacrylates, polylactic-co-glycolic acid, polymers, such as conjugates between dextran and methacrylates, polyphosphoesters, various polysaccharides and polyamines and poly-s-caprolactone, albumin, chitosan, collagen or modified gelatine and crosslinked or amphipathic block copolymers of hydrogels.

Suitable for enteral administration (oral or rectal) are, in particular, tablets, dragees, capsules, syrups, juices, drops or suppositories, and suitable for topical use are ointments, creams, pastes, lotions, gels, sprays, foams, aerosols, solutions (for example solutions in alcohols, such as ethanol or isopropanol, acetonitrile, DMF, dimethylacetamide, 1,2-propanediol or mixtures thereof with one another and/or with water) or powders. Also, particularly suitable for topical uses are liposomal preparations.

In the case of formulation to give an ointment, the active compound can be employed either with a paraffinic or a water-miscible cream base. Alternatively, the active compound can be formulated to a cream with an oil-in-water cream base or a water-in-oil base.

Medicaments adapted to transdermal administration can be delivered as independent plasters for extended, close contact with the epidermis of the recipient. Thus, for example, the active compound can be supplied from the plaster by means of iontophoresis, as described in general terms in Pharmaceutical Research, 3 (6), 318 (1986).

It goes without saying that, besides the constituents particularly mentioned above, the medicaments according to the invention may also comprise other agents usual in the art with respect to the particular type of pharmaceutical formulation.

The invention also relates to a set (kit) consisting of separate packs of a) an effective amount of a compound of the present invention and/or physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios, and b) an effective amount of a further medicament active compound.

The set comprises suitable containers, such as boxes or cartons, individual bottles, bags or ampoules. The set may, for example, comprise separate ampoules each containing an effective amount of a compound of the present inventionand/or pharmaceutically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios, and an effective amount of a further medicament active compound in dissolved or lyophilised form.

Furthermore, the medicaments according to the invention can be used in order to provide additive or synergistic effects in certain known therapies and/or can be used in order to restore the efficacy of certain existing therapies.

Besides the compounds according to the invention, the pharmaceutical preparations according to the invention may also comprise further medicament active compounds, for example for use in the treatment of cancer, other anti-tumor medicaments. For the treatment of the other diseases mentioned, the pharmaceutical preparations according to the invention may also, besides the compounds according to the invention, comprise further medicament active compounds which are known to the person skilled in the art in the treatment thereof.

In one principal embodiment, methods are provided for enhancing an immune response in a host in need thereof. The immune response can be enhanced by reducing T cell tolerance, including by increasing IFN-y release, by decreasing regulatory T cell production or activation, or by increasing antigen-specific memory T cell production in a host. In one embodiment, the method comprises administering a compound of the present invention to a host in combination or alternation with an antibody. In particular subembodiments, the antibody is a therapeutic antibody. In one particular embodiment, a method of enhancing efficacy of passive antibody therapy is provided comprising administering a compound of the present invention in combination or alternation with one or more passive antibodies. This method can enhance the efficacy of antibody therapy for treatment of abnormal cell proliferative disorders such as cancer or can enhance the efficacy of therapy in the treatment or prevention of infectious diseases. The compound of the present invention can be administered in combination or alternation with antibodies such as rituximab, herceptin or erbitux, for example.

In another principal embodiment, a method of treating or preventing abnormal cell proliferation is provided comprising administering a compound of the present invention to a host in need thereof substantially in the absence of another anticancer agent.

In another principal embodiment, a method of treating or preventing abnormal cell proliferation in a host in need thereof is provided, comprising administering a first compound of the present invention substantially in combination with a first anticancer agent to the host and subsequently administering a second compound of the present invention receptor antagonist. In one subembodiment, the second antagonist is administered substantially in the absence of another anti-cancer agent. In another principal embodiment, a method of treating or preventing abnormal cell proliferation in a host in need thereof is provided, comprising administering a compound of the present invention substantially in combination with a first anticancer agent to the host and subsequently administering a second anti-cancer agent in the absence of the antagonist.

Thus, the cancer treatment disclosed here can be carried out as therapy with a compound of the present invention or in combination with an operation, irradiation or chemotherapy. Chemotherapy of this type can include the use of one or more active compounds of the following categories of antitumour active compounds:

(i) antiproliferative/antineoplastic/DNA-damaging active compounds and combinations thereof, as used in medical oncology, such as alkylating active compounds (for example cis-platin, parboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines such as 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea and gemcitabine); antitumour antibiotics (for example anthracyclines, such as adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic active compounds (for example vinca alkaloids, such as vincristine, vin- biastine, vindesine and vinorelbine, and taxoids, such as taxol and taxotere); topoisomerase inhibitors (for example epipodophyllotoxins, such as etoposide and teniposide, amsacrine, topotecan, irinotecan and camptothecin) and celldifferentiating active compounds (for example all-trans-retinoic acid, 13-cis-retinoic acid and fenretinide);

(ii) cytostatic active compounds, such as anti-oestrogens (for example tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene), oestrogen receptor regulators (for example fulvestrant), anti-androgens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progesterones (for example megestrol acetate), aromatase inhibitors (for example anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5a-reductase, such as finasteride;

(iii) active compounds which inhibit cancer invasion including for example metalloproteinase inhibitors, like marimastat, and inhibitors of urokinase plasminogen activator receptor function;

(iv) inhibitors of growth factor function, for example growth factor antibodies, growth factor receptor antibodies, for example the anti-erbb2 antibody trastuzumab [Herceptin™] and the anti-erbbl antibody cetuximab [C225]), farnesyl transferase inhibitors, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors, such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6- (3- morpholinopropoxy) quinazolin-4-amine (gefitinib, AZD1839), N-(3-ethynylphenyl)- 6,7-bis (2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido- N-(3-chloro-4-fluorophenyl)-7-(3-morpholinopropoxy)quinazoli n-4-amine (Cl 1033), for example inhibitors of the platelet-derived growth factor family and, for example, inhibitors of the hepatocyte growth factor family;

(v) anti-angiogenic active compounds, such as bevacizumab, angiostatin, endostatin, linomide, batimastat, captopril, cartilage derived inhibitor, genistein, interleukin 12, lavendustin, medroxypregesterone acetate, recombinant human platelet factor 4, tecogalan, thrombospondin, TNP-470, anti-VEGF monoclonal antibody, soluble VEGF-receptor chimaeric protein, anti-VEGF receptor antibodies, anti-PDGF receptors, inhibitors of integrins, tyrosine kinase inhibitors, serine/threonine kinase inhibitors, antisense oligonucleotides, antisense oligodexoynucleotides, siRNAs, anti-VEGF aptamers, pigment epithelium derived factor and compounds which have been published in the international patent applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354);

(vi) vessel-destroying agents, such as combretastatin A4 and compounds which have been published in the international patent applications WO 99/02166,

WO 00/40529, WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213;

(vii) antisense therapies, for example those directed to the targets mentioned above, such as ISIS 2503, an anti-Ras antisense;

(viii) gene therapy approaches, including, for example, approaches for replacement of abnormal, modified genes, such as abnormal p53 or abnormal BRCA1 or BRCA2, GDEPT approaches (gene-directed enzyme pro-drug therapy), such as those which use cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme, and approaches which increase the tolerance of a patient to chemotherapy or radiotherapy, such as multi-drug resistance therapy; and

(ix) immunotherapy approaches, including, for example, ex-vivo and in-vivo approaches for increasing the immunogenicity of tumour cells of a patient, such as transfection with cytokines, such as interleukin 2, interleukin 4 or granulocyte macrophage colony stimulating factor, approaches for decreasing T-cell anergy, approaches using transfected immune cells, such as cytokine-transfected dendritic cells, approaches for use of cytokine-transfected tumour cells and approaches for use of anti-idiotypic antibodies

(x) chemotherapeutic agents including foor example abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, asparaginase, BCG live, bevaceizumab, bexarotene, bleomycin, bortezomib, busulfan, calusterone, camptothecin, capecitabine, carboplatin, carmustine, celecoxib, cetuximab, chlorambucil, cinacalcet, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, daunorubicin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin, dromostanolone, epirubicin, epoetin alfa, estramustine, etoposide, exemestane, filgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant and gemcitabine.

The medicaments from table 1 can preferably, but not exclusively, be combined with the compounds of the present invention.

Even without further embodiments, it is assumed that a person skilled in the art will be able to use the above description in the broadest scope. The preferred embodiments should therefore merely be regarded as descriptive disclosure which is absolutely not limiting in any way.

The following examples are thus intended to explain the invention without limiting it. Unless indicated otherwise, per cent data denote per cent by weight. All temperatures are indicated in degrees Celsius. “Conventional work-up”: water is added if necessary, the pH is adjusted, if necessary, to values between 2 and 10, depending on the constitution of the end product, the mixture is extracted with ethyl acetate or dichloromethane, the phases are separated, the organic phase is dried over sodium sulfate or magnesium sulfate, filtered and evaporated, and the product is purified by chromatography on silica gel and/or by crystallisation.

Rf values on silica gel; mass spectrometry: El (electron impact ionisation): M + , FAB (fast atom bombardment): (M+H) + , THF (tetrahydrofuran), NMP (N-methlpyrrolidone), DMSO (dimethyl sulfoxide), EtOAc (ethyl acetate), MeOH (methanol), EtOH (ethanol), TLC (thin-layer chromatography)

List of Abbreviations

AUC Area under the plasma drug concentration-time curve

Cmax Maximum plasma concentration

CL Clearance

CV Coefficient of variation CYP Cytochrome P450

DMSO Dimethyl sulfoxide

F Bioavailability f a Fraction absorbed iv Intravenous

LC-MS/MS Liquid chromatography tandem mass spectrometry

LLOQ Lower limit of quantification

NC Not calculated

ND Not determined

PEG Polyethylene glycol

Pgp Permeability glycoprotein

PK Pharmacokinetic(s) po Per os (oral) rt Room temperature ti/2 Half-life Time at which maximum plasma concentration of drug is reached

UPLC Ultra performance liquid chromatography

Vss Volume of distribution (at steady state) v/v Volume to volume

Preparation of the compounds of the present invention and analytical methods

The invention especially relates to the compounds of the following examples and physiologically acceptable salts, derivatives, solvates, prodrugs and stereoisomers thereof, including mixtures thereof in all ratios.

All solvents used were commercially available and used without further purification. Reactions were typically run using anhydrous solvents under an inert atmosphere of nitrogen or argon. Flash column chromatography was carried out using Silica gel 60 (0.035-0.070 mm particle size). Flash column chromatography was also carried out using a Biotage purification system using SNAP KP-Sil cartridges or on reversephase mode using SNAP Ultra C18 cartridges. Microwave-assisted reactions were carried out using a Biotage Initiator microwave system. 1 H NMR spectra were recorded on Bruker DPX-300, DRX-400, Avance II-400, Avance III HD-400, Avance II+-500, Avance III-500, Avance Neo 600 or on a Avance III-700 spectrometer, using residual signal of deuterated solvent as internal reference. Chemical shifts (5) are reported in ppm relative to tetramethylsilane (TMS), referenced to the internal deuterated solvent. 1 H NMR data are reported as follows: chemical shift (multiplicity, coupling constants, and number of hydrogens). Multiplicity is abbreviated as follows: s (singlet), d (doublet), t (triplet), q (quartet), sext (sextet), hept (heptet), m (multiplet), br (broad).

HPLC/MS spectra of the products were recorded on an Agilent 1100 HPLC system (1100 high pressure gradient pump, 1100 diode array detector, wavelength: 220 nm) interfaced to an Agilent 1100 mass spectrometer detector (positive mode). LC-MS analyses were performed on a SHIMADZU LC-MS machine consisting of an LIFLC 20-AD system and LCMS 2020 MS detector.

Details of the applied conditions for HPLC/MS spectra recorded on a Shimadzu LCMS-2020 system or an Agilent 1200 system:

(A): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.1 % FA, mobile phase B: ACN with 0.1% FA; gradient: 5% B to 100% B till min 1.50, hold till min 1.80, 100% B to 5% B till min 1.81 , stop after 2.00; flow: 1.5 mL/min.

(B): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.05% TFA, mobile phase B: ACN with 0.05% TFA; gradient: 5% B to 100% B till min 1.20, hold till min 1.80, 100% B to 5% B till min 1.82, stop after 2.00; flow: 1.5 mL/min.

(C): column: HALO C18 90A, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.05% TFA, mobile phase B: ACN with 0.05% TFA; gradient: 5% B to 100% B till min 1.20, hold till min 1.80, 100% B to 5% B till min 1.82, stop after 2.00; flow: 1.2 mL/min.

(D): column: Shim-pack C18, 3.0x33 mm, 3.0 pm; mobile phase A: water with 5 mM NH4CO3, mobile phase B: ACN; gradient: 10% B to 95% B till min 1.2, hold till min 1.8 min, 95% B to 10% B till min 1.82, stop after 2.00; flow: 1.5 mL/min. (E): column: HALO C18, 3.0*30 mm, 3.0 pm; mobile phase A: water with 0.05% TFA, mobile phase B: ACN with 0.05% TFA; gradient: 5% B to 100% B till min 1.20, hold till min 1.80, 100% B to 5% B till min 1.82, stop after 2.00; flow: 1.5 mL/min.

(F): column: HALO C18 90A, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.05% TFA, mobile phase B: ACN with 0.05% TFA; gradient: 5% B to 60% B till min 1.80, 60% B to 95% B till min 2.50, hold till min 2.80, 95% B to 5% B till min 2.81 , stop after 3.00; flow: 1.2 mL/min.

(G): column: Chromolith HR C18 RP-18e, 4.6x50 mm; mobile phase A: water with 0.05% FA, mobile phase B: ACN with 0.04% FA + 1% water; gradient: 0% B to 100% B till min 2.0, hold till min 2.5, 100% B to 0% B till min 2.51 , stop after 2.95; flow: 3.3 mL/min.

(H): column: Kinetex EVO C18, 4.6x50 mm, 5.0 pm; mobile phase A: water with 0.05% FA, mobile phase B: ACN with 0.04% FA and 1 % water; gradient: 1% B to 99% B till min 0.8, 99% B to 1% B till min 1.1 , stop after 1.50; flow: 3.3 mL/min.

(I): column: Kinetex EVO C18 100A, 3.0x50 mm, 2.6 pm; mobile phase A: water with 0.04% NH4OH, mobile phase B: ACN; gradient: 10% B to 60% B in 2.2 min, 60% B to 95% B in 2.7 min, hold till min 3.2, 95% B to 10% B till min 3.3, stop after 3.5; flow: 1.2 mL/min.

(J): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.05% TFA, mobile phase B: ACN with 0.05% TFA; gradient: 5% B to 100% B till min 0.70, hold till min 1.10, 100% B to 5% B till min 1.12, stop after 1.20; flow: 1.5 mL/min.

(K): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.1 % FA, mobile phase B: ACN with 0.1% FA; gradient: 5% B to 95% B till min 2.10, hold till min 2.75, 95% B to 5% B till min 2.81 , stop after 3.00; flow: 1.5 mL/min.

(L): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.1 % FA, mobile phase B: ACN with 0.1% FA; gradient: 10% B to 95% B till min 1.20, hold till min 1.80, 95% B to 10% B till min 1.82, stop after 2.00; flow: 1.5 mL/min. (M) column: Sunfire C18, 3.0x100 mm, 5 pm; mobile phase A: water with 0.05% FA, mobile phase B: ACN with 0.04% FA and 1% water; gradient: 1% B to 99% B in 2.0 min, hold till min 3.5, 99% B to 1% B till min 2.71 , stop after 3.5; flow: 1.4 mL/min.

(N): column: Chromolith HR C18, 4.6x50 mm, 5 pm; mobile phase A: water with 0.1 % TFA, mobile phase B: ACN with 0.1% TFA; gradient: 1 % B to 99% B till min 2.0, hold till min 2.5, 99% B to 1% B till min 2.51 , stop after 2.95; flow: 3.3 mL/min.

(O): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.05% TFA, mobile phase B: ACN with 0.05% TFA; gradient: 5% B to 50% B till min 1.80, 50% B to 95% B till min 2.50, hold till min 2.80, 95% B to 5% B till min 2.81 , stop after 3.00; flow: 1 .5 mL/min.

(P): column: Shim-pack velox, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.1% FA, mobile phase B: ACN with 0.1% FA; gradient: 5% B to 100% B till min 1.50, hold till min 1.80, 100% B to 5% B till min 1.81 , stop after 2.00; flow: 1.5 mL/min.

(Q): column: Kinetex EVO C18, 3.0x50 mm, 2.6 pm; mobile phase A: 6.5 mM NH4HCO3 + NH4OH (pH10), mobile phase B: ACN; gradient: 10% B to 70% B in 2.2 min, 70% B to 95% B in 2.7 min, hold till min 3.2, 95% B to 10% B till min 3.3, stop after 3.5; flow: 1.2 mL/min.

(R): column: HALO C18, 3.0*30 mm, 2.0 pm; mobile phase A: water with 0.1% FA, mobile phase B: ACN with 0.1% FA; gradient: 5% B to 100% B till min 1.20, hold till min 1.80, 100% B to 5% B till min 1.82, stop after 2.00; flow: 1.5 mL/min.

(S): column: Kinetex EVO C18, 3.0x50 mm, 2.6 pm; mobile phase A: 6.5 mM NH4HCO3 + NH4OH (pH10), mobile phase B: ACN; gradient: 10% B to 95% B in 1.9 min, hold till min 2.7, 95% B to 10% B till min 2.75, stop after 3.0; flow: 1.2 mL/min.

(T): column: Kinetex EVO C18, 2.1x30 mm, 5.0 pm; mobile phase A: 0.0375% TFA in water (v/v), mobile phase B: 0.01875% TFA in Acetonitrile (v/v); gradient: 5% B to 95% B in 0.8 min, 95% B till min 1.2, 5% B til stop after 1.55 min; flow: 1.5 mL/min. (II): column: Kinetex EVO C18, 2.1x30 mm, 5.0 pm; mobile phase A: 0.0375% TFA in water (v/v), mobile phase B: 0.01875% TFA in Acetonitrile (v/v); gradient: 0% B to 60% B in 0.8 min, 60% B till min 1.2, 0% B til stop after 1.55 min; flow: 1.5 mL/min.

(V): column: Kinetex EVO C18, 2.1x30 mm, 5.0 pm; mobile phase A: 0.025% NH3 H2O in water (v/v), mobile phase B: Acetonitrile; gradient: 5% B to 95% B in 0.8 min, 95% B till min 1.2, 5% B til stop after 1.55 min; flow: 1.5 mL/min.

(W): column: Poroshell HPH-C18, 3.0x50 mm, 2.7 pm; mobile phase A: water with 5 mM NH4CO3, mobile phase B: AON; gradient: 10% B to 95% B till min 2.00, hold till min 2.70 min, 95% B to 10% B till min 2.75, stop after 3.00; flow: 1.2 mL/min.

(X): column: Poroshell HPH-C18, 3.0x50 mm, 2.7 pm; mobile phase A: water with 5 mM NH4CO3, mobile phase B: ACN; gradient: 10% B to 70% B till min 2.20, 70% B to 95% B till min 2.70, hold till min 3.20 min, 95% B to 10% B till min 3.30, stop after 3.50; flow: 1.2 mL/min.

Details of the applied conditions for HPLC/MS spectra on an Agilent 1200 series HPLC and diode array detector coupled to a 6210 time of flight mass spectrometer with dual multimode APCI/ESI source.

(AA): Analytical separation was carried out at 40°C on a Merck Chromolith Flash column (RP-18e, 25 x 2 mm) using a flow rate of 1.5 mL/min in a 2 minute gradient elution with detection at 254 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 5:95 (A/B) to 100:0 (A/B) over 1.25 min, 100:0 (A/B) for 0.5 min, and then reversion back to 5:95 (A/B) over 0.05 min, finally 5:95 (A/B) for 0.2 min.

(AB) Analytical separation was carried out at 30°C on a Merck Chromolith Flash column (RP-18e, 25 x 2 mm) using a flow rate of 0.75 mL/min in a 4 minute gradient elution with detection at 254 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 5:95 (A/B) to 100:0 (A/B) over 2.5 min, 100:0 (A/B) for 1 min, and then reversion back to 5:95 (A/B) over 0.1 min, finally 5:95 (A/B) for 0.4 min.

Details of the applied conditions for HPLC/MS spectra on a Waters Acquity LIPLC and diode array detector coupled to a Waters G2 QToF mass spectrometer fitted with a multimode ESI/APCI source.

(AC) Analytical separation was carried out at 30°C on a Phenomenex Kinetex C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.5 mL/min in a 2 minute gradient elution with detection at 254 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 1.25 min, 90:10 (A/B) for 0.5 min, and then reversion back to 10:90 (A/B) over 0.15 min, finally 10:90 (A/B) for 0.1 min.

(AD) Analytical separation was carried out at 30°C on a Phenomenex Kinetex C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.3 mL/min in a 4 minute gradient elution with detection at 254 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 3 min, 90:10 (A/B) for 0.5 min, and then reversion back to 10:90 (A/B) over 0.3 min, finally 10:90 (A/B) for 0.2 min.

(AE) Analytical separation was carried out at 30°C on an Agilent Poroshell C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.5 mL/min in a 2 minute gradient elution with detection at 254 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 1.25 min, 90:10 (A/B) for 0.5 min, and then reversion back to 10:90 (A/B) over 0.15 min, finally 10:90 (A/B) for 0.1 min.

(AF) Analytical separation was carried out at 30°C on an Agilent Poroshell C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.3 mL/min in a 4 minute gradient elution with detection at 254 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 3 min, 90:10 (A/B) for 0.5 min, and then reversion back to 10:90 (A/B) over 0.3 min, finally 10:90 (A/B) for 0.2 min.

Details of the applied conditions for HPLC/MS spectra on an Agilent 1260 Infinity II series LIPLC and diode array detector coupled to a 6530 Quadrupole time of flight mass spectrometer with Agilent Jet Stream ESI source.

(AG) Analytical separation was carried out at 40°C on an Agilent Poroshell C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.6 mL/min in a 2 minute gradient elution with detection at 254, 280 and 214 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 1.25 min, 90:10 (A/B) for 0.5 min, and then reversion back to 10:90 (A/B) over 0.15 min, finally 10:90 (A/B) for 0.1 min.

(AH) Analytical separation was carried out at 40°C on an Agilent Poroshell C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.4 mL/min in a 4 minute gradient elution with detection at 254, 280 and 214 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 2.5 min, 90:10 (A/B) for 1 min, and then reversion back to 10:90 (A/B) over 0.3 min, finally 10:90 (A/B) for 0.2 min.

(Al) Analytical separation was carried out at 40°C on a Phenomenex Kinetex C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.6 mL/min in a 2 minute gradient elution with detection at 254, 280 and 214 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 1.25 min, 90:10 (A/B) for 0.5 min, and then reversion back to 10:90 (A/B) over 0.15 min, finally 10:90 (A/B) for 0.1 min.

(AJ) Analytical separation was carried out at 40°C on a Phenomenex Kinetex C18 column (30 x 2.1 mm, 2.6u, 100A) using a flow rate of 0.4 mL/min in a 4 minute gradient elution with detection at 254, 280 and 214 nm. The mobile phase was a mixture of methanol (solvent A) and water (solvent B), both containing formic acid at 0.1%. Gradient elution was as follows: 10:90 (A/B) to 90:10 (A/B) over 2.5 min, 90:10 (A/B) for 1 min, and then reversion back to 10:90 (A/B) over 0.3 min, finally 10:90 (A/B) for 0.2 min

Synthesis of intermediates:

A1 : 3-(3-methyl-1,2-oxazol-5-yl)benzoic acid

A1.1: A solution of 1-(3-bromophenyl)ethan-1-one (4.75 g; 23.864 mmol) and (1,1- dimethoxyethyl)dimethylamine (9.53 g; 71.552 mmol) was stirred for 16 h at 100 °C. The resulting mixture was concentrated under vacuum. Then NH2OH*HCI (4.98 g; 71.664 mmol) and EtOH (100.0 mL) were added, and the resulting mixture was stirred at 90 °C for 28 h. The reaction mixture was cooled to ambient temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 3:2) to afford 3.6 g (63%) of 5-(3-bromophenyl)-3-methyl-1 ,2-oxazole as a colorless solid. HPLC/MS m/z: 238.05 [M+H] + , Rt (A): 1.18 min.

A1.2: To a stirred solution of 5-(3-bromophenyl)-3-methyl-1,2-oxazole (900.0 mg; 3.784 mmol) and Et 3 N (1.53 g; 15.120 mmol) in DMF (10.0 mL) were added Pd(dppf)Cl2.CH2Cl2 (309 mg; 0.378 mmol) and formyl acetate (2.63 g; 29.865 mmol). The resulting mixture was stirred under nitrogen atmosphere at 90 °C for 2 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 2:1) to afford 468.0 mg (61%) of 3-(3-methyl-1 ,2- oxazol-5-yl)benzoic acid as a colorless solid. HPLC/MS m/z: 204.2 [M+H] + , Rt (B): 0.73 min.

A2: 3-(5-methyl-1,3-oxazol-2-yl)benzoic acid

A2.1: To a stirred solution of 3-bromobenzoic acid (4.75 g; 23.630 mmol) and prop- 2-yn-1-amine (1.30 g; 23.630 mmol) in DMF (50.0 mL) were added TCFH (9.95 g; 35.445 mmol) and 1-methyl-1 H-imidazole (3.88 g; 47.257 mmol) and the resulting mixture was stirred at room temperature for 1 h. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 3:1) to afford 4.51 g (80%) of 3-bromo-N-(prop-2-yn-1-yl)benzamide as a yellow solid. HPLC/MS m/z: 240.1 [M+H] + , Rt (B): 0.77 min.

A2.2: A solution of 3-bromo-N-(prop-2-yn-1-yl)benzamide (1.80 g; 7.569 mmol) and AuCh (114.80 mg; 0.378 mmol) in ACN (20.0 mL) was stirred under nitrogen atmosphere at 50 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 2:1) to give 587 mg (33%) of 2-(3-bromophenyl)-5-methyl-1 ,3-oxazole as a colorless solid. HPLC/MS m/z: 238.0 [M+H] + , Rt (E): 1.08 min.

A2.3: 2-(3-bromophenyl)-5-methyl-1,3-oxazole (342.70 mg; 1.439 mmol) was converted as described for A1.2. Yield: 230 mg (79%) colorless solid. HPLC/MS m/z: 204.15 [M+H] + , Rt (A): 0.84 min.

A3: 3-(4-methyl-1,3-oxazol-2-yl)benzoic acid A3.1: To a stirred solution of [3-(methoxycarbonyl)phenyl]boronic acid (475.0 mg;

2.639 mmol) and 2-bromo-4-methyl-1,3-oxazole (427.5 mg; 2.639 mmol) in THF (5.0 mL) and water (0.50 mL) were added sodium carbonate (559.5 mg; 5.279 mmol) and Pd(dppf)Cl2.CH2Cl2 (215.5 mg; 0.264 mmol) and the resulting mixture was stirred under N2 atmosphere at 70 °C for 16 h. The reaction mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1 :1) to afford 200 mg (35%) of methyl 3-(4-methyl-1,3-oxazol-2-yl)benzoate as a yellow solid.

HPLC/MS m/z: 218.2 [M+H] + , Rt (A): 1.01 min.

A3.2: To a stirred solution of methyl 3-(4-methyl-1 ,3-oxazol-2-yl)benzoate (180.00 mg; 0.829 mmol) in THF (2.0 mL) and water (1.0 mL) was added LiOH (39.7 mg; 1.658 mmol) and the resulting mixture was stirred at room temperature for 4 h. The mixture was acidified to pH1 with 2N HCI solution and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/MeOH = 92:8) to afford 120 mg (71%) of 3-(4-methyl-1,3-oxazol-2- yl)benzoic acid as a colorless solid. HPLC/MS m/z: 204.1 [M+H] + , Rt (E): 0.76 min.

A4: 3-(1-chloro-7-isoquinolyl)-5-methyl-1 ,2,4-oxadiazole

A4.1: Nitrogen gas was bubbled through a mixture of 7-bromoisoquinolin-1-ol (0.500 g, 2.23 mmol) and zinc cyanide (0.341 g, 2.90 mmol) in DMF (12.4 mL) for 15 min. Palladium tetrakis(triphenylphosphine) (0.155 g, 0.13 mmol) was added and the mixture heated at 100 °C in a sealed vial for 16 h. The reaction mixture was diluted with brine (100 mL) and extracted with ethyl acetate (100 mL). The organic layer was washed with brine (2 x 50 mL). Aqueous layers containing some precipitated product were re-extracted with dichloromethane (2 x 50 mL). Combined organic layers were dried over anhydrous sodium sulfate, filtered, preabsorbed onto silica and purified by column chromatography (eluent: methanol/dichloromethane = 0 to 10% gradient) to afford 0.314 g (83%) of 1-hydroxyisoquinoline-7-carbonitrile.

HPLC/MS m/z: 171.05 [M+H] + , Rt (AC): 0.84 min. 1 H NMR (500 MHz, DMSO-d 6 ): 5 11.64 (s, 1 H), 8.53-8.47 (m, 1 H), 8.03 (dd, J = 8.3, 1.8 Hz, 1H), 7.84 (d, J = 8.3 Hz, 1H), 7.38 (d, J = 7.1 Hz, 1 H), 6.65 (d, J = 7.2 Hz, 1H).

A4.2: 1-Hydroxyisoquinoline-7-carbonitrile (0.215 g, 1.26 mmol) and hydroxylamine hydrochloride (0.176 g, 2.52 mmol) in 1-butyl-3-methylimidazolium acetate (1.3 mL) were heated at 80 °C for 30 min. Water (50 mL) was added and the resulting precipitate filtered, washed with water (50 mL) and dried to afford 0.235 g (92%) of N',1-dihydroxyisoquinoline-7-carboxamidine. HPLC/MS m/z: 204.07 [M+H] + , Rt (AC): 0.36 min.

A4.3: N',1-Dihydroxyisoquinoline-7-carboxamidine (0.235 g, 1.16 mmol) and acetic anhydride (0.13 mL, 1.39 mmol) in acetonitrile (4.6 mL) were heated at 180 °C for 10 min by microwave irradiation. Water (25 mL) was added, and the resulting precipitate filtered, washed with water (25 mL), diethyl ether (2 x 10 mL) and dried to afford 0.180 g (68%) of 7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1-ol. HPLC/MS m/z: 228.08 [M+H] + , Rt (AC): 1.07 min.

A4.4: 7-(5-Methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1-ol (50 mg, 0.22 mmol) in phosphorus oxychloride (2 mL) was heated at 100 °C for 1 h. The mixture was concentrated to afford 0.054 g (100%) of 3-(1-chloro-7-isoquinolyl)-5-methyl-1 ,2,4- oxadiazole. HPLC/MS m/z: 246/248 Cl split [M+H] + , Rt (AC): 1.32 min.

A5: 5-Methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1,2,4-oxadiazole A5.1 : lsoquinoline-7-carbonitrile (4.00 g, 25.9 mmol), triethylamine (7.23 mL, 51.9 mmol) and [bmim]OAc (26 mL) were mixed and heated to 80 °C. Hydroxylamine hydrochloride (3.61 g, 51.9 mmol) was added. The reaction mixture was continued to stir at 80 °C for 1.5 h. The reaction mixture was cooled to room temperature and mixed thoroughly with EtOAc (250 mL). The emulsion was then mixed with water (750 mL). The layers were separated, and the aqueous layer was further extracted with EtOAc (5 x 100 mL). The combined organic layer was washed with saturated NaCI (3 x 100 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure to yield 3.70 g (76%) of N'-hydroxyisoquinoline-7-carboxamidine as an off-white solid. HPLC/MS m/z: 188.0829 [M+H] + , Rt (AD): 0.37 min.

A5.2: Four individual 20 mL microwave vials were each charged with a quarter of the following: N'-hydroxyisoquinoline-7-carboxamidine (3.67 g, 19.6 mmol) was mixed with acetonitrile (40 mL) and acetic anhydride (2.2 mL, 23.5 mmol) under an argon atmosphere. Each portion of the reaction mixture was heated at 180 °C under microwave irradiation for 10 min. The combined reaction mixture was evaporated onto silica gel and purified by flash chromatography (20-80% EtOAc in cyclohexane) to yield 3.59 g (87%) of 3-(7-isoquinolyl)-5-methyl-1 ,2,4-oxadiazole as an off-white solid. HPLC/MS m/z: 212.1 [M+H] + , Rt (AC): 0.88 min.

A5.3: 3-(7-isoquinolyl)-5-methyl-1 ,2,4-oxadiazole (3.59 g, 17.0 mmol) was suspended in anhydrous chloroform (57 mL) under an argon atmosphere and cooled in an ice bath. 3-Chloroperoxybenzoic acid (4.57 g, 20.4 mmol) was added. The stirred reaction mixture was allowed to warm to ambient temperature and continued to stir overnight. Potassium carbonate (9.40 g, 68.0 mmol) was added. The mixture was stirred at room temperature for 4 h before filtering through a pad of anhydrous MgSCL. The filtrate was concentrated under reduced pressure to yield 5- methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole (3.12 g, 81%) as an off- white powder. HPLC/MS m/z: 228.0713 [M+H] + , Rt (AH): 1.73 min.

B1 : Ethyl 2-(2-aminoethyl)-3-oxo-2,3-dihydro-1 H-isoindole-5-carboxylate dihydrochloride

B1.1: To a stirred solution of methyl 5-bromo-2-(bromomethyl)benzoate (28.5 g; 92.542 mmol) and tert-butyl N-(2-aminoethyl)carbamate (62.3 g; 388.676 mmol) in MeOH (400.0 mL) was added triethylamine (18.7 g; 185.08 mmol) at room temperature and the resulting mixture was stirred at 40 °C for 16 h. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1 :1) to yield 32 g (96%) of tert-butyl N-[2-(6-bromo-1-oxo-2,3-dihydro-1 H-isoindol-2- yl)ethyl]carbamate as an off-white solid. HPLC/MS m/z: 355.1-357.1 [M+H] + , Rt (A): 0.84 min.

B1.2: To a stirred solution of tert-butyl N-[2-(6-bromo-1-oxo-2,3-dihydro-1H-isoindol- 2-yl)ethyl]carbamate (1.59 g, 4.476 mmol) and triethylamine (930.7 pL, 6.714 mmol) in MeOH (18.0 mL) and THF (52.0 mL) in a pressure tank was added Pd(dppf)Cl2.CH2Cl2 (218.4 mg. 0.273 mmol) and 1,1-bis-(diphenylphosphino)- ferrocen (198.5 mg, 0.358 mmol). The mixture was purged with nitrogen for 3 min and then pressurized to 4.1 bar with carbon monoxide at 100 °C overnight. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The resulting mixture was concentrated under vacuum. The residue was purified by column chromatography to give 1.32 g (88%) of methyl 2-(2-{[(tert- butoxy)carbonyl]amino}ethyl)-3-oxo-2,3-dihydro-1H-isoindole- 5-carboxylate as a red foam. HPLC/MS m/z: 234.9 [M+H-Boc] + , Rt (G): 1.38 min.

B1.3: Methyl 2-(2-{[(tert-butoxy)carbonyl]amino}ethyl)-3-oxo-2,3-dihydro- 1 H- isoindole-5-carboxylate (469.0 mg, 1.403 mmol) was suspended in dioxane (4.8 mL) and a solution of HCI in dioxane ((4.0 M, 2.1 mL) was added. A clear solution was formed which turned into a suspension after 5 min. The reaction was stirred at room temperature for overnight. A solution of HCI in dioxane ((4.0 M, 0.7 mL) was added, the reaction mixture was stirred at room temperature for overnight and evaporated to dryness to give 422.0 mg (98%) of methyl 2-(2-aminoethyl)-3-oxo-2,3-dihydro-1H- isoindole- 5-carboxylate dihydrochloride as a pale-pink solid. HPLC/MS m/z: 234.9 [M+H] + , Rt (G): 0.96 min.

B2: 2-(2-Aminoethyl)-6-bromo-3H-isoindol-1-one

To a stirred solution of methyl 5-bromo-2-(bromomethyl)benzoate (22.8 g, 74.034 mmol) and ethane-1,2-diamine (22.3 g, 370.906 mmol) in methanol (26.00 mL) was added triethylamine (12.4 g, 123.024 mmol) at room temperature, and the resulting mixture was stirred at 40 °C for 4 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: chloroform/methanol - 75:25) to afford 15 g (79%) of 2-(2-aminoethyl)-6-bromo-3H-isoindol-1-one as an off-white solid. HPLC/MS m/z: 255.05 [M+H] + , Rt (J): 0.47 min.

B3: N1-[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]ethane-1 ,2-diamine

1-Chloro-7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinoline [Intermediate A4](900.0 mg, 3.664 mmol) and ethylendiamine (3.71 mL, 54.953 mmol) were suspended in a 5 ml microwave vessel in dry 1-Methyl-2-pyrrolidinone (16 mL) and the mixture was heated under microwave irradiation at 160 °C for 45 min. The reaction mixture was diluted with saturated aqueous NaHCOs-solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. After addition of ethyl acetate and n-heptane to the residue a precipitate was formed, which was filtered with suction, rinsed with n- heptane, and dried under high vacuum to give 726 mg (74%) of N1-[7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]ethane-1 ,2-diamine as a light-yellow solid.

HPLC/MS m/z: 270.0 [M+H] + , Rt (G): 0.90 min.

Example 1: Methyl 2-(2-{[3-(2-methyl-2H-1 ,2,3,4-tetrazol-5- yl)phenyl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5- carboxylate

Intermediate B1 (422.0 mg, 1.374 mmol) and N-Ethyldiisopropylamine (532.7 mg, 4.122 mmol) was dissolved in DMF (5.8 mL). 3-(2-methyl-2H-1,2,3,4-tetrazol-5- yl)benzoic acid (280.6 mg, 1.374 mmol) and [Dimethylamino-([1,2,3]triazolo[4,5- b]pyridin-3-yloxy)-methylene]-dimethyl-ammonium; hexafluoro phosphate (574.7 mg, 1.511 mmol) were added and the mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with a mixture of water and saturated aqueous NaHCOs-solution (1:2, 60 mL) and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash-chromatography to yield 577 mg (100%) as a pale-red solid. HPLC/MS m/z: 421.9 [M+H] + , Rt (G): 1.33 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.86 (t, J = 5.9 Hz, 1 H), 8.45 (t, J = 1.8 Hz, 1 H), 8.18-8.17 (m, 1H), 8.17-8.16 (m, 1H), 8.14-8.13 (m, 1 H), 7.93-7.91 (m, 1 H), 7.79-7.77 (m, 1H), 7.64 (t, J = 7.7 Hz, 1 H), 4.66 (s, 2H), 4.44 (s, 3H), 3.88 (s, 3H), 3.76 (t, J = 6.0 Hz, 2H), 3.61-3.57 (m, 2H).

Example 2: Ethyl 2-(2-{[3-(2-methyl-2H-1 ,2,3,4-tetrazol-5- yl)phenyl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5- carboxylate

Methyl 2-(2-{[3-(2-methyl-2H-1,2,3,4-tetrazol-5-yl)phenyl]formamido }ethyl)-3-oxo- 2,3-dihydro-1H-isoindole-5-carboxylate (73.6 mg. 0.175 mmol) was suspended in dry ethanol (4.90 mL). A solution of HCI in dioxane ((4.0 M, 1.09 mL, 4.377 mmol) was added. A clear solution was formed, which was stirred at 55 °C for 60 h, cooled to room temperature and evaporated to dryness. The residue was purified by chromatography to afford 56 mg (74%) as a colorless solid. HPLC/MS m/z: 434.9 [M+H] + , Rt (G): 1.42 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.86 (t, J = 5.9 Hz, 1 H), 8.45 (t, J = 1.8 Hz, 1H), 8.18-8.16 (m, 2H), 8.14-8.13 (m, 1 H), 7.93-7.91 (m, 1H), 7.79-7.77 (m, 1H), 7.64 (t, J = 7.7 Hz, 1H), 4.66 (s, 2H), 4.44 (s, 3H), 4.33 (q, J = 7.1 Hz, 2H), 3.76 (t, J = 6.0 Hz, 2H), 3.59 (q, J = 5.9 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H).

Example 3: Propan-2-yl 2-(2-{[3-(2-methyl-2H-1 ,2,3,4-tetrazol-5- yl)phenyl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5- carboxylate

Example 3.1: Methyl 2-(2-{[3-(2-methyl-2H-1 ,2,3,4-tetrazol-5- yl)phenyl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5- carboxylate (137.9 mg, 0.328 mmol) was dissolved in THF (6.60 mL) and water (3.30 mL). Lithium hydroxide (19.7 mg, 0.821 mmol) was added, and the reaction mixture was stirred at room temperature for 2 h. THF was removed in vacuo, the aqueous residue was diluted with water (20 mL)and acidified to pH 3-4 with aqueous HCI-solution. A solid was formed, which was filtered by suction, washed with demineralized water, and dried under vacuum to give 116 mg (86%) of 2-(2-{[3-(2-methyl-2H-1,2,3,4-tetrazol- 5-yl)phenyl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole- 5-carboxylic acid as a pale-beige solid. HPLC/MS m/z: 407.9 [M+H] + , Rt (H): 0.79 min.

Example 3.2: 2-(2-{[3-(2-methyl-2H-1 ,2,3,4-tetrazol-5-yl)phenyl]formamido}ethyl)-3- oxo-2, 3-dihydro-1H-isoindole-5-carboxylic acid (115.0 mg, 0.283 mmol), 4- (dimethylamino)-pyridin (13.8 mg, 0.113 mmol) and DCC (93.3 mg, 0.452 mmol) were dissolved in dry THF (0.7 mL) and dry DMF (0.4 mL). 2-Propanol (216 pL, 2.826 mmol) was added and the reaction was stirred at room temperature overnight. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash-chromatography to give 88 mg (69%) of the title compound as a colorless solid. HPLC/MS m/z: 449.9 [M+H] + , Rt (G): 1.49 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.86 (t, J = 5.9 Hz, 1 H), 8.45 (t, J = 1.7 Hz, 1 H), 8.18-8.16 (m, 1 H), 8.15 (dd, J = 7.9, 1.6 Hz, 1 H), 8.13-8.12 (m, 1 H), 7.93-7.91 (m, 1 H), 7.78-7.76 (m, 1 H), 7.64 (t, J = 7.7 Hz, 1 H), 5.15 (quint, J = 6.3 Hz, 1 H), 4.66 (s, 2H), 4.44 (s, 3H), 3.76 (t, J = 5.9 Hz, 2H), 3.59 (q, J = 5.9 Hz, 2H), 1.33 (d, J = 6.2 Hz, 6H).

Example 4: tert-Butyl 2-(2-{[3-(2-methyl-2H-1 ,2,3,4-tetrazol-5- yl)phenyl]formamido}ethyl)-3-oxo-2,3-dihydro-1 H-isoindole-5-carboxylate

A mixture of intermediate 3.1. (37.8 mg, 0.093 mmol) and O-(tert-Butyl)-N,N'- diisopropylisourea (93.7 mg, 0.465 mmol) in dry toluene (0.7 mL) was stirred at 90 °C for 16 h. The reaction mixture was cooled to room temperature, diluted with water, and extracted with dichloromethane. The combined organic layers were dried with sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by RP flash-chromatography to yield 28 mg (65%) of the title compound as a colorless solid. HPLC/MS m/z: 463.9 [M+H] + , Rt (H): 0.93 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.86 (t, J = 5.8 Hz, 1 H), 8.46 (t, J = 1.8 Hz, 1 H), 8.19-8.16 (m, 1 H), 8.11 (dd, J = 7.9, 1.6 Hz, 1 H), 8.08-8.07 (m, 1 H), 7.93-7.91 (m, 1 H), 7.75 (d, J = 8.0 Hz, 1 H), 7.64 (t, J = 7.8 Hz, 1 H), 4.65 (s, 2H), 4.44 (s, 3H), 3.76 (t, J = 6.0 Hz, 2H), 3.59 (q, J = 5.9 Hz, 2H), 1.56 (s, 9H).

Example 5: Ethyl 2-(2-{[3-(3-methyl-1 ,2-oxazol-5-yl)phenyl]formamido}ethyl)-3-oxo- 2,3-dihydro-1 H-isoindole-5-carboxylate

To a solution of intermediate A1 (79.8 mg; 0.393 mmol) and intermediate B1 (97.5 mg; 0.393 mmol) in DMF (2.0 mL) were added 1-methyl-1 H-imidazole (64.5 mg; 0.785 mmol) and TCFH (165.3 mg; 0.589 mmol) and the mixture was stirred at room temperature for 2 h. The reaction mixture was evaporated, and the residue purified by RP flash-chromatography to afford 77 mg (45%) of the title compound as an off- white solid. HPLC/MS m/z: 434.15 [M+H] + , Rt (D): 1.09 min. 1 H NMR (300 MHz, DMSO-d 6 ): 58.69 (t, J= 5.8 Hz, 1H), 8.10-7.99 (m, 3H), 7.83 (dt, J= 7.8, 1.4 Hz, 1H), 7.73 (dt, J= 7.9, 1.4 Hz, 1H), 7.65 (d, J= 7.9 Hz, 1H), 7.48 (t, J= 7.8 Hz, 1H), 6.78 (s, 1H), 4.54 (s, 2H), 4.28-4.15 (m, 2H), 3.68-3.58 (m, 2H), 3.52-3.41 (m, 2H), 2.18 (s, 3H), 1.22 (t, J= 7.1 Hz, 3H).

Example 6: Ethyl 2-(2-{[3-(5-methyl-1,3-oxazol-2-yl)phenyl]formamido}ethyl)-3 -oxo- 2,3-dihydro-1H-isoindole-5-carboxylate

Preparation as described for example 5 using A2 (100.00 mg; 0.49 mmol) and B1 (122.1 mg; 0.49 mmol). Purification by reverse flash chromatography. Yield: 24 mg (11%) colorless solid. HPLC/MS m/z: 434.15 [M+H] + , Rt (F): 1.55 min. 1 H NMR (400 MHz, DMSO-d 6 ): 58.83 (t, J= 5.8 Hz, 1H), 8.30 (t, J= 1.8 Hz, 1H), 8.20-8.12 (m, 2H), 8.04 (dt, J= 7.8, 1.4 Hz, 1H), 7.86 (dt, J= 7.9, 1.4 Hz, 1H), 7.81-7.74 (m, 1H), 7.59 (t, J= 7.8 Hz, 1H), 7.02 (t, J= 1.3 Hz, 1H), 4.66 (s, 2H), 4.38-4.29 (m, 2H), 3.79-3.72 (m, 2H), 3.63-3.54 (m, 2H), 2.39 (d, J= 1.3 Hz, 3H), 1.34 (t, J= 7.1 Hz, 3H).

Example 7: Ethyl 2-(2-{[3-(4-methyl-1,3-oxazol-2-yl)phenyl]formamido}ethyl)-3 -oxo- 2,3-dihydro-1H-isoindole-5-carboxylate

Preparation as described for example 5 using A3 (100.0 mg; 0.49 mmol) and B1 (122.2 mg; 0.49 mmol). Purification by reverse flash chromatography. Yield: 48 mg (23%) colorless solid. HPLC/MS m/z: 434.15 [M+H] + , Rt (C): 0.90 min. 1 H NMR (400 MHz, DMSO-d 6 ): 58.84 (t, J =5.9 Hz, 1H), 8.33 (t, J = 1.8 Hz, 1H), 8.20-8.11 (m, 2H), 8.05 (dt, J= 7.8, 1.4 Hz, 1H), 7.93 (q, J= 1.2 Hz, 1H), 7.88 (dt, J= 7.8, 1.4 Hz, 1H), 7.77 (dd, J= 7.9, 0.8 Hz, 1H), 7.59 (t, J= 7.8 Hz, 1H), 4.66 (s, 2H), 4.39-4.29 (m, 2H), 3.79-3.72 (m, 2H), 3.63-3.54 (m, 2H), 2.17 (d, J = 1.3 Hz, 3H), 1.34 (t, J =

7.1 Hz, 3H).

Example 8: Ethyl 2-(2-{[3-(5-methyl-1 ,2,4-oxadiazol-3-yl)phenyl]formamido}ethyl)-3- oxo-2, 3-dihydro-1H-isoindole-5-carboxylate

Example 8.1. To a stirred solution of intermediate B2 (599.3 mg, 2.349 mmol) and 3- (5-methyl-1,2,4-oxadiazol-3-yl)benzoic acid (575.6 mg, 2.819 mmol) in DMF (6.0 mL) was added 1-methyl-1 H-imidazole (578.6 mg, 7.047 mmol) at room temperature. After stirring the mixture for 15 min at room temperature [chloro(dimethylamino)methylidene]dimethylazanium; hexafluoro- 5 -phosphanuide (791.0 mg, 2.819 mmol) was added at room temperature, and the mixture was stirred for 2 h. The reaction mixture was concentrated under vacuum, and the residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 9:1) to afford 666 mg (64%) of N-[2-(6-bromo-1-oxo- 2,3-dihydro-1 H-isoindol-2-yl)ethyl]-3-(5-methyl-1 ,2,4-oxadiazol-3-yl)benzamide as a colorless solid. HPLC/MS m/z: 441.05 [M+H] + , Rt (J): 0.66 min.

Example 8.2. To a solution of intermediate 8.1 (174.0 mg, 0.394 mmol) and triethylamine (190.00 pL, 1.299 mmol) in EtOH (16.0 mL) was added Pd(dppf)Cl2.CH2Cl2 (35.1 mg, 0.043 mmol) in a pressure tank. The mixture was purged with nitrogen for 3 min and then was pressurized to 30 atm with carbon monoxide at 100°C for 32 h. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids, and the filtrate was concentrated under vacuum. The crude product was purified by prep-HPLC to give 29 mg (17%) of ethyl 2-(2-{[3-(5-methyl-1,2,4-oxadiazol-3-yl)phenyl]formamido}eth yl)-3-oxo-2,3-dihydro- 1H-isoindole-5-carboxylate as a colorless solid. HPLC/MS m/z: 435.20 [M+H] + , Rt (I): 1.85 min. 1 H NMR (300 MHz, DMSO-cfe): 6 8.87 (t, J = 5.8 Hz, 1H), 8.39 (t, J = 1.7 Hz, 1 H), 8.22-8.07 (m, 3H), 8.01-7.92 (m, 1H), 7.78 (d, J = 8.0 Hz, 1 H), 7.65 (t, J = 7.8 Hz, 1H), 4.66 (s, 2H), 4.34 (q, J = 7.1 Hz, 2H), 3.76 (t, J = 5.9 Hz, 2H), 3.59 (q, J = 5.9 Hz, 2H), 2.68 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

The following examples were prepared in an analogous manner: Example 9: Ethyl 2-(2-{[6-(5-methyl-1,2,4-oxadiazol-3-yl)pyridin-2- yl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5-carboxy late

36 mg colorless solid. HPLC/MS m/z: 436.9 [M+H] + , Rt (G): 1.42 min. 1 H NMR (700 MHz, DMSO-d 6 ): 58.83 (t, J= 6.3 Hz, 1H), 8.21 (dd, J=7.7, 1.3 Hz, 1H), 8.19-8.17 (m, 1H), 8.17-8.16 (m, 1H), 8.13 (dd, J=7.7, 1.3 Hz, 1H), 8.12-8.11 (m, 1H), 7.78- 7.76 (m, 1H), 4.69 (s, 2H), 4.33 (q, J= 7.1 Hz, 2H), 3.79 (t, J= 5.9 Hz, 2H), 3.65 (q, J= 6.1 Hz, 2H), 2.70 (s, 3H), 1.34 (t, J= 7.1 Hz, 3H).

Example 10: Propan-2-yl 2-(2-{[6-(5-methyl-1,2,4-oxadiazol-3-yl)pyridin-2- yl]formamido}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5-carboxy late

60 mg colorless solid. HPLC/MS m/z: 450.9 [M+H] + , Rt (G): 1.49 min. 1 H NMR (700 MHz, DMSO-d 6 ): 58.82 (t, J= 6.2 Hz, 1H), 8.21 (dd, J=7.7, 1.3 Hz, 1H), 8.18 (t, J = 7.7 Hz, 1H), 8.15 (dd, J= 7.9, 1.6 Hz, 1H), 8.13 (dd, J=7.7, 1.3 Hz, 1H), 8.11- 8.10 (m, 1H), 7.77-7.75 (m, 1H), 5.18-5.12 (m, 1H), 4.69 (s, 2H), 3.79 (t, J= 5.9 Hz, 2H), 3.65 (q, J = 6.1 Hz, 2H), 2.70 (s, 3H), 1.33 (d, J= 6.2 Hz, 6H).

Example 11: Ethyl 2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2,3-dihydro-1H-isoindole-5-carboxylate

Example 11.1: To a stirred solution of intermediate B2 (1.00 g, 3.920 mmol) and 1- chloro-7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinoline (0.96 g, 3.920 mmol) in NMP (8.0 mL) was added diethylamine (1.45 g, 11.181 mmol) at room temperature. The resulting mixture was heated to 160 °C and stirred for 16 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by RP flash-chromatography to afford 1.2 g (66%) of 6-bromo-2-(2-[[7-(5- methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino]ethyl)-3H-isoindo l-1-one as a yellow solid. HPLC/MS m/z: 466.10 [M+H] + , Rt (J): 0.62 min.

Example 11.2: To a solution of 6-bromo-2-(2-[[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino]ethyl)-3H-isoindol-1-one (48.8 mg, 0.105 mmol) in ethanol (6.0 mL) was added Pd(dppf)Cl2.CH2Cl2 (10.6 mg, 0.013 mmol), triethylamine (28.5 mg, 0.282 mmol) in a pressure tank. The mixture was purged with nitrogen for 1 min and then was pressurized to 30 atm with carbon monoxide at 100 °C for 16 h. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 1 :1) and by preparative HPLC to give 18 mg (38%) of the title compound as a colorless solid. HPLC/MS m/z: 458.0 [M+HJ+, Rt (K): 0.93 min. 1 H NMR (300 MHz, DMSO-d6): 5 8.83 (s, 1 H), 8.19-8.12 (m, 3H), 8.04 (d, J = 5.6 Hz, 1 H), 7.94 (d, J = 5.7 Hz, 1 H), 7.85 (d, J = 8.6 Hz, 1 H), 7.76 (d, J = 7.9 Hz, 1 H), 6.95 (d, J = 5.8 Hz, 1 H), 4.69 (s, 2H), 4.34 (q, J = 7.1 Hz, 2H), 3.92-3.74 (m, 4H), 2.68 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

Example 12: Ethyl 2-(2-{[7-(1-methyl-1 H-pyrazol-4-yl)isoquinolin-1-yl]amino}ethyl)-3- oxo-2, 3-dihydro-1 H-isoindole-5-carboxylate

Preparation in an analogous manner. 28 mg off-white solid. HPLC/MS m/z: 456.15 [M+H] + , Rt (L): 0.59 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.33 (s, 1 H), 8.19-8.10 (m, 3H), 7.95 (s, 1 H), 7.84-7.70 (m, 3H), 7.66 (d, J = 8.5 Hz, 1 H), 7.58 (s, 1 H), 6.83 (d, J = 5.8 Hz, 1 H), 4.68 (s, 2H), 4.32 (q, J = 7.1 Hz, 2H), 3.92-3.73 (m, 7H), 1.32 (t, J = 7.1 Hz, 3H).

Example 13: Propan-2-yl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2,3-dihydro-1 H-isoindole-5-carboxylate

Example 13.1 : To a stirred solution of intermediate 11.1. (480.0 mg, 1.034 mmol) and formyl acetate (0.85 mL, 10.269 mmol) in DMF (6.0 mL) were added triethylamine (0.76 mL, 5.18 mmol) and Pd(dppf)Cl2.CH2Cl2 (64.5 mg, 0.079 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90 °C under nitrogen atmosphere. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by RP flash-chromatography to afford 292 mg (66%) of 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol- 3-yl)isoquinolin-1-yl]amino}ethyl)-3-oxo-2,3-dihydro-1 H-isoindole-5-carboxylic acid as a yellow solid. HPLC/MS m/z: 430.15 [M+H] + , Rt (J): 0.57 min.

Example 13.2: A solution of intermediate 12.1. (87.6 mg, 0.204 mmol) in H2SO4 (0.25 mL, 4.660 mmol) and 2-propanol (1.0 mL) was stirred at 80 °C for 3 h. The mixture was cooled to room temperature, rendered basic to pH8 with saturated aqueous NaHCOs solution, and extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by prep-HPLC to afford 32 mg (33%) of the title compound as a colorless solid. HPLC/MS m/z: 472.10 [M+H] + , Rt (B): 0.76 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.82 (s, 1 H), 8.18-8.08 (m, 3H), 8.03 (t, J = 5.6 Hz, 1 H), 7.93 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.75 (d, J = 7.8 Hz, 1 H), 6.94 (d, J = 5.7 Hz, 1 H), 5.22-5.08 (m, 1 H), 4.68 (s, 2H), 3.87 (t, J = 5.7 Hz, 2H), 3.81 (t, J = 5.5 Hz, 2H), 2.67 (s, 3H), 1 .33 (d, J = 6.3 Hz, 6H).

Example 14: tert-Butyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2,3-dihydro-1 H-isoindole-5-carboxylate

To O-(tert-Butyl)-N,N'-diisopropylisoure (1.0 mL) was added intermediate 12.1. (87.6 mg, 0.204 mmol) and the mixture was stirred at 60 °C for 3 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The crude product was purified by prep. HPLC to give 23 mg (23%) of the title compound as a colorless solid. HPLC/MS m/z: 472.10 [M+H] + , Rt (B): 0.79 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.82 (d, J = 1.5 Hz, 1 H), 8.17-8.08 (m, 2H), 8.06-8.01 (m, 2H), 7.93 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.73 (d, J = 7.9 Hz, 1 H), 6.94 (d, J = 5.7 Hz, 1 H), 4.67 (s, 2H), 3.86 (t, J = 5.7 Hz, 2H), 3.83-3.77 (m, 2H), 2.67 (s, 3H), 1.55 (s, 9H).

Example 15: 6-(Difluoromethoxy)-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-

1-yl]amino}ethyl)-2,3-dihydro-1 H-isoindol-1-one

Example 15.1 : To stirred suspension of methyl 2-(bromomethyl)-5- methoxybenzoate (200.0 mg, 0.772 mmol) and intermediate B3 (228.7 mg, 0.849 mmol) in dry methanol (4.8 mL) was added triethylamine (321.0 pL, 2.316 mmol). The reaction mixture was heated to 45 °C while a clear solution was formed, which was stirred for 90 min at this temperature. The reaction mixture was cooled to room temperature and evaporated to dryness. The residue was purified by RP flashchromatography to obtain 137 mg (43%) of 6-methoxy-2-(2-{[7-(5-methyl- 1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-2,3-dihydro-1 H-isoindol-1-one as a beige solid. HPLC/MS m/z: 415.9 [M+H] + , Rt (H): 0.71 min.

Example 15.2: 6-methoxy-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-2,3-dihydro-1 H-isoindol-1-one (124.0 mg, 0.293 mmol) was suspended in dry dichloromethane (4.7 mL). Boron tribromide (142.4 pL, 1.463 mmol) was dissolved in dry dichloromethane (1.9 mL), added slowly to the suspension, and the mixture was stirred at room temperature for 4 h. The reaction mixture was poured onto a 1 :1 mixture of water/aqueous saturated NaHCOs-solution (60 mL) and extracted with dichloromethane. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and evaporated to dryness. The residue was purified by chromatography to afford 63 mg (54%) of 6-hydroxy-2-(2- {[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-2,3-dihydr o-1 H- isoindol-1-one as a beige solid. HPLC/MS m/z: 401.9 [M+H] + , Rt (H): 0.67 min. Example 15.3: Intermediate 15.2 (53.0 mg, 0.132 mmol) and cesium carbonate (64.5 mg, 0.198 mmol) were suspended in DMF (770.0 pL). Methyl 2-chloro-2,2- difluoroacetate (19.5 pL, 0.185 mmol) was added and the mixture was stirred at 80 °C for 3 h. Further methyl 2-chloro-2,2-difluoroacetate (14.0 pL, 0.132 mmol) and cesium carbonate (43.5 mg, 0.132 mmol) were added, and the reaction was stirred at 80 °C overnight. The reaction mixture was cooled to room temperature, filtered, and evaporated to dryness. The residue was purified by RP flash- chromatography to give 21 mg (35%) of the title compound as a colorless solid. HPLC/MS m/z: 451.8 [M+H] + , Rt (M): 1.78 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.84-8.82 (m, 1 H), 8.14 (dd, J = 8.4, 1.5 Hz, 1 H), 8.03 (t, J = 5.6 Hz, 1H), 7.95 (d, J = 5.7 Hz, 1H), 7.84 (d, J = 8.5 Hz, 1H), 7.65 (d, J = 8.1 Hz, 1 H), 7.40 (d, J = 2.4 Hz, 1H), 7.38 (dd, J = 8.2, 2.4 Hz, 1 H), 7.32 (t, J = 73.9 Hz, 1H), 6.95 (d, J = 5.7 Hz, 1 H), 4.58 (s, 2H), 3.87- 3.84 (m, 2H), 3.81-3.78 (m, 2H), 2.68 (s, 3H).

Example 16: 6-(difluoromethoxy)-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl) isoquinolin- 1-yl]amino}ethyl)-2,3-dihydro-1H-isoindol-1-one

Example 16.1: Methyl 2-bromo-5-(trifluoromethoxy)benzoate (1.43 g, 4.782 mmol) and zinc cyanide (2.28 g, 19.417 mmol) were dissolved in dry DMF (22.0 mL) under nitrogen atmosphere. Tetrakis(triphenylphosphine)palladium(0) (551.2 mg; 0.477 mmol) was added and the reaction mixture was heated to 120 °C and stirred for 1 h. After cooling down to room temperature, the reaction mixture was diluted with water and extracted with dichloromethane. The combined organic layers were washed with water and brine, dried over sodium sulfate, filtered, and concentrated under vacuum. The crude product was purified by flash-chromatography to yield 951 mg (81 %) of methyl 2-cyano-5-(trifluoromethoxy)benzoate as a colorless solid.

HPLC/MS m/z: 245.9 [M+H] + , Rt (G): 1.59 min.

Example 16.2: To a solution of methyl 2-cyano-5-(trifluoromethoxy)benzoate (742.0 mg, 3.027 mmol) in methanol (10.0 mL) was added Pd/C (5%, 800 mg) and HCI solution (0.76 mL, 32%) in a pressure tank. The mixture was hydrogenated at room temperature under 3.6 bar of hydrogen pressure over night, filtered and evaporated to dryness. The residue (824 mg, 95%) was used in the next step without further purification. Example 16.3: Intermediate 16.2 (824.0 mg, 2.885 mmol) was dissolved in methanol (25.0 mL) and treated with an aqueous NaOH solution (7N, 5 mL). The reaction was stirred at room temperature for 20 min. The reaction mixture was concentrated under reduced pressure, the aqueous residue was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and evaporated to dryness to give 571 mg (91 %) 6- (trifluoromethoxy)-2,3-dihydro-1 H-isoindol-1-one as colorless crystals. HPLC/MS m/z: 217.9 [M+H] + , Rt (G): 1.32 min.

Example 16.4: Intermediate 16.3 (97.0 mg, 0.447 mmol) was dissolved in THF (3.6 mL) and treated with sodium hydride (60% dispersion in mineral oil; 53.6 mg, 1.340 mmol) while cooling in an ice bath under nitrogen atmosphere. The suspension was stirred for 45 min. Bromoacetonitrile (214.3 mg, 1.787 mmol) was added and the reaction mixture was allowed to warm to room temperature and stirred for 15 min. The reaction was quenched by the addition of water and the mixture extracted with ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuum. The residue was purified by RP flashchromatography to yield 65 mg (57%) of 2-[1-oxo-6-(trifluoromethoxy)-2,3-dihydro- 1 H-isoindol-2-yl]acetonitrile as an orange solid. HPLC/MS m/z: 256.9 [M+H] + , Rt (G): 1.42 min.

Example 16.5: To a solution of intermediate 16.4 (65 mg, 0.254 mmol) in methanol (3.0 mL) was added Pd/C (5%, 100 mg) and HCI solution (64 pL, 32%) in a pressure tank. The mixture was hydrogenated at room temperature under 3.2 bar of hydrogen pressure for 17 h, filtered and evaporated to dryness. The crude product was purified by RP flash-chromatography to afford 24 mg (31%) of 2-[1-oxo-6- (trifluoromethoxy)-2,3-dihydro-1 H-isoindol-2-yl]ethan-1-ammonium formate as colourless solid. HPLC/MS m/z: 260.9 [M+H] + , Rt (G): 1.16 min.

Example 16.6: 7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-2-ium-2olate (17.0 mg, 0.075 mmol) and intermediate 16.5 (24.1 mg, 0.079 mmol) were dissolved in dry dichloromethane (0.4 mL) under an argon atmosphere. DIPEA (85.9 pL, 0.505 mmol) and bromo-tripyrrolidinophosphonium-hexafluorphosphate (52.2 mg, 0.112 mmol) were added and the reaction mixture was stirred at room temperature for 3 d. The reaction mixture was evaporated under reduced pressure and the crude product was purified by RP flash-chromatography to give 11 mg (31%) of 2-(2-{[7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-6-(trifluo romethoxy)-2,3-dihydro- 1 H-isoindol-1-one as a colorless solid. HPLC/MS m/z: 470.8 [M+H] + , Rt (M): 1.87 min.

Example 17: 6-(2,3-Difluoropropoxy)-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-2,3-dihydro-1 H-isoindol-1-one

Example 17.1 : Methyl 5-hydroxy-2-methylbenzoate (122.5 mg, 0.737 mmol), 2,3- difluoropropan-1-ol (141.6 mg, 1.474 mmol) and triphenylphosphine (271.0 mg, 1.033 mmol) were dissolved in dry dichloromethane (2.2 mL) and the solution was cooled to 0 °C. Di-tert-butyl azodicarboxylate (238.0 mg, 1.033 mmol) was added and the light-yellow solution was stirred at 0 °C for 10 min and then overnight at room temperature. Triphenylphosphine (97.0 mg, 0.370 mmol) and di-tert-butyl azodicarboxylate (85.2 mg, 0.370 mmol) were added, the mixture was heated to 40 °C and stirred for 4.5 h. The reaction mixture was diluted with dichloromethane, washed with water, dried with sodium sulfate, filtered, and evaporated to dryness. The oily residue was purified by flash chromatography to yield 170 mg (94%) of methyl 5-(2,3-difluoropropoxy)-2-methylbenzoate as a light-yellow oil. HPLC/MS m/z: 245.1 [M+H] + , Rt (N): 1.60 min.

Example 17.2: Intermediate 16.1 (205.0 mg, 0.839 mmol), NBS (149.3 mg, 0.839 mmol) and dibenzoyl peroxide (with 25% water, 13.8 mg, 0.042 mmol) were suspended in carbon tetrachloride (7.0 mL) under argon and the mixture was heated to 77 °C for 4 h. Further dibenzoyl peroxide (with 25% water, 16.0 mg, 0.049 mmol) was added and the reaction mixture stirred at 77 °C overnight. The reaction mixture was cooled to room temperature, diluted with dichloromethane (20 mL), washed with water, dried with sodium sulfate, filtered, and evaporated to dryness. The residue was purified by flash chromatography to afford 222 mg (82%) of methyl 2- (bromomethyl)-5-(2,3-difluoropropoxy)benzoate as a colorless oil. HPLC/MS m/z: 345.0 [M+Na] + , Rt (N): 1.65 min.

Example 17.3. The reaction of intermediate B3 (48.5 mg, 0.180 mmol) and intermediate 17.2 (70.0 mg, 0.217 mmol) was performed as described for intermediate 15.1. Yield: 42 mg (49%) pale-brown solid. HPLC/MS m/z: 480.2 [M+H] + , Rt (N): 1.38 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.85-8.83 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.04 (t, J = 5.6 Hz, 1H), 7.95 (d, J = 5.7 Hz, 1H), 7.84 (d, J = 8.4 Hz, 1H), 7.51-7.49 (m, 1 H), 7.19 (s, 1 H), 7.19-7.17 (m, 1H), 6.95 (d, J = 5.6 Hz, 1H), 5.20-5.07 (m, 1H), 4.85-4.67 (m, 2H), 4.51 (s, 2H), 4.39-4.33 (m, 1 H), 4.30- 4.24 (m, 1 H), 3.84 (t, J = 5.9 Hz, 2H), 3.78 (q, J = 5.8 Hz, 2H), 2.68 (s, 3H).

Example 18: 2-(2-((7-(5-Methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1-yl)ami no)ethyl)-6- propoxyisoindolin-1-one

Example 18.1: Potassium carbonate (333 mg, 2.4 mmol) was added to a mixture of 5-hydroxy-3-methyl benzoic acid methyl ester (200 mg, 1.2 mmol) and 1- iodopropane (0.24 mL, 2.4 mmol) in acetonitrile (3.3 mL) at rt. The obtained mixture was heated at reflux overnight. After 19h the reaction mixture was partitioned between ethyl acetate (60 mL) and water (40 mL). After phase separation, the organic layer was washed with brine (30 mL), dried with magnesium sulfate, filtered, and evaporated to dryness, to afford the methyl 2-methyl-5-propoxy-benzoate (220 mg, 80%, 1.0 mmol) that was taken through to the next step without further purification. HPLC/MS m/z: (doesn’t ionise), Rt (Y): 1.601 min.

Example 18.2: Methyl 2-methyl-5-propoxy-benzoate (220 mg, 1.0 mmol) was dissolved in chloroform (1.5 mL). Azobis-isobutyronitrile (AIBN) (2.6 mg, 0.14 mmol) and N-bromosuccinimide (207 mg, 1.2 mmol) were carefully added to the solution and the obtained mixture was refluxed at 65°C overnight. After 17h reaction, the reaction mixture was evaporated into silica (dry load) and purified by silica column chromatography (eluent: 0-30% ethyl acetate in cyclohexane) to afford the required product methyl 2-(bromomethyl)-5-propoxy-benzoate (230 mg, 76%, 0.8 mmol). HPLC/MS m/z: (doesn’t ionise), Rt (Y): 1.625 min.

Example 18.3: A solution of methyl 2-(bromomethyl)-5-propoxy-benzoate (230 mg, 0.8 mmol), 1-Boc-ethylenediamine (0.15 mL, 0.9 mmol) and triethylamine (0.17 mL, 1.2 mmol) in methanol (4 mL) was refluxed for 23h. After being cooled down to room temperature, the reaction mixture was concentrated and purified by silica column chromatography (eluent: 20-90% ethyl acetate in cyclohexane) to afford the tert- butyl N-[2-(1-oxo-6-propoxy-isoindolin-2-yl)ethyl]carbamate (140 mg, 53%, 0.4 mmol) as a white amorphous solid. HPLC/MS m/z: 357.2 [M+Na] + , Rt (Y): 1.493 min.

Example 18.4: Tert-butyl N-[2-(1-oxo-6-propoxy-isoindolin-2-yl)ethyl]carbamate (140 mg, 0.4 mmol) was mixed with 4 M HCI in dioxane (10 mL, 42 mmol) and 1 ,4- dioxane (10 mL) and stirred for 18 h. The volatiles were removed under reduced pressure and the obtained crude (176 mg) was dissolved in methanol, followed by purification using a SCX-II (2 g, 15 mL) cartridge and methanol and a 2M solution of ammonia in methanol as eluents. Basic fractions were combined to afford the product 2-(2-aminoethyl)-6-propoxy-isoindolin-1 -one (98 mg, 100%, 0.4 mmol) as a white amorphous powder. HPLC/MS m/z: 235.1 [M+H] + , Rt (Y): 0.946 min.

Example 18.5: 2-(2-Aminoethyl)-6-propoxy-isoindolin-1-one (40 mg, 0.17 mmol), 5- methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole [Intermediate A5] (46 mg, 0.2 mmol), PyBrop (96 mg, 0.2 mmol), N,N-diisopropylethylamine (0.11 mL, 0.6 mmol) and anhydrous dichloromethane (0.85 mL) were placed in a microwave vial at rt under nitrogen. The reaction mixture was heated at 60°C by microwave irradiation for 1 h. The volatiles were removed under reduced pressure and the obtained crude was purified directly by reverse phase flash chromatography (eluent: 20-100% MeOH in water modified with 0.1 % Formic acid) followed by purification using a SCX-II (2 g, 15 mL) cartridge and methanol and a 2M ammonia solution in methanol as eluants. Basic fractions were concentrated to dryness in vacuum to give 2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-6-propoxy- isoindolin-1-one (22 mg, 29%, 0.05 mmol) as a pale-yellow solid. HPLC/MS m/z: 444.2 [M+H] + , Rt (Y): 1.287 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.84 (br s, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.03 (t, J = 5.5 Hz, 1 H), 7.95 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.46 (dd, J = 8.1 , 0.9 Hz, 1 H), 7.15-7.08 (m, 2H), 6.95 (dd, J = 5.9, 0.8 Hz, 1 H), 4.49 (s, 2H), 3.96 (t, J = 6.6 Hz, 2H), 3.83 (t, J = 5.9 Hz, 2H), 3.78 (q, J = 5.6 Hz, 2H), 2.68 (s, 3H), 1.89-1.62 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H).

Example 19: 2-(2-{[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-6-

(trifluoromethyl)-2,3-dihydro-1 H-isoindol-1-one Example 19.1: Preparation as described for intermediate B2 using methyl 2- (bromomethyl)-5-(trifluoromethyl)benzoate (2.38 g, 8.012 mmol), ethane-1,2- diamine (2.41 g, 40.058 mmol) and triethylamine (2.43 g, 24.036 mmol) in methanol (26.00 mL). Yield: 1.69 g (86%) of 2-(2-aminoethyl)-6-(trifluoromethyl)-2,3-dihydro- 1 H-isoindol-1-one as a yellow solid. HPLC/MS m/z: 245.20 [M+H] + , Rt (B): 0.57 min. Example 19.2: Preparation as described for intermediate 11.1. Yield: 31 mg (9%) of 2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1-yl]ami no}ethyl)-6- (trifluoromethyl)-2,3-dihydro-1 H-isoindol-1-one as a yellow solid. HPLC/MS m/z: 454.00 [M+H] + , Rt (O): 1.54 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.82 (d, J = 1.6 Hz, 1H), 8.17-8.10 (m, 1H), 8.03 (t, J = 5.6 Hz, 1H), 7.99-7.90 (m, 2H), 7.90-7.81 (m, 3H), 6.97-6.91 (m, 1H), 4.70 (s, 2H), 3.92-3.84 (m, 2H), 3.82 (t, J = 5.6 Hz, 2H), 2.67 (s, 3H).

Example 20: 2-(2-{[7-(5-Methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1-yl]ami no}ethyl)-6-

[1-(trifluoromethyl)cyclopropyl]-2,3-dihydro-1 H-isoindol-1-one

Example 20.1: To a stirred solution of [3-(methoxycarbonyl)-4-methylphenyl]boronic acid (1.90 g, 9.794 mmol and 2-bromo-3,3,3-trifluoroprop-1-ene (2.91 g, 16.633 mmol) in THF (14.0 mL) and water (7.0 mL) was added CS2CO3 (4.47 g, 13.719 mmol) and Pd(dppf)Cl2.CH2Cl2 (0.40 g, 0.490 mmol). The resulting mixture was stirred for 16 h at 80 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 3:1) to afford 1.89 g (79%) of methyl 2-methyl-5-(3,3,3-trifluoroprop-1-en-2-yl)benzoate as a yellow liquid. HPLC/MS m/z: 245.15 [M+H] + , Rt (J): 0.81 min.

Example 20.2: A stirred solution of intermediate 20.1 (1.79 g, 7.329 mmol) and diphenyl methyl sulfonium tetrafluoroborate (2.74 g, 9.516 mmol) in THF (20.0 mL) was treated with sodium bis(trimethylsilyl)amide (2M in THF, 5.86 mL, 11.711 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was warmed to room temperature and stirred for 1 h. The reaction was quenched with water and the mixture extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/petroleum ether - 1 :2) to afford 851 mg (45%) of methyl 2-methyl- 5-[1-(trifluoromethyl)cyclopropyl]benzoate as a yellow liquid. HPLC/MS m/z: 259.15 [M+H] + , Rt (B): 1.09 min.

Example 20.3: To a stirred solution of intermediate 20.2 (789.0 mg, 3.055 mmol) and NBS (598.0 mg. 3.360 mmol) in CCI4 (10.0 mL) was added AIBN (100.3 mg, 0.611 mmol). The resulting mixture was stirred at 70 °C for 2 h. The resulting mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 1 :3) to afford 526 mg (51%) of methyl 2-(bromomethyl)-5-[1-(trifluoromethyl)cyclo- propyl]benzoate as a yellow solid. HPLC/MS m/z: 339.05 [M+H] + , Rt (E): 1.14 min. Example 20.4: Preparation as described for intermediate 19.1. Yield: 149 mg (72%) of 2-(2-aminoethyl)-6-[1-(trifluoromethyl)cyclopropyl]-2,3-dihy dro-1 H-isoindol-1-one as a yellow solid. HPLC/MS m/z: 285.15 [M+H] + , Rt (B): 0.68 min.

Example 20.5: To a stirred solution of intermediate 20.4 (134.0 mg, 0.471 mmol) and 7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-2-ium-2-olate (107.1 mg, 0.471 mmol) in dichloromethane (3.0 mL) were added PyBrOP (285.5 mg, 0.612 mmol) and DI PEA (243.5 mg, 1.884 mmol). The resulting mixture was stirred at room temperature for 2 h and concentrated under reduced pressure. The residue was purified by RP flash-chromatography to give 52 mg (22%) of 2-(2-{[7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-6-[1-(trif luoromethyl)cyclopropyl]- 2,3-dihydro-1 H-isoindol-1-one as a colorless solid. HPLC/MS m/z: 494.15 [M+H] + , Rt (F): 1.59 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.85-8.80 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.02 (t, J = 5.5 Hz, 1 H), 7.95 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.70-7.63 (m, 2H), 7.61 (dd, J = 7.6, 1.0 Hz, 1 H), 6.98-6.92 (m, 1 H), 4.58 (s, 2H), 3.89-3.83 (m, 2H), 3.83-3.75 (m, 2H), 2.67 (s, 3H), 1.41-1.33 (m, 2H), 1.16 (s, 2H).

Example 21 : Methyl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5H,6H,7H-pyrrolo[3,4-b]pyridine-3-carb oxylate

Example 21.1 : Ethyl 5-bromo-2-methylnicotinate (1.00 g, 4.097 mmol) was brominated with NMB in CCI4 as described for intermediate 17.2. Yield: 855 mg (64%) of ethyl 5-bromo-2-(bromomethyl)pyridine-3-carboxylate as a colorless oil. HPLC/MS m/z: 323.7 [M+H] + , Rt (G): 1.72 min.

Example 21.2: Intermediate 21.1 (150.0 mg, 0.464 mmol) was treated with intermediate B3 (160.0 mg, 0.594 mmol) and triethylamine (141.0 mg, 1.393 mmol) as described for compound 17.2. Yield: 187 mg (87%) of 3-bromo-6-(2-{[7-(5- methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-5H,6H,7H-p yrrolo[3,4- b]pyridin-5-one as an amorphous beige powder. HPLC/MS m/z: 465.1/467.0 [M+H] + , Rt (N): 1.28 min.

Example 21.3: Carbonylation of intermediate 21.2 to the title compound was performed as described for compound 11.2. Yield: 31 mg (41 %), beige solid. HPLC/MS m/z: 444.9 [M+H] + , Rt (G): 1.28 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 9.24 (d, J = 2.0 Hz, 1 H), 8.81-8.79 (m, 1 H), 8.34 (d, J = 2.0 Hz, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 7.98 (t, J = 5.4 Hz, 1 H), 7.91 (d, J = 5.7 Hz, 1 H), 7.83 (d, J = 8.5 Hz, 1 H), 6.93 (d, J = 5.6 Hz, 1 H), 4.76 (s, 2H), 3.91 (s, 3H), 3.91-3.87 (m, 2H), 3.87- 3.81 (m, 2H), 2.67 (s, 3H).

The following examples were prepared in an analogous manner:

Example 22: Ethyl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5H,6H,7H-pyrrolo[3,4-b]pyridine-3-carb oxylate

20 mg colorless solid. HPLC/MS m/z: 458.8 [M+H] + , Rt (G): 1.37 min. 1 H NMR (700 MHz, DMSO-d 6 ): 6 9.24 (d, J = 2.0 Hz, 1 H), 8.80-8.79 (m, 1 H), 8.34 (d, J = 1.9 Hz, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.00 (t, J = 5.7 Hz, 1 H), 7.91 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.94 (d, J = 5.7 Hz, 1 H), 4.76 (s, 2H), 4.37 (q, J = 7.1 Hz, 2H), 3.91-3.88 (m, 2H), 3.86-3.82 (m, 2H), 2.67 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

Example 23: Propan-2-yl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5H,6H,7H-pyrrolo[3,4-b]pyridine-3-carb oxylate

31 mg colorless solid. HPLC/MS m/z: 472.9 [M+H] + , Rt (M): 1.77 min. 1 H NMR (700 MHz, DMSO-d 6 ): 6 9.22 (d, J = 2.0 Hz, 1 H), 8.80-8.79 (m, 1 H), 8.32 (d, J = 2.0 Hz, 1 H), 8.14 (dd, J = 8.5, 1.6 Hz, 1 H), 8.00 (t, J = 5.6 Hz, 1 H), 7.91 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.93 (d, J = 5.9 Hz, 1 H), 5.21-5.15 (m, 1 H), 4.76 (s, 2H), 3.91-3.88 (m, 2H), 3.86-3.82 (m, 2H), 2.67 (s, 3H), 1.34 (d, J = 6.2 Hz, 6H).

Example 24: Ethyl 2-methyl-6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5H,6H,7H-pyrrolo[3,4-b]pyridine-3-carb oxylate

50 mg colorless solid. HPLC/MS m/z: 473.2 [M+H] + , Rt (N): 1.34 min. 1 H NMR (400 MHz, DMSO-d 6 ): 6 8.81-8.79 (m, 1H), 8.27 (s, 1H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.01-7.95 (m, 1H), 7.92 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1H), 6.94 (d, J = 5.8 Hz, 1H), 4.67 (s, 2H), 4.33 (q, J = 7.1 Hz, 2H), 3.90-3.85 (m, 2H), 3.85-3.80 (m, 2H), 2.81 (s, 3H), 2.67 (s, 3H), 1.33 (t, J = 7.1 Hz, 3H).

Example 25: 2-Methyl-6-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1- yl]amino}ethyl)-3-propoxy-5H,6H,7H-pyrrolo[3,4-b]pyridin-5-o ne

Example 25.1: A solution of methyl 2-hydroxy-6-methylpyridine-3-carboxylate (4.28 g, 25.604 mmol) and NBS (4.56 g, 25.620 mmol) in DMF (100.0 mL) was stirred for 4 h at room temperature under nitrogen atmosphere and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 7:1) to afford 6.0 g (95%) of methyl 5-bromo-2- hydroxy-6-methylpyridine-3-carboxylate as a colorless solid. HPLC/MS m/z: 246.05 [M+H] + , Rt (B): 0.61 min.

Example 25.2: A solution of intermediate 25.1 (5.96 g, 24.222 mmol) and 1,1,1- trifluoro-N-phenyl-N-trifluoromethanesulfonylmethanesulfonam ide (12.45 g, 34.849 mmol) and DIPEA (9.00 g, 69.684 mmol) in DMF (50.0 mL) was stirred at room temperature under nitrogen atmosphere overnight. The reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 1 :1) to give 6.69 g (73%) of methyl 5-bromo-6-methyl-2-(trifluoromethanesulfonyloxy)pyridine-3- carboxylate as a yellow oil. HPLC/MS m/z: 380.00 [M+H] + , Rt (B): 1.12 min. Example 25.3: Intermediate 25.2 (620.8 mg, 1.642 mmol) and tributyl(ethenyl)stannane (542.5 mg, 1.711 mmol) was dissolved in DMF (10.0 mL). Pd(PPh 3 ) 2 CI 2 (115.0 mg, 0.164 mmol) was added and the reaction mixture stirred at 90 °C under nitrogen atmosphere for 16 h. The mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 3:2) to afford 415.0 mg (99%) of methyl 5- bromo-2-ethenyl-6-methylpyridine-3-carboxylate as a brown oil. HPLC/MS m/z: 256.10 [M+H] + , Rt (B): 1.08 min.

Example 25.4: To a stirred solution of intermediate 25.3 (0.88 g, 3.467 mmol) and NalC>4 (1.48 g, 6.919 mmol) in THF (10.0 mL) and water (2.0 mL) was added potassium osmate(VI) dihydrate (0.13 g, 0.345 mmol) and the mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated under vacuum and the crude product (1.20 g) was used in the next step without further purification. Example 25.5: A solution of the crude aldehyde (192.0 mg, 0.744 mmol) in DCE (5.0 mL) was treated with tert-butyl N-(2-aminoethyl)carbamate (119.2 mg, 0.744 mmol) and sodium triacetoxyborohydride (315.4 mg, 1.488 mmol) and stirred at room temperature for 16 h. The reaction mixture was concentrated under reduced pressure, and the residue was purified by RP flash-chromatography to give 139 mg (51 %) of N-(2-{3-bromo-2-methyl-5-oxo-5H,6H,7H-pyrrolo[3,4-b]pyridin- 6-yl}ethyl)- carbamate as a colorless solid. HPLC/MS m/z: 372.15 [M+H] + , Rt (B): 0.81 min. Example 25.6: To a solution of intermediate 25.4 (308.0 mg, 0.832 mmol) and propan-1-ol (50.0 mg, 0.832 mmol) in toluene (5.0 mL) were added CS2CO3 (542.2 mg, 1.664 mmol) and tBuXPhos Pd G3 (66.1 mg, 0.083 mmol) and the resulting mixture was stirred at 90 °C under nitrogen atmosphere for 16 h. The reaction mixture was concentrated under reduced pressure and the residue was purified by RP flash-chromatography to yield 198 mg (68%) of tert-butyl N-(2-{2-methyl-5-oxo- 3-propoxy-5H,6H,7H-pyrrolo[3,4-b]pyridin-6-yl}ethyl)carbamat e as a yellow solid. HPLC/MS m/z: 350.30 [M+H] + , Rt (P): 1.03 min.

Example 25.7: Intermediate 25.5 (198.0 mg, 0.566 mmol) was dissolved in dioxane (2.0 mL), treated with a solution of HCI in dioxane (4 M, 2.0 mL) and the mixture was stirred at room temperature for 4 h. The mixture was basified to pH8 by addition of with aqueous NaHCO 3 solution. The mixture was concentrated under vacuum and the residue was purified by RP flash-chromatography to afford 120 mg (84%) of 6- (2-aminoethyl)-2-methyl-3-propoxy-5H,6H,7H-pyrrolo[3,4-b]pyr idin-5-one as a colorless solid. HPLC/MS m/z: 250.25 [M+H] + , Rt (P): 0.69 min.

Example 25.8: Preparation as described for compound 20.5. Yield: 28 mg (15%) of 2-methyl-6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-3- propoxy-5H,6H,7H-pyrrolo[3,4-b]pyridin-5-one as a colorless solid. HPLC/MS m/z: 459.15 [M+H] + , Rt (D): 1.19 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.83 (s, 1 H), 8.14 (d, J = 8.6 Hz, 1 H), 8.01 (t, J = 5.6 Hz, 1 H), 7.95 (d, J = 5.8 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.46 (s, 1 H), 6.95 (d, J = 5.7 Hz, 1 H), 4.50 (s, 2H), 4.03 (t, J = 6.4 Hz, 2H), 3.88-3.79 (m, 4H), 2.68 (s, 3H), 2.46 (s, 3H), 1.80-1.69 (m, 2H), 1.00 (t, J = 7.4 Hz, 3H).

Example 26: 2-(2-{[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-6- propoxy-1 H,2H,3H-pyrrolo[3,4-c]pyridin-1-one

Example 26.1 : To a stirred solution of methyl 5-bromo-2-oxo-1 ,2-dihydropyridine-4- carboxylate (3.80 g, 16.377 mmol) and 1-iodopropane (5.57 g, 32.754 mmol) in toluene (40.0 mL) was added Ag2COs (6.77 g, 24.566 mmol), and the reaction mixture was stirred at 100 °C under nitrogen atmosphere for 2 h. The mixture was concentrated under vacuum, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 1 :4) to afford 4.0 g (89%) of methyl 5-bromo-2-propoxypyridine-4-carboxylate as a colorless solid. HPLC/MS m/z: 276.1 [M+H] + , Rt (P): 1.28 min.

Example 26.2: Intermediate 26.1(1.92 g, 7.004 mmol) was reacted with tributyl(ethenyl)stannane as described for compound 27.3. Yield: 1.16 g (75%) of methyl 5-ethenyl-2-propoxypyridine-4-carboxylate as a yellow liquid. HPLC/MS m/z: 222.05 [M+H] + , Rt (B): 1.05 min. Example 26.3: To a solution of intermediate 26.2 (965.0 mg, 4.361 mmol) and NalC>4 (1.87 g, 8.743 mmol) in 1 ,4-dioxane (10.0 mL) and water (2.0 mL) was added KMnC>4 (1.03 g, 6.518 mmol), and the mixture was stirred at room temperature for 30 min. The reaction mixture was concentrated under vacuum, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 2:3) to afford 507 mg (52%) of methyl 5-formyl-2-propoxypyridine-4-carboxylate as a yellow solid. HPLC/MS m/z: 224.05 [M+H] + , Rt (D): 1.21 min.

Example 26.4: To a solution of intermediate 26.3 (507.0 mg, 2.271 mmol) and benzyl N-(2-aminoethyl)carbamate (441.1 mg, 2.271 mmol) in dichloroethane (10.0 mL) was added sodium triacetoxyborohydride (721.6 mg, 3.405 mmol) and the mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated under vacuum, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 2:3) to afford 300.0 mg (35%) of benzyl N-(2-{1-oxo-6-propoxy-1 H,2H,3H-pyrrolo[3,4-c]pyridin-2- yl}ethyl)carbamate as a yellow solid. HPLC/MS m/z: 370.25 [M+H] + , Rt (P): 1.05 min.

Example 26.5: Intermediate 26.4 (277.0 mg, 0.750 mmol) was stirred in an aqueous HCI solution (2N, 5.0 mL) at 50 °C for 2 h. The mixture was cooled to room temperature, neutralized to pH7 with aqueous NaHCOs solution, and concentrated under vacuum. The residue was purified by reverse flash chromatography to yield 158 mg (89%) of 2-(2-aminoethyl)-6-propoxy-1 H,2H,3H-pyrrolo[3,4-c]pyridin-1-one as an off-white solid. HPLC/MS m/z: 236.25 [M+H] + , Rt (P): 0.62 min.

Example 26.6: Preparation as described for compound 20.5. Yield: 23 mg (9%) of 2- (2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-6-propoxy- 1 H,2H,3H-pyrrolo[3,4-c]pyridin-1-one as an off-white solid. HPLC/MS m/z: 445.25 [M+H] + , Rt (D): 1.51 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.70 (d, J = 1.5 Hz, 1 H), 8.29 (d, J = 1.0 Hz, 1 H), 8.02 (dd, J = 8.5, 1.5 Hz, 1 H), 7.92 (d, J = 5.6 Hz, 1 H), 7.82 (d, J = 5.7 Hz, 1 H), 7.72 (d, J = 8.5 Hz, 1 H), 6.87-6.77 (m, 2H), 4.47 (s, 2H), 4.11 (t, J = 6.7 Hz, 2H), 3.76-3.64 (m, 4H), 2.56 (s, 3H), 1.69-1.51 (m, 2H), 0.83 (t, J = 7.4 Hz, 3H).

Example 27: Ethyl 1-methyl-5-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-4-oxo-1 H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridine-2-carboxylate

Example 27.1: A solution of piperidine-2, 4-dione (14.25 g, 125.978 mmol), 2,2- dimethoxyethan-1-amine (13.24 g, 125.978 mmol) and 4-methylbenzene-1 -sulfonic acid (2.17 g, 12.601 mmol) in toluene (150.0 mL) was stirred at 110 °C overnight. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was dissolved in dichloromethane (200 mL), treated with TFA (143.74 g, 1260.612 mmol) and the resulting mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 9:1) to afford 8.31 g (48%) of 1H,4H,5H,6H,7H- pyrrolo[3,2-c]pyridin-4-one as a yellow solid. HPLC/MS m/z: 137.20 [M+H] + , Rt (B): 0.22 min.

Example 27.2: A solution of 1,3-dibromo-5,5-dimethylimidazolidine-2, 4-dione (6.20 g, 21.684 mmol) and intermediate 27.1 (8.31 g, 61.035 mmol) in DMF (100.0 mL) was stirred for 20 min at -60 °C under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 9:1) to afford 11.79 g (90%) of 2-bromo-1 H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridin-4-one as a colorless solid. HPLC/MS m/z: 215.10/217.05 [M+H] + , Rt (B): 0.47 min.

Example 27.3: Intermediate 27.2 (4.70 g, 21.856 mmol) was dissolved in DMF (60.0 mL) and treated with CS2CO3 (14.44 g, 44.319 mmol) and methyl iodide (3.47 g, 24.447 mmol). The reaction mixture was stirred at room temperature overnight and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 13:1) to give 4.08 g (82%) of 2-bromo-1-methyl-1 H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridin-4-one as a colorless solid. HPLC/MS m/z: 229.10/231.15 [M+H] + , Rt (B): 0.61 min. Example 27.4: To a solution of intermediate 27.3 (1.70 g, 7.421 mmol) in ethanol (15.0 mL) in a pressure tank was added Pd(dppf)Cl2.CH2Cl2 (0.68 g, 0.833 mmol) and triethylamine (2.53 g, 25.002 mmol). The mixture was purged with nitrogen for 5 min and then pressurized to 20 atm with carbon monoxide and stirred at 100 °C overnight. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 10:1) to yield 1.56 g (95%) of ethyl 1-methyl-4-oxo- 1H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridine-2-carboxylate as a brown semi solid.

HPLC/MS m/z: 223.20 [M+H] + , Rt (B): 0.62 min.

Example 27.5: Intermediate 27.4 (785.4 mg, 3.534 mmol was dissolved in THF (10.0 mL) and sodium hydride (60%, 0.76 g, 19.000 mmol) was added at 0 °C. The mixture was warmed to room temperature and stirred for 16 h. 2-bromoacetonitrile (2.85 g, 23.760 mmol) was added and the mixture was stirred for 2 d at room temperature. The reaction was quenched with EtOH the mixture concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 10:1) to yield 199 mg (22%) of ethyl 5- (cyanomethyl)-1-methyl-4-oxo-1 H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridine-2-carboxylate as a light-brown solid. HPLC/MS m/z: 262.30 [M+H] + , Rt (B): 0.70 min.

Example 27.6: Intermediate 27.5 (184.0 mg, 0.704 mmol) was hydrogenated in EtOH (5.0 mL) in presence of Raney-Ni (10%, 200.0 mg, 0.233 mmol) at room temperature for 16 h. The reaction mixture was filtered, and the filter cake was washed with dichloromethane. The filtrate was concentrated under reduced pressure, and the residue was purified by RP flash chromatography to give 49 mg (26%) of ethyl 5-(2-aminoethyl)-1-methyl-4-oxo-1H,4H,5H,6H,7H-pyrrolo[3,2- c]pyridine-2-carboxylate as a brown solid. HPLC/MS m/z: 266.25 [M+H] + , Rt (B): 0.56 min.

Example 27.7: Preparation as described for compound 20.5. Yield: 40 mg (46%) of ethyl 1-methyl-5-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1-yl]amino}ethyl)- 4-oxo-1 H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridine-2-carboxylate as a colorless solid. HPLC/MS m/z: 475.00 [M+H] + , Rt (L): 0.63 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.86 (s, 1H), 8.22-8.08 (m, 1 H), 8.03-7.91 (m, 2H), 7.82 (d, J = 8.6 Hz, 1H), 7.00 (s, 1 H), 6.93 (d, J = 5.7 Hz, 1 H), 4.19 (q, J = 7.1 Hz, 2H), 3.73 (s, 3H), 3.70-3.66 (m, 3H), 3.63 (t, J = 6.8 Hz, 3H), 2.87 (t, J = 6.8 Hz, 2H), 2.71-2.64 (m, 3H), 2.28-2.24 (m, 1 H), 1.25 (t, J = 7.1 Hz, 3H). The following compounds were prepared analogously:

Example 28: Propan-2-yl 1-methyl-5-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-4-oxo-1H,4H,5H,6H,7H-pyrrol o[3,2-c]pyridine-2- carboxylate

9 mg colorless solid. HPLC/MS m/z: 489.05 [M+H] + , Rt (B): 0.72 min. 1 H NMR (300 MHz, DMSO-d 6 ): 68.97 (s, 1H), 8.19 (s, 1H), 7.92 (d, J=5.8Hz, 2H), 7.07 (s, 1H), 6.98 (s, 1H), 5.11-4.97 (m, 1H), 3.79-3.63 (m, 8H), 2.91 (t, J= 6.7 Hz, 2H), 2.77- 2.67 (m, 4H), 1.27 (d, J = 6.2 Hz, 6H).

Example 29: Ethyl 1-ethyl-5-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin- 1- yl]amino}ethyl)-4-oxo-1H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridine- 2-carboxylate

11 mg colorless solid. HPLC/MS m/z: 489.20 [M+H] + , Rt (O): 1.49 min. 1 H NMR (400 MHz, DMSO-d 6 ): 58.89 (s, 1H), 8.18-8.11 (m, 1H), 8.05-8.01 (m, 1H), 7.97 (d, J = 5.7 Hz, 1H), 7.85 (d, J= 8.5 Hz, 1H), 7.04 (s, 1H), 6.96 (d, J= 5.8 Hz, 1H), 4.31- 4.17 (m, 3H), 3.90 (q, J = 7.1 Hz, 1 H), 3.73- 3.57 (m, 6H), 2.96-2.81 (m, 2H), 2.70 (s, 3H), 1.28 (t, J = 7.1 Hz, 3H), 1.24-1.14 (m, 3H).

Example 30: Propan-2-yl 1-ethyl-5-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin- 1-yl]amino}ethyl)-4-oxo-1 H,4H,5H,6H,7H-pyrrolo[3,2-c]pyridine-2-carboxylate

2 mg. HPLC/MS m/z: 503.35 [M+H] + , Rt (B): 0.88 min. 1 H NMR (300 MHz, DMSO- d 6 ): 6 8.89 (s, 1 H), 8.15 (dd, J = 8.5, 1.5 Hz, 1 H), 8.06-7.93 (m, 2H), 7.85 (d, J = 8.5 Hz, 1 H), 7.01 (s, 1 H), 6.96 (d, J = 5.8 Hz, 1 H), 5.12-4.98 (m, 1 H), 4.25 (q, J = 7.0 Hz, 2H), 3.87-3.49 (m, 6H), 2.91 (t, J = 6.7 Hz, 2H), 2.70 (s, 3H), 1.27 (d, J = 6.2 Hz, 6H), 1.21 (t, J = 7.1 Hz, 3H).

Example 31 : Ethyl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-7-oxo-4H,5H,6H,7H-thieno[2,3-c]pyridine-2-ca rboxylate

Example 31.1 : 2-Bromo-4H,5H,6H,7H-thieno[2,3-c]pyridin-7-one (447.0 mg, 1.926 mmol) was dissolved in dry THF (15.0 mL) and sodium hydride (60% dispersion in mineral oil; 231.1 mg, 5.779 mmol) was added, and the suspension was cooled in an ice bath under nitrogen atmosphere and stirred for 45 min. Bromoacetonitrile (69.2 mg, 5.779 mmol) was added, the reaction mixture was allowed to warm up to room temperature and stirred for 2 h. The reaction was quenched with aqueous saturated NH4CI solution and extracted with ethyl acetate. The combined organic layers were dried with sodium sulfate, filtered, and evaporated to dryness. The crude product was purified by flash chromatography to yield 415 mg (79 %) of 2-{2- bromo-7-oxo-4H,5H,6H,7H-thieno[2,3-c]pyridin-6-yl}acetonitri le as colorless crystals. HPLC/MS m/z: 270.7/272.8 [M+H] + , Rt (G): 1.40 min.

Example 31.2: Intermediate 31.1 (229.3 mg, 0.846 mmol) was dissolved in dry methanol (3.5 mL) and dry THF (11 mL). Triethylamine (175.9 pl, 1.269 mmol), Pd(dppf)Cl2.CH2Cl2 (41.3 mg, 0.052 mmol) and 1 ,1-bis-(diphenylphosphino)- ferrocen (37.5 mg, 0.068 mmol) were added under nitrogen atmosphere. The reactor was pressurized to 4.0 bar with carbon monoxide and the reaction mixture stirred at 100 °C overnight. The reaction mixture was filtered, the filtrate evaporated to dryness and the residue purified by flash chromatography to give 194 mg (92%) of methyl 6-(cyanomethyl)-7-oxo-4H,5H,6H,7H-thieno[2,3-c]pyridine-2-ca rboxylate as a red oil. HPLC/MS m/z: 250.9 [M+H] + , Rt (G): 1.26 min.

Example 31.3: Hydrogenation of intermediate 31.2 (194.0 mg, 0.775 mmol) with Raney-Ni was performed Yield: 88 mg (39%) of methyl 6-(2-aminoethyl)-7-oxo- 4H,5H,6H,7H-thieno[2,3-c]pyridine-2-carboxylate hydrochloride as a colorless solid. HPLC/MS m/z: 254.9 [M+H] + , Rt (G): 0.97 min.

Example 31.4: Preparation as described for compound 20.5. Yield: 20 mg (22%) of methyl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-7-oxo- 4H,5H,6H,7H-thieno[2,3-c]pyridine-2-carboxylate as a colorless oil. HPLC/MS m/z: 463.9 [M+H] + , Rt (M): 1.73 min.

Example 31.5: Transesterification of intermediate 31.4 (12.0 mg, 0.026 mmol) was performed as described for example 2. Yield: 9.5 mg (77 %) of ethyl 6-(2-{[7-(5- methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-7-oxo-4H,5 H,6H,7H- thieno[2,3-c]pyridine-2-carboxylate as a colorless oil. HPLC/MS m/z: 477.8 [M+H] + , Rt (M): 1.80 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 13.47-12.58 (m, 1 H), 9.05-8.92 (m, 1 H), 8.35-8.21 (m, 1 H), 8.05-7.89 (m, 1 H), 7.92-7.86 (m, 1 H), 7.69 (s, 1 H), 7.21- 7.04 (m, 1 H), 4.30 (q, J = 7.1 Hz, 2H), 3.82-3.75 (m, 4H), 3.71 (t, J = 7.0 Hz, 2H), 2.90 (t, J = 7.0 Hz, 2H), 2.70 (s, 3H), 1.29 (t, J = 7.1 Hz, 3H).

Example 32: Ethyl 5-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-4H,5H,6H-thieno[2,3-c]pyrrole-2-carboxylate

Example 32.1: Methyl 4,5-bis(chloromethyl)thiophene-2-carboxylate (47.5 mg, 0.199 mmol) was dissolved in dry ethanol (0.5 mL). Anhydrous sodium carbonate (56.8 mg, 0.536 mmol) and N-(2-aminoethyl)carbamic acid tert-butyl ester (35.0 mg, 0.219 mmol) were added and the mixture was stirred at 100°C for 1 h. The reaction was filtered and washed with ethanol. The filtrate was evaporated to dryness. And the residue was purified by flash-chromatography to give 38 mg (59%) of methyl 5-(2- {[(tert-butoxy)carbonyl]amino}ethyl)-4H,5H,6H-thieno[2,3-c]p yrrole-2-carboxylate as a colorless solid. HPLC/MS m/z: 326.9 [M+H] + , Rt (H): 0.66 min.

Example 32.2: Intermediate 32.1 (585.0 mg, 1.792 mmol) was suspended in 1,4- dioxane (6.0 mL). A solution of HCI in dioxane (4N, 2.69 mL) was added and the suspension was stirred at room temperature overnight. The reaction mixture was evaporated to dryness to yield 535 mg (100%) of methyl 5-(2-aminoethyl)- 4H,5H,6H-thieno[2,3-c]pyrrole-2-carboxylate dihydrochloride as grey-green solid. HPLC/MS m/z: 226.9 [M+H] + , Rt (H): 0.19 min.

Example 32.3: Under nitrogen atmosphere 1-chloro-7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinoline (133.0 mg, 0.541 mmol), intermediate 32.2 (194.4 mg, 0.650 mmol), (R)-(+)-2,2’-Bis(diphenylphosphino)-1,1’-binaphthalene (20.2 mg, 0.032 mmol) and Palladium(ll)-acetate ((47% Pd; 7.3 mg, 0.032 mmol) were suspended in dry toluene (0.6 mL). Potassium tert-butylate (182.3 mg, 1.624 mmol) was added and the reaction mixture was heated to 85 °C and stirred overnight. The mixture was cooled to room temperature, filtered over Celite and washed with dichloromethane/methanol. The filtrate was diluted with demineralized water and extracted with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash-chromatography to afford 54 mg (23%) of methyl 5-(2-{[7-(5-methyl-1,2,4- oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-4H,5H,6H-thieno [2,3-c]pyrrole-2- carboxylate as a colorless solid. HPLC/MS m/z: 435.8 [M+H] + , Rt (H): 0.69 min. 1 H NMR (500 MHz, DMSO-d 6 ): 5 8.90-8.88 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 7.97 (d, J = 5.7 Hz, 1 H), 7.90-7.86 (m, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.61 (s, 1 H), 6.94 (d, J = 5.4 Hz, 1 H), 4.12-4.04 (m, 2H), 3.94-3.87 (m, 2H), 3.80 (s, 3H), 3.69 (q, J = 6.3 Hz, 2H), 3.10-3.03 (m, 2H), 2.70 (s, 3H).

Example 32.4. Transesterification of intermediate 32.3 (18.0 mg, 0.041 mmol) was performed as described for example 2. Yield: 3 mg (16%) of ethyl 5-(2-{[7-(5-methyl- 1,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-4H,5H,6H- thieno[2,3-c]pyrrole-2- carboxylate. HPLC/MS m/z: 449.8 [M+H] + , Rt (M): 1.72 min.

Example 33: Propan-2-yl 5-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-4H,5H,6H-thieno[2,3-c]pyrrole-2-carboxylate

Saponification of intermediate 32.3 (48.0 mg, 0.110) and esterification of the acid was performed as described for example 3. Yield: 9 mg (21%) of propan-2-yl 5-(2- {[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-4H,5H,6H-t hieno[2,3- c]pyrrole-2-carboxylate as a colorless solid. HPLC/MS m/z: 463.8 [M+H] + , Rt (H): 0.73 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.90-8.88 (m, 1 H), 8.15 (dd, J = 8.5, 1.6 Hz, 1 H), 7.97 (d, J = 5.7 Hz, 1 H), 7.92-7.87 (m, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.57 (s, 1 H), 6.95 (d, J = 5.8 Hz, 1 H), 5.12-5.02 (m, 1 H), 4.17-4.06 (m, 2H), 4.00-3.88 (m, 2H), 3.70 (q, J = 6.2 Hz, 2H), 3.16-3.05 (m, 2H), 2.70 (s, 3H), 1.28 (d, J = 6.2 Hz, 6H).

Example 34: Ethyl 1-methyl-5-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-1 H,4H,5H,6H-pyrrolo[2,3-c]pyrrole-2-carboxylate

Example 34.1: To a stirred solution of POCI3 (50.1 g, 327.760 mmol) in dichloroethane (300 mL) was added DMF (23.9 g, 347.760 mmol) very slowly at 0 °C. The resulting mixture was allowed to warm up to room temperature and stirred for 1 h. Ethyl 4-bromo-1H-pyrrole-2-carboxylate(15.00 g, 65.352 mmol, 1.00 equiv, 95%) was added at room temperature and the resulting mixture was heated to at 80 °C and stirred for 1 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 3:1) to afford 14.6 g (91%) of ethyl 4-bromo-5-formyl-1 H-pyrrole-2-carboxylate as a yellow solid. HPLC/MS m/z: 245.9 [M+H] + , Rt (A): 0.78 min.

Example 34.2: To a stirred solution of intermediate 34.1 (14.85 g, 56.287 mmol) in toluene (160.0 mL) was added tetramethylammonium fluoride (15.72 g, 168.796 mmol) at room temperature. The resulting mixture was heated to 100 °C and stirred for 16 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - (92:8) to afford 13 g (88%) of ethyl 4-bromo- 5-formyl-1-methylpyrrole-2-carboxylate as a colorless solid. HPLC/MS m/z: 262.0 [M+H] + , Rt (J): 0.78 min.

Example 34.3: To a stirred solution of intermediate 34.2 (9.93 g, 38.180 mmol) and (Tributylstannyl)-methanol (17.17 g, 53.473 mmol) in toluene (115.0 mL) was added Pd(PPh3)4 (0.44 g, 0.381 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 110 °C and stirred for 4 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 6:4) to yield 7.9 g (98%) of ethyl 5-formyl-4-(hydroxymethyl)-1- methylpyrrole-2-carboxylate as a yellow solid. HPLC/MS m/z: 212.1 [M+H] + , Rt (J): 0.57 min.

Example 34.4: Intermediate B3 (266.6 mg, 0.990 mmol) and intermediate 34.3 (250.8 mg, 1.187 mmol) were dissolved in dichloroethane (5.0 mL), traces of acetic acid were added, and the mixture was stirred at 50 °C for 16 h. The reaction mixture was cooled to room temperature, sodium borohydride (187.2 mg, 4.948 mmol) was added, and the mixture was at room temperature for 1 h. The reaction was quenched with aqueous saturated NH4CI solution (10 mL) and the resulting mixture was extracted with dichloromethane. The combined organic layers were dried with sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 9/1) to give 106 mg (23%) of ethyl 4-(hydroxymethyl)-1-methyl-5-{[(2-{[7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)amino]methy l}-1 H-pyrrole-2- carboxylate as a colorless. HPLC/MS m/z: 465.25 [M+H] + , Rt (J): 0.58 min.

Example 34.5: To a stirred solution of intermediate 34.4 (106.0 mg, 0.228 mmol) in dichloromethane (2.0 mL) was added thionyl chloride (56.0 mg, 0.471 mmol) at room temperature, and the resulting mixture was stirred room temperature for 1 h. The reaction mixture was concentrated under vacuum to afford 109 mg (99%) of ethyl 4-(chloromethyl)-1-methyl-5-{[(2-{[7-(5-methyl-1,2,4-oxadiaz ol-3-yl)isoquinolin- 1-yl]amino}ethyl)amino]methyl}-1 H-pyrrole-2-carboxylate as an off-white solid. HPLC/MS m/z: 479.25 [M+H] + , Rt (B): 0.74 min.

Example 34.6: A solution of intermediate 34.5 (95.0 mg, 0.196 mmol) in DMF (2.0 mL) was treated with potassium carbonate (94.0 mg, 0.682 mmol) and the mixture was heated to 80 °C and stirred for 16 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The crude product was purified by prep-HPLC to afford 3 mg (3%) of ethyl 1-methyl-5-(2-{[7-(5-methyl-1,2,4-oxadiazol- 3-yl)isoquinolin-1-yl]amino}ethyl)-1 H,4H,5H,6H-pyrrolo[2,3-c]pyrrole-2-carboxylate as a brown oil. HPLC/MS m/z: 447.10 [M+H] + , Rt (B): 0.62 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.89 (s, 1 H), 8.18-8.11 (m, 1H), 7.97 (d, J = 5.7 Hz, 1 H), 7.90-7.80 (m, 2H), 6.94 (d, J = 5.7 Hz, 1 H), 6.62 (s, 1H), 4.17 (q, J = 7.1 Hz, 2H), 3.89 (s, 2H), 3.75 (d, J = 6.8 Hz, 4H), 3.67 (q, J = 6.4 Hz, 2H), 3.03 (t, J = 6.8 Hz, 2H), 2.70 (s, 3H), 1.24 (t, J = 7.2 Hz, 4H).

Example 35: Ethyl 1-methyl-5-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1- yl]amino}ethyl)-4-oxo-1H,4H,5H,6H-pyrrolo[2,3-c]pyrrole-2-ca rboxylate

Example 35.1: To a stirred solution of intermediate 34.2 (803.0 mg, 3.087 mmol) and formyl acetate (2.91 g, 33.045 mmol) in DMF (1.0 mL) was added Pd(dppf)Cl2.CH2Cl2 (1.52 g; 1.861 mmol) at room temperature under nitrogen atmosphere followed by traces of triethylamine. The resulting mixture was stirred at 90 °C for 3 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by RP flash-chromatography to give 530 mg (76%) of 5-(ethoxycarbonyl)-2-formyl-1-methyl-1 H-pyrrole-3- carboxylic acid as a yellow solid. HPLC/MS m/z: 226.15 [M+H] + , Rt (J): 0.65 min. Example 35.2: Intermediate 35.1 (488.9 mg; 2.171 mmol) was converted as described for compound 34.4. After purification by silica gel column chromatography (eluent: dichloromethane/methanol - 9:1) 832 mg (80%) of 5-(ethoxycarbonyl)-1- methyl-2-{[(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}- ethyl)amino]methyl}-1 H-pyrrole-3-carboxylic acid was isolated as a brown solid. HPLC/MS m/z: 479.25 [M+H] + , Rt (J): 0.63 min.

Example 35.3: To a stirred solution of intermediate 35.2 (677.6 mg, 1.416 mmol) in DMF (6.0 mL) was added 1-methyl-1H-imidazole (348.7 mg, 4.247 mmol) at room temperature and the mixture was stirred for 15 min at room temperature. [Chloro(dimethylamino)methylidene]dimethylazanium; hexafluoro- 5 -phosphanuide (476.9 mg, 1.700 mmol) was added at room temperature and the mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated under vacuum and the residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol - 9:1) to afford 584 mg (90%) of ethyl 1-methyl-5-(2-{[7- (5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl) -4-oxo-1H,4H,5H,6H- pyrrolo[2,3-c]pyrrole-2-carboxylate as a colorless solid. HPLC/MS m/z: 461.25 [M+H] + , Rt (J): 0.71 min. 1 H NMR (300 MHz, DMSO-d 6 ): 6 8.73 (s, 1 H), 8.02 (d, J = 8.6 Hz, 1 H), 7.92-7.80 (m, 2H), 7.72 (d, J = 8.5 Hz, 1 H), 6.83 (d, J = 5.8 Hz, 1 H), 6.76 (d, J = 1.1 Hz, 1 H), 4.40 (s, 2H), 4.11 (q, J = 7.1 Hz, 2H), 3.71 (s, 3H), 3.61 (s, 4H), 2.57 (s, 3H), 1.21-1.10 (m, 3H).

The following example was prepared in an analogous manner:

Example 36: Propan-2-yl 1-methyl-5-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-4-oxo-1 H,4H,5H,6H-pyrrolo[2,3-c]pyrrole-2- carboxylate

Saponification of compound 35 and esterification of the corresponding acid using standard conditions provided compound 36. 25 mg colorless solid. HPLC/MS m/z: 475.30 [M+H] + , Rt (Q): 2.06 min. 1 H NMR (300 MHz, DMSO-d 6 ): 6 8.73 (s, 1 H), 8.07-7.98 (m, 1 H), 7.92-7.80 (m, 2H), 7.72 (d, J = 8.5 Hz, 1 H), 6.83 (d, J = 5.7 Hz, 1 H), 6.73 (s, 1 H), 5.01-4.84 (m, 1 H), 4.39 (s, 2H), 3.70 (s, 3H), 3.60 (d, J = 2.5 Hz, 4H), 2.57 (s, 3H), 1 .16 (d, J = 6.2 Hz, 6H).

Example 37: 2-{2-[7-(5-Methyl-[1 ,2,4]oxadiazol-3-yl)-isoquinolin-1-ylamino]-ethyl}-1- oxo-1 , 2-dihydro-isoquinoline-7-carboxylic acid ethyl ester

Example 37.1: A solution of 7-bromoisoquinolin-1-ol (1.00 g, 4.463 mmol) and tertbutyl N-(2-chloroethyl)carbamate (963 mg, 5.356 mmol) and potassium carbonate (1.90 g, 13.748 mmol) in DMF (11.0 mL) was heated to 80 °C and stirred for 12 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by PP flash-chromatography to give 1.48 g (90%) of tert-butyl N-[2-(7-bromo-1-oxoisoquinolin-2-yl)ethyl]carbamate as a colorless solid. HPLC/MS m/z: 367.15 [M+H] + , Rt (B): 0.97 min.

Example 37.2: To a solution of intermediate 37.1 (494.5 mg, 1.342 mmol) in ethanol (5.0 mL) in a pressure tank Pd(dppf)Cl2.CH2Cl2 (100.8 mg, 0.123 mmol) and triethylamine (373.0 mg, 3.686 mmol) were added. The mixture was purged with nitrogen for 5 min, pressurized to 20 atm with carbon monoxide and stirred at 100 °C overnight. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The filtrate was evaporated to dryness and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 1 :4) to afford 475 mg (98%) of ethyl 2-[2-[(tert-butoxycarbonyl)amino]ethyl]-1- oxoisoquinoline-7-carboxylate as a brown solid. HPLC/MS m/z: 361.25 [M+H] + , Rt (B): 0.94 min.

Example 37.3: Deprotection of intermediate 37.2 (475 mg, 1.318 mmol) with HCI in 1,4-dioxane was performed as described for compound 25.7. Yield: 340 mg (99%) of ethyl 2-(2-aminoethyl)-1-oxo-1,2-dihydroisoquinoline-7-carboxylate as a brown solid. HPLC/MS m/z: 261.15 [M+H] + , Rt (B): 0.60 min.

Example 37.4: A solution of intermediate 37.3 (100 mg, 0.384 mmol) and 1-chloro-7- (5-methyl-1,2,4-oxadiazol-3-yl)isoquinoline (18.9 mg, 0.077 mmol) and potassium carbonate (25.2 mg, 0.182 mmol) in dimethylsulfoxide (2.6 mL) was stirred at 120 °C for 2 d. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by prep-HPLC to afford 3.5 mg (10%) of ethyl 2-(2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino]ethyl)-1-oxoisoquinoline-7-carboxylate as a colorless solid. HPLC/MS m/z: 470.05 [M+H] + , Rt (K): 1.07 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.85-8.75 (m, 2H), 8.20-8.08 (m, 2H), 8.02-7.96 (m, 1 H), 7.92-7.79 (m, 2H), 7.70 (d, J = 8.4 Hz, 1 H), 7.35 (d, J = 7.3 Hz, 1 H), 6.93 (d, J = 5.8 Hz, 1 H), 6.55 (d, J = 7.4 Hz, 1 H), 4.42- 4.24 (m, 4H), 3.89-3.82 (m, 2H), 2.65 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

The following examples were prepared in a similar manner.

Example 38: Ethyl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5,6-dihydro-1 ,6-naphthyridine-3-carboxylate

25 mg off-white solid. HPLC/MS m/z: 471.10 [M+H] + , Rt (R): 0.64 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 9.29 (d, J = 2.2 Hz, 1 H), 8.93 (d, J = 2.3 Hz, 1 H), 8.76 (s, 1 H), 8.12 (d, J = 8.5 Hz, 1 H), 7.96 (s, 1 H), 7.82 (d, J = 7.1 Hz, 2H), 7.62 (d, J = 7.6 Hz, 1 H), 6.91 (d, J = 5.8 Hz, 1 H), 6.61 (d, J = 7.5 Hz, 1 H), 4.49-4.35 (m, 2H), 4.30 (s, 2H), 3.87 (s, 2H), 2.65 (s, 3H), 1.36 (t, J = 7.1 Hz, 3H).

Example 39: Propan-2-yl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5,6-dihydro-1 ,6-naphthyridine-3-carboxylate

9 mg colorless solid. HPLC/MS m/z: 485.20 [M+H] + , Rt (B): 0.86 min. 1 H NMR (300 MHz, DMSO-d 6 ): 6 9.28 (d, J = 2.2 Hz, 1 H), 8.91 (d, J = 2.2 Hz, 1 H), 8.75 (s, 1 H), 8.15-8.10(m,1 H), 7.97 (s, 1 H), 7.84-7.79 (m, 2H), 7.62 (d, J = 7.6 Hz, 1 H), 6.91 (d, J = 5.8 Hz, 1 H), 6.61 (d, J = 7.5 Hz, 1 H), 5.25-5.14 (m, J = Q.2 Hz, 1 H), 4.29 (t, J = 5.7 Hz, 2H), 3.87 (d, J = 5.4 Hz, 2H), 2.65 (s, 3H), 1 .35 (d, J = 6.3 Hz, 6H).

Example 40: tert-Butyl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5,6-dihydro-1 ,6-naphthyridine-3-carboxylate

25 mg yellow solid. HPLC/MS m/z: 499.15 [M+H] + , Rt (B): 0.91 min. 1 H NMR (300 MHz, DMSO-d 6 +D 2 O): 5 9.22 (d, J = 2.2 Hz, 1 H), 8.84 (d, J = 2.3 Hz, 1 H), 8.73 (d, J = 1.5 Hz, 1 H), 8.14-8.08 (m, 1 H), 7.84-7.76 (m, 2H), 7.61 (d, J = 7.6 Hz, 1 H), 6.90 (d, J = 5.8 Hz, 1 H), 6.59 (d, J = 7.6 Hz, 1 H), 4.28 (t, J = 5.6 Hz, 2H), 3.85 (t, J = 5.6 Hz, 2H), 2.63 (s, 3H), 1.56 (s, 9H). Example 41: Ethyl 6-(2-{[3-(5-methyl-1,2,4-oxadiazol-3-yl)phenyl]formamido}eth yl)-

5-oxo-5,6-dihydro-1,6-naphthyridine-3-carboxylate

10 mg colorless solid. HPLC/MS m/z: 448.15 [M+H] + , Rt (B): 0.88 min. 1 H NMR (300 MHz, DMSO-d 6 ): 69.31 (d, J = 2.2 Hz, 1H), 8.93 (d, J = 2.2 Hz, 1H), 8.80 (t, J = 5.8 Hz, 1H), 8.34 (t, J = 1.7 Hz, 1H), 8.13-8.07 (m, 1H), 7.95-7.89 (m, 1H), 7.78 (d, J = 7.6 Hz, 1H), 7.62 (t, J = 7.8 Hz, 1H), 6.71 (d, J = 7.5 Hz, 1H), 4.42-4.33 (m, 2H), 4.19 (t, J = 5.6 Hz, 2H), 3.66 (d, J = 5.8 Hz, 2H), 2.65 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

Example 42: Ethyl 6-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5,6,7,8-tetrahydro-1,6-naphthyridine-3 -carboxylate

53 mg colorless solid. HPLC/MS m/z: 473.20 [M+H] + , Rt (B): 0.77 min. 1 H NMR (400 MHz, DMSO-d 6 ): 59.07 (d, J = 2.2 Hz, 1H), 8.84 (s, 1H), 8.51 (d, J =2.2 Hz, 1H), 8.18-8.11 (m, 1H), 8.02 (t, J= 5.3 Hz, 1H), 7.95 (d, J= 5.7 Hz, 1H), 7.84 (d, J= 8.5 Hz, 1 H), 6.95 (d, J = 5.8 Hz, 1 H), 4.40-4.30 (m, 2H), 3.87-3.76 (m, 4H), 3.73 (t, J = 6.7 Hz, 2H), 3.17 (t, J = 6.7 Hz, 2H), 2.68 (s, 3H), 1.33 (t, J = 7.1 Hz, 3H).

Example 43: Propan-2-yl 6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5,6,7,8-tetrahydro-1 ,6-naphthyridine-3-carboxylate

38 mg colorless solid. HPLC/MS m/z: 487.15 [M+H] + , Rt (B): 0.82 min. 1 H NMR (300 MHz, DMSO-d 6 ): 6 8.92 (d, J = 2.2 Hz, 1 H), 8.71 (d, J = 1.5 Hz, 1 H), 8.36 (d, J = 2.2 Hz, 1 H), 8.04-7.99 (m, 1 H), 7.91 (d, J = 5.6 Hz, 1 H), 7.82 (d, J = 5.7 Hz, 1 H), 7.70 (d, J = 8.5 Hz, 1 H), 6.81 (d, J = 5.7 Hz, 1 H), 5.13-4.95 (m, 1 H), 3.60 (t, J = 6.7 Hz, 6H), 3.04 (t, J = 6.6 Hz, 2H), 2.55 (s, 3H), 1 .20 (d, J = 6.2 Hz, 6H).

Example 44: Ethyl 2-methyl-6-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-5-oxo-5,6,7,8-tetrahydro-1 ,6-naphthyridine-3-carboxylate

Example 44.1 : Ethyl 5-cyano-2-hydroxy-6-methylpyridine-3-carboxylate (10.0 g, 48.497 mmol) was stirred in POCI3 (22.3 g, 145.436 mmol) at 90 °C for 16 h. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and was quenched by the addition of ice water (20 mL). The resulting mixture was extracted with ethyl acetate, the combined organic layers were washed with brine, dried with sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 2:3) to afford 7.71 g (71%) of ethyl 2-chloro-5-cyano- 6-methylpyridine-3-carboxylate as a yellow solid. HPLC/MS m/z: 225.0 [M+H] + , Rt (B): 0.87 min.

Example 44.2: To a solution of intermediate 44.1 (7.71 g, 34.316 mmol) and tributyl(ethenyl)stannane (13.06 g, 41.186 mmol) in DMF (80.0 mL) was added Pd(PPh 3 ) 2 CI 2 (2.41 g, 3.434 mmol). The reaction mixture was heated to 100 °C under nitrogen atmosphere and stirred for 16 h. The reaction mixture was cooled to room temperature, diluted with water (80 mL), and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 1 :2) to yield 6.11 g (82%) of ethyl 5-cyano-2-ethenyl-6-methylpyridine-3-carboxylate as a colorless solid. HPLC/MS m/z: 217.25 [M+H] + , Rt (B): 0.98 min.

Example 44.3: A solution of intermediate 44.2 (5.92 g, 27.390 mmol) and benzyl N- (2-aminoethyl)carbamate (5.32 g; 27.390 mmol) in methanol (15.0 mL) and acetonitrile (15.0 mL) was treated with diethyl amine (7.08 g, 54.799 mmol) and irradiated in a microwave at 150 °C for 1 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether - 1 :1) and by RP flash-chromatography to yield 2 g (20%) of benzyl N-[2-(3-cyano-2-methyl-5- oxo-5,6,7,8-tetrahydro-1 ,6-naphthyridin-6-yl)ethyl]carbamate as a brown solid. HPLC/MS m/z: 365.20 [M+H] + , Rt (B): 0.85 min.

Example 44.4: Intermediate 44.3 (993.0 g, 2.724 mmol) was stirred in HCI (5.0 mL) at 100 °C for 16 h. The reaction mixture was cooled to room temperature, neutralized to pH7 with aqueous NaHCO 3 solution, and concentrated under vacuum. The residue was purified by RP flash-chromatography to give 663 mg (98%) of 6-(2- aminoethyl)-2-methyl-5-oxo-5,6,7,8-tetrahydro-1 ,6-naphthyridine-3-carboxylic acid as a yellow solid. HPLC/MS m/z: 250.20 [M+H] + , Rt (B): 0.13 min.

Example 44.5: Intermediate 44.4 (351.5 mg, 1.410 mmol) was dissolved in ethanol (5.0 mL), treated with H2SO4 (0.02 mL, 0.28 mmol) and stirred at 80 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by RP flash-chromatography to yield 206 mg (53%) of ethyl 6-(2-aminoethyl)-2-methyl-5-oxo-5,6,7,8-tetrahydro-1,6- naphthyridine-3-carboxylate as a yellow solid. HPLC/MS m/z: 278.10 [M+H] + , Rt (B): 0.53 min.

Example 44.6: Preparation as described for compound 20.5. Yield: 28 mg (9%) of ethyl 2-methyl-6-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1-yl]amino}ethyl)- 5-oxo-5,6,7,8-tetrahydro-1,6-naphthyridine-3-carboxylate as an off-white solid.

HPLC/MS m/z: 487.20 [M+H] + , Rt (B): 0.83 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.85 (s, 1H), 8.45 (s, 1H), 8.15 (dd, J= 8.5, 1.5 Hz, 1H), 8.04 (d, J= 5.2 Hz, 1H), 7.96 (d, J= 5.7 Hz, 1H), 7.85 (d, J= 8.6 Hz, 1H), 6.96 (d, J= 5.8 Hz, 1H), 4.38-4.25 (m, 2H), 3.84-3.75 (m, 4H), 3.71 (t, J= 6.7 Hz, 2H), 3.10 (t, J= 6.6 Hz, 2H), 2.72 (d, J= 17.5 Hz, 6H), 1.32 (t, J= 7.1 Hz, 3H).

The following example was obtained in an analogous manner.

Example 45: Propan-2-yl 2-methyl-6-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-5-oxo-5,6,7,8-tetrahydro-1, 6-naphthyridine-3- carboxylate

68 mg colorless solid. HPLC/MS m/z: 501.30 [M+H] + , Rt (S): 1.58 min. 1 H NMR (300 MHz, DMSO-d 6 ): 68.85 (s, 1H), 8.42 (s, 1H), 8.15 (dd, J= 8.5, 1.5 Hz, 1H), 7.92- 8.05 (m, 2H), 7.85 (d, J= 8.5 Hz, 1H), 6.96 (d, J= 5.8 Hz, 1H), 5.05-5.22 (m, 1H), 3.77-3.84 (m, 3H), 3.70 (t, J= 6.6 Hz, 3H), 3.09 (t, J= 6.6 Hz, 2H), 2.72 (d, J= 15.9 Hz, 6H), 1.32 (d, J= 6.3 Hz, 6H).

Example 46: Methyl 2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2H,3H-imidazo[1,5-a]pyrazine-6-carboxy late

Example 46.1: To a suspension of methyl 5-formylpyrazine-2-carboxylate (27.7 mg, 0.173 mmol) and intermediate B3 (56.0 mg, 0.208 mmol) in dry dichloromethane (2.2 mL) was added sodium triacetoxyborohydride (55.1 mg, 0.260 mmol) and the mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure to give a pale-red residue, which was purified by RP flash-chromatography to yield 41 mg (56%) of methyl 5-{[(2-{[7-(5-methyl- 1,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)amino]meth yl}pyrazine-2- carboxylate as a yellow solid. HPLC/MS m/z: 419.8 [M+H] + , Rt (H): 0.62 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 9.06 (d, J = 1.4 Hz, 1H), 8.88-8.87 (m, 1H), 8.85 (d, J = 1.4 Hz, 1H), 8.14 (dd, J = 8.4, 1.5 Hz, 1 H), 7.93 (d, J = 5.7 Hz, 1H), 7.83 (d, J = 8.5 Hz, 1 H), 7.81 (t, J = 5.4 Hz, 1 H), 6.92 (d, J = 5.6 Hz, 1H), 4.02 (s, 2H), 3.90 (s, 3H), 3.62 (q, J = 6.2 Hz, 2H), 2.88 (t, J = 6.4 Hz, 2H), 2.70 (s, 3H), 2.75-2.62 (m, 1 H).

Example 46.2: Intermediate 46.1 (41.0 mg, 0.098 mmol) was suspended in dichloromethane (0.25 mL) and N-ethyldiisopropylamine (42.9 pL, 0.253 mmol) and the mixture was cooled to 0 °C in an ice bath. Bis(trichloromethyl) carbonate (29.1 mg, 0.098 mmol), dissolved in dichloromethane (0.49 mL), was added slowly. A clear solution was formed, which was allowed to warm to room temperature and was stirred overnight. The reaction was evaporated to dryness and the residue purified by flash chromatography to afford 23 mg (53%) of methyl 2-(2-{[7-(5-methyl-1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-3-oxo-2H,3H-imi dazo[1,5-a]pyrazine-6- carboxylate as a yellow solid. HPLC/MS m/z: 445.8 [M+H] + , Rt (M): 1.61 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 9.14-9.13 (m, 1 H), 9.09-9.09 (m, 1 H), 8.54 (dd, J = 8.4, 1.5 Hz, 1 H), 8.30-8.29 (m, 1H), 8.26-8.25 (m, 1H), 8.12 (d, J = 8.4 Hz, 1 H), 7.81 (d, J = 6.9 Hz, 1H), 7.37 (d, J = 6.9 Hz, 1 H), 4.43-4.40 (m, 2H), 4.12-4.09 (m, 2H), 3.90 (s, 3H), 2.69 (s, 3H). Example 47: Ethyl 2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2H,3H-imidazo[1,5-a]pyrazine-6-carboxy late

Example 47.1: Example 46 (265.5 mg, 0.596 mmol) was dissolved in dry THF (14.8 mL) and water (7.4 mL), treated with lithium hydroxide (36.0 mg, 1.490 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction was diluted with water (40 mL), acidified with HCI solution to pH3-4. A precipitate was formed, which was filtered by suction, rinsed with water, and dried under high vacuum to give 145 mg (56%) of 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin- 1-yl]amino}ethyl)-3-oxo-2H,3H-imidazo[1,5-a]pyrazine-6-carbo xylic acid as a yellow solid. HPLC/MS m/z: 431.8 [M+H] + , Rt (M): 1.57 min.

Example 47.2: Intermediate 47.1 (48.5 mg, 0.112 mmol), 4-(dimethylamino)-pyridine (5.5 mg, 0.045 mmol) and DCC (37.1 mg, 0.180 mmol) were suspended in dry THF (0.3 mL) and DMF (0.9 mL). Ethanol (65.6 pl, 1.124 mmol) was added and the reaction was stirred at room temperature overnight. The reaction was concentrated under reduced pressure and the residue purified by RP flash- chromatography to afford 22 mg (43%) of ethyl 2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2H,3H-imidazo[1,5-a]pyrazine-6-carboxy late as a yellow solid. HPLC/MS m/z: 459.8 [M+H] + , Rt (G): 1.30 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.80-8.79 (m, 1H), 8.46 (d, J = 1.7 Hz, 1 H), 8.15 (dd, J = 8.4, 1.5 Hz, 1 H), 7.99-7.98 (m, 1 H), 7.96 (t, J = 5.7 Hz, 1 H), 7.93 (d, J = 5.7 Hz, 1H), 7.85 (d, J = 8.4 Hz, 1 H), 7.33 (d, J = 0.9 Hz, 1H), 6.97 (d, J = 5.8 Hz, 1 H), 4.25 (q, J = 7.1 Hz, 2H), 4.15-4.12 (m, 2H), 3.84 (q, J = 5.6 Hz, 2H), 2.68 (s, 3H), 1.28 (t, J = 7.1 Hz, 3H).

Example 48: Propan-2-yl 2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3-oxo-2H,3H-imidazo[1,5-a]pyrazine-6-carboxy late

Preparation as described for example 47. Yield: 17 mg (32%) yellow solid.

HPLC/MS m/z: 473.9 [M+H] + , Rt (H): 0.71 min. 1 H NMR (700 MHz, DMSO-d 6 ): 5 8.80-8.79 (m, 1 H), 8.46 (d, J = 1.6 Hz, 1 H), 8.15 (dd, J = 8.6, 1.5 Hz, 1 H), 7.97-7.94 (m, 2H), 7.93 (d, J = 5.6 Hz, 1 H), 7.86 (d, J = 8.4 Hz, 1 H), 7.33 (s, 1 H), 6.97 (d, J = 5.7 Hz, 1 H), 5.11-5.05 (m, 1 H), 4.14 (t, J = 5.7 Hz, 2H), 3.84 (q, J = 5.7 Hz, 2H), 2.69 (s, 3H), 1.28 (d, J = 6.2 Hz, 6H).

Example 49: Ethyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-2,3-dihydro-1 H-isoindole-5-carboxylate

Example 49.1 : To a stirred solution of methyl 2,3-dihydro-1 H-isoindole-5-carboxylate hydrochloride (2.85 g, 13.339 mmol) and tert-butyl N-(2-chloroethyl)carbamate (4.09 g, 22.767 mmol) in DMF (40.0 mL) was added triethylamine (3.99 g, 39.431 mmol) and the resulting mixture was stirred at 80 °C overnight. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate - 1 :1.5) to afford 1.95 g (46%) of methyl 2-(2-{[(tert-butoxy)carbonyl]amino}ethyl)-2,3- dihydro-1 H-isoindole-5-carboxylate as a brown oil. HPLC/MS m/z: 321.20 [M+H] + , Rt (J): 0.54 min.

Example 49.2: A solution of intermediate 49.1 (1.95 g, 6.086 mmol) in HCI/MeOH (20.0 mL) was stirred at room temperature for 2 h. The mixture was neutralized to pH7 with aqueous saturated NaHCOs solution (20 mL) and extracted with dichloromethane. The combined organic layers were dried with sodium sulfate, filtered, and concentrated under reduced pressure to give 1.28 g (86%) of methyl 2- (2-aminoethyl)-2,3-dihydro-1 H-isoindole-5-carboxylate as a yellow solid. HPLC/MS m/z: 221.10 [M+H] + , Rt (J): 0.29 min.

Example 49.3: Preparation as described for compound 20.5. Yield: 38 mg (31%) of methyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-2,3- dihydro-1 H-isoindole-5-carboxylate as a brown oil. HPLC/MS m/z: 430.20 [M+H] + , Rt (J): 0.53 min.

Example 49.4: A solution of intermediate 49.3 (96.9 mg, 0.225 mmol) and lithium hydroxide (22.8 mg, 0.952 mmol) in water (4.0 mL) and 1 ,4-dioxane (2.0 ml) was stirred at 60 °C overnight. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by RP flashchromatography to afford 75 mg (80%) of 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-2,3-dihydro-1 H-isoindole-5-carboxylic acid.

HPLC/MS m/z: 416.25 [M+H] + , Rt (J): 0.52 min.

Example 49.5: Esterification was performed as described for intermediate 44.5. Yield: 9 mg (6%) of ethyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-2,3-dihydro-1 H-isoindole-5-carboxylate as a yellow oil. HPLC/MS m/z: 444.20 [M+H] + , Rt (J): 0.58 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.88 (d, J = 1.5 Hz, 1 H), 8.17-8.08 (m, 1 H), 7.96 (d, J = 5.8 Hz, 1 H), 7.89 (t, J = 5.4 Hz, 1 H), 7.86-7.76 (m, 3H), 7.37 (d, J = 8.3 Hz, 1 H), 6.93 (d, J = 5.7 Hz, 1 H), 4.34-4.20 (m, 2H), 4.00 (s, 4H), 3.76-3.64 (m, 2H), 3.00 (t, J = 6.7 Hz, 2H), 2.68 (s, 3H), 1.30 (t, J = 7.1 Hz, 3H).

The following example was obtained in an analogous manner.

Example 50: Ethyl 2-(2-{[7-(1-methyl-1 H-pyrazol-4-yl)isoquinolin-1-yl]amino}ethyl)-

2,3-dihydro-1 H-isoindole-5-carboxylate

14 mg off-white solid. HPLC/MS m/z: 442.25 [M+H] + , Rt (J): 0.57 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 8.39 (d, J = 1.5 Hz, 1 H), 8.19 (s, 1 H), 8.00 (d, J = 0.8 Hz, 1 H), 7.90-7.77 (m, 4H), 7.66 (d, J = 8.5 Hz, 1 H), 7.39 (t, J = 8.0 Hz, 2H), 6.83 (d, J = 5.8 Hz, 1H), 4.28 (q, J = 7.1 Hz, 2H), 4.01 (s, 4H), 3.88 (s, 3H), 3.78-3.61 (m, 2H), 3.00 (t, J = 6.8 Hz, 2H), 1.30 (t, J= 7.1 Hz, 3H).

Example 51: Ethyl 1-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1- yl]amino}ethyl)-3-oxo-2,3-dihydro-1 H-indazole-5-carboxylate

Example 51.1: 7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-2-ium-2olate (708.0 mg, 3.116 mmol) and aminoacetaldehyde diethyl acetal (519.0 mg, 3.897 mmol) were dissolved in dichloromethane (16.0 mL) under an argon atmosphere. N- Ethyldiisopropylamine (1.91 g, 14.778 mmol) and PyBroP (1.89 g, 4.054 mmol) were added, and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with dichloromethane and washed with water. The organic layer was dried with sodium sulfate, filtered, and evaporated to dryness. The crude product was purified by flash-chromatography to yield 681 mg (64%) of N- (2,2-diethoxyethyl)-7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquin olin-1-amine as a beige solid. HPLC/MS m/z: 342.9 [M+H] + , Rt (M): 1.67 min.

Example 51.2: Intermediate 51.1 (681.0 mg, 1.989 mmol) was dissolved in THF (5.0 mL), hydrochloric acid (2N, 10.0 mL) was added, and the reaction mixture was heated to 60 °C and stirred for 3 h. The reaction mixture was cooled down to room temperature and basified with 6N NaOH. A precipitate was formed, which was filtered off, washed with water, and dried to give 468 mg (88%) of 2-{[7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}acetaldehyde as a beige solid. HPLC/MS m/z: 268.9 [M+H] + , Rt (M): 1.40 min.

Example 51.3: Intermediate 51.2 (448.0 mg, 1.670 mmol) was dissolved in methanol (11.0 mL) and N-methyl(tert-butoxy)carbohydrazide (293-0 mg, 2.004 mmol) and glacial acetic acid (430 pl, 7.515 mmol) were added, and the reaction mixture was stirred at room temperature for 1 h. Sodium cyanoborohydride (157.4 mg, 2.505 mmol) was added slowly and the reaction was stirred for 24 h. The reaction mixture was evaporated, the residue was dissolved in dichloromethane and treated with aqueous saturated NaHCCh solution. The mixture was rigorously stirred until the gas formation ceased. The phases were separated, and the aqueous phase was extracted with dichloromethane, the combined organic extracts were washed with brine, dried with sodium sulfate, filtered, and concentrated under reduced pressure to afford 665 mg (100%) of N-methyl-N'-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)(tert-butoxy)carbohydrazide as a yellow oil. HPLC/MS m/z: 398.9 [M+H] + , Rt (M): 1.74 min.

Example 51.4: 2-Bromo-5-(methoxycarbonyl)benzoic acid (528.2 mg, 2.039 mmol) was dissolved in thionyl chloride (1.23 mL, 16.991 mmol) and stirred at 75 °C for 1 h. The excess of thionyl chloride was removed in vacuo, the residue was dissolved in little dichloromethane and added slowly to a solution of intermediate 51.3 (677.0 mg, 1.699 mmol) and triethylamine (7.07 ml, 50.974 mmol) in dichloromethane (11.0 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure and the residue was purified by RP flash-chromatography to yield 826 mg (76%) of methyl 4-bromo-3-{N'-[(tert- butoxy)carbonyl]-N'-methyl-N-(2-{[7-(5-methyl-1,2,4-oxadiazo l-3-yl)isoquinolin-1- yl]amino}ethyl)hydrazinecarbonyl}benzoate as a yellow oil. HPLC/MS m/z: 639.1/641.1 [M+H] + , Rt (N): 1.61 min.

Example 51.5: Intermediate 51.4 (826.0 mg, 1.292 mmol) was dissolved in a solution of HCI in 1 ,4-dioxane (4.0 M, 11.2 mL) and stirred at room temperature for 2.5 h while a precipitate was formed. The reaction mixture was concentrated in vacuo, the residue was dissolved in dichloromethane and washed with aqueous saturated NaHCCh solution. The organic phase was dried with sodium sulfate, filtered, and evaporated. The crude product (650 mg) was used in the next step without further purification. HPLC/MS m/z: 539.1/541.1 [M+H] + , Rt (N): 1.35 min. Example 51.6: Intermediate 51.5 (650.0 mg, 1.205 mmol) was dissolved in DMF (3.50 mL), cesium carbonate (784.8 mg, 2.409 mmol), copper iodide (22.9 mg, 0.120 mmol) and 2-acetylcyclohexanone (33.8 mg, 0.241 mmol) were added under an argon atmosphere, and the reaction mixture was heated to 90 °C and stirred for 5 h. After cooling down to room temperature, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried with sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by RP flash-chromatography to give 442 mg (80%) of methyl 1-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1- yl]amino}ethyl)-3-oxo-2,3-dihydro-1H-indazole-5-carboxylate as a colorless solid. HPLC/MS m/z: 459.1 [M+H] + , Rt (N): 1.25 min.

Example 51.7: Saponification of intermediate 51.6 (274.0 mg, 0.598) and esterification of the corresponding acid (50 mg, 0.090 mmol) was performed as described for example 3. Yield: 34 mg (80%) of ethyl 1-methyl-2-(2-{[7-(5-methyl- 1,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-3-oxo-2,3 -dihydro-1H-indazole-5- carboxylate as a colorless solid. HPLC/MS m/z: 472.8 [M+H] + , Rt (M): 1.72 min. 1 H NMR (700 MHz, DMSO-d 6 ): 58.78 (s, 1H), 8.18 (d, J= 1.7 Hz, 1H), 8.15 (dd, J = 8.3, 1.6 Hz, 1H), 8.09 (dd, J= 8.6, 1.7 Hz, 1H), 8.04 (t, J= 5.7 Hz, 1H), 7.96 (d, J = 5.7 Hz, 1H), 7.86 (d, J= 8.5 Hz, 1H), 7.57 (d, J= 8.7 Hz, 1H), 6.98 (d, J= 5.7 Hz, 1H), 4.30 (q, J= 7.1 Hz, 2H), 4.20 (t, J= 6.5 Hz, 2H), 3.73 (q, J= 6.3 Hz, 2H), 3.48 (s, 3H), 2.68 (s, 3H), 1.33 (t, J= 7.1 Hz, 3H).

The following example was obtained in an analogous manner.

Example 52: Propan-2-yl 1-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-3-oxo-2,3-dihydro-1H-indazo le-5-carboxylate

56 mg colorless solid. HPLC/MS m/z: 486.9 [M+H] + , Rt (M): 1.79 min. 1 H NMR (700 MHz, DMSO-d 6 ): 58.78 (s, 1H), 8.18-8.13 (m, 2H), 8.10-8.06 (m, 1H), 8.03 (t, J = 5.7 Hz, 1H), 7.96 (d, J= 5.7 Hz, 1H), 7.86 (d, J= 8.4 Hz, 1H), 7.57 (d, J= 8.6 Hz, 1H), 6.98 (d, J= 5.7 Hz, 1H), 5.12 (p, J= 6.2 Hz, 1H), 4.20 (t, J= 6.6 Hz, 2H), 3.73 (q, J = 6.3 Hz, 2H), 3.48 (s, 3H), 2.69 (s, 3H), 1.32 (d, J = 6.2 Hz, 6H).

Example 53: 1-[6-(2-{[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)- 4H,5H,6H,7H-thieno[2,3-c]pyridin-2-yl]butan-1-one

Example 53.1: Ethyl 4H,5H,6H,7H-thieno[2,3-c]pyridine-2-carboxylate hydrochloride (33.2 mg, 0.157 mmol) and 2-(Boc-amino)ethyl bromide (70.6 mg, 0.315 mmol) were placed in a vial and suspended in dry 1 ,4-dioxane (0.50 mL). Triethylamine (76.4 pl, 0.551 mmol) was added, the mixture was heated to 80 °C and stirred for 18 h. The reaction mixture was cooled to room temperature, diluted with 5 mL dichloromethane and 5 ml water. The aqueous phase was washed twice with dichloromethane, the combined organic layer was dried with sodium sulfate, filtered, and evaporated to dryness. The residue wss purified by flash chromatography to give 45 mg (81%) of ethyl 6-(2-{[(tert-butoxy)carbonyl]amino}ethyl)-4H,5H,6H,7H- thieno[2,3-c]pyridine-2-carboxylate as a yellow oil. HPLC/MS m/z: 354.9 [M+H] + , Rt (H): 0.70 min.

Example 53.2: Intermediate 53.1 (157.4 mg, 0.444 mmol) was dissolved in dry THF (7 mL) and water (4 mL). While stirring lithium hydroxide (26.6 mg, 1.110 mmol) was added and the reaction mixture was stirred at room temperature overnight. A light yellow/orange solution was formed, which was stirred at room temperature for further 48h. The reaction mixture was diluted with water (40 mL) and acidified to pH3-4 with 1.0 N HCI solution. The mixture was extracted twice with ethyl acetate, the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The aqueous phase was neutralized with saturated aqueous NaHCCh solution and evaporated to dryness. The colorless solid was combined with the solid isolated after extraction, triturated with a mixture of dicloromethane/methanol/dimethylformamide, filtered, washed and the filtrate was evaporated to dryness to yield 145 mg (100%) of 6-(2-{[(tert-butoxy)carbonyl]- amino}ethyl)-4H,5H,6H,7H-thieno[2,3-c]pyridine-2-carboxylic acid as a beige solid. HPLC/MS m/z: 326.9 [M+H] + , Rt (H): 0.63 min.

Example 53.3: Intermediate 53.2 (145.0 mg, 0.444 mmol) and N,O- dimethylhydroxylamine hydrochloride (52.0 mg, 0.533 mmol) were suspended in N,N-Dimethylformamide (1.6 mL). [Dimethylamino-([1,2,3]triazolo[4,5-b]pyridin-3- yloxy)-methylene]-dimethyl-ammonium; hexafluoro phosphate (HATU) (168.8 mg, 0.444 mmol) and N-ethyldiisopropylamine (0.38 mL, 2.221 mmol) were added and the mixture was stirred at room temperature for 2 h. Further N,O- dimethylhydroxylamine (21.7 mg, 0.222 mmol) and [Dimethylamino- ([1 ,2,3]triazolo[4,5-b]pyridin-3-yloxy)-methylene]-dimethyl-amm onium; hexafluoro phosphate (HATLI) (84.4 mg, 0.222 mmol) were added and the reaction was stirred at room temperature overnight. The mixture was diluted with ethyl acetate and extracted two times with water and saturated aqueous NaHCOs-solution (1 :1). The combined aqueous phase was extracted twice using ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash chromatography to afford 121 mg (73%) of tert-butyl N-(2-{2-[methoxy(methyl)carbamoyl]-4H,5H,6H,7H- thieno[2,3-c]pyridin-6-yl}ethyl)carbamate as a beige oil. HPLC/MS m/z: 370.2 [M+H] + , Rt (N): 1.15 min.

Example 53.4: tert-butyl N-(2-{2-[methoxy(methyl)carbamoyl]-4H,5H,6H,7H- thieno[2,3-c]pyridin-6-yl}ethyl)carbamate (119.7 mg, 0.324 mmol) was dissolved in dry THF (0.50 mL). The clear solution was cooled in an ice bath to 0 °C. Bromo(propyl)magnesium (3M solution, 119 pl, 0.357 mmol) was added slowly and the yellow solution was allowed to warm to room temperature overnight. Further bromo(propyl)magnesium (119 pl, 0.357 mmol) was added and the reaction mixture was stirred at room temperature. After 30 min a yellow thick gum precipitated on the bottom of the flask. Dry THF (0.50 mL) was added, and the mixture was stirred at room temperature overnight. The reaction was quenched with saturated aqueous NH4CI-solution and extracted twice using ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash chromatography to yield 61 mg (53%) of tert-butyl N-(2-{2-butanoyl-4H,5H,6H,7H-thieno[2,3-c]pyridin-6-yl}ethyl )carbamate as a yellow oil (53%). HPLC/MS m/z: 352.9 [M+H] + , Rt (H): 0.71 min.

Example 53.5: Deprotection of intermediate 53.4 (61 mg, 0.172 mmol) with HCI in 1 ,4-dioxane was performed as described for compound 25.7 afforded 43.4 mg (100%) of 1-[6-(2-aminoethyl)-4H,5H,6H,7H-thieno[2,3-c]pyridin-2-yl]bu tan-1-one as a brown oil, which was used in the next steps without further purification.

Example 54.6: Preparation as described for example 32.3. Yield: 21 mg orange solid. HPLC/MS m/z: 461.9 [M+H] + , Rt (M): 1.76 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.87-8.85 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 7.96 (d, J = 5.8 Hz, 1 H), 7.83 (d, J = 8.4 Hz, 1 H), 7.84-7.80 (m, 1 H), 7.67 (s, 1 H), 6.94-6.92 (m, 1 H), 3.81-3.78 (m, 2H), 3.71 (q, J = 6.4 Hz, 2H), 2.87-2.79 (m, 6H), 2.70 (s, 3H), 2.72-2.66 (m, 2H), 1.67-1.56 (m, 2H), 0.91 (t, J = 7.4 Hz, 3H).

Example 54: Ethyl 6-methyl-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-1-oxo-3,4-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate

Example 54.1: Ethyl pyruvate (0.67 mL, 6.0282 mmol) and Ethylenediamine (0.40 mL, 6.0282 mmol) were added to MeCN (10.05 mL, 0.3700 M) and stirred for 30 min at 20 °C vigorously. Ethyl 3-bromo-2-oxobutanoate (1260.14 mg, 6.0282 mmol) in MeCN (6.03 mL, 0.3700 M) and iron(lll) chloride (195.56 mg, 1.2056 mmol) were added to the mixture and heating at reflux for 5 h. After completion of the reaction, as indicated by LCMS, the mixture was cooled to room temperature. The solution was passed through celite to remove the residual FeO, the organics were then evaporated, and the crude was subjected to normal phase column chromatography (0-100% EtOAc:CycHex -> 10% MeOH) to afford crude ethyl 6-methyl-1-oxo-3,4- dihydro-2H-pyrrolo[1,2-a]pyrazine-7-carboxylate (567.5 mg, 42%, 2.5535 mmol) as a dark brown solid. The product was used in the next step without further purification. HPLC/MS mlz 223.1078 [M+H] + , Rt (Al): 1.05 min.

Example 54.2: To a solution of ethyl 6-methyl-1-oxo-3,4-dihydro-2H-pyrrolo[1,2- a]pyrazine-7-carboxylate (550.00 mg, 2.4748 mmol) in THF (24.75 mL, 0.1000 M) at 0°C was added Sodium hydride (60% dispersion in mineral oil) (217.78 mg, 5.4446 mmol) and the mixture was stirred and allowed to warm to room temperature over 1 h. Bromoacetonitrile (0.57 mL, 8.1668 mmol) was then added and the reaction was allowed to stir at room temperature overnight. The reaction was quenched with NH4CI, then DCM was added, and the reaction was passed through a phase separator and washed with DCM several times. The combined organic layer was evaporated in vacuo to give a crude product that was subjected to silica gel normal phase column chromatography (40-100% EtOAc:CycHex) to give ethyl 2- (cyanomethyl)-6-methyl-1-oxo-3,4-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (142 mg, 22%, 0.5435 mmol) as a light yellow/brown solid. HPLC/MS m/z 262.1195 [M+H] + , m/z 284.0994 [M+Na] + , Rt (AC): 1.06 min. 1 H NMR (600 MHz, Chloroform- d): 5 7.40 (s, 1 H), 4.53 (s, 2H), 4.28 (q, J = 7.1 Hz, 2H), 4.17-4.12 (m, 2H), 3.86- 3.79 (m, 2H), 2.56 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

Example 54.3: To a solution of ethyl 2-(cyanomethyl)-6-methyl-1-oxo-3,4- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (140.00 mg, 0.5358 mmol) in EtOH (8.00 mL, 0.0500 M), AcOH (0.15 mL, 0.0500 M) and water (2.50 mL, 0.0500 M) was added . The reaction mixture was placed under a H2 atmosphere, and the solution was stirred at room temperature for 16 h. Pd/C was filtered through a pad of celite and washed with MeOH (5 mL). The crude sample was evaporated and subjected to reverse phase column chromatography (10-80% MeOH:water). The pure fractions were run through an SCX-II column and released using 2N NH3 in methanol to afford ethyl 2-(2-aminoethyl)-6-methyl-1-oxo-3,4-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (80 mg, 56%, 0.3015 mmol) as a pale-yellow semi-solid. HPLC/MS m/z 249.1238 [M+H] + , Rt (AC): 0.66 min. 1 H NMR (600 MHz, DMSO-d 6 ): 56.90 (s, 1 H), 4.18 (q, J = 7.1 Hz, 2H), 4.12-4.05 (m, 2H), 3.73-3.65 (m, 2H), 3.46 (t, J = 6.5 Hz, 2H), 2.78 (t, J = 6.5 Hz, 2H), 2.47 (s, 3H), 1.26 (t, J = 7.1 Hz, 3H). Example 54.4: 5-Methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole [Intermediate A5] (43.16 mg, 0.1900 mmol), DIPEA (0.10 mL, 0.5936 mmol) and Bromotri(pyrrolidino)phosphonium hexafluorophosphate (88.56 mg, 0.1900 mmol) in anhydrous DCM (0.75 mL, 0.1300 M) were stirred under a nitrogen atmosphere for 30 min at 40 °C in a microwave vial. Ethyl 2-(2-aminoethyl)-6-methyl-1-oxo-3,4- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (42.00 mg, 0.1583 mmol) was then added in anhydrous DCM (0.50 mL, 0.1300 M) and the the reaction mixture was heated at 60 °C by microwave irradiation for 2 h. Volatiles were removed under reduced pressure. The crude was purified directly by reverse phase flash chromatography (10-80% MeOH in water) followed. One fraction was clean by LCMS, so this was run through an SCX-II column to afford clean product ethyl 6- methyl-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-1-oxo-3,4- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (20.3 mg, 27%, 0.0421 mmol). Further material was recovered from the other fractions after they were washed through an SCX-II column, evaporated to afford ethyl 6-methyl-2-[2-[[7-(5-methyl-1,2,4- oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-1-oxo-3,4-dihydro pyrrolo[1,2-a]pyrazine-7- carboxylate (11.7 mg, 15%, 0.0242 mmol) as a fine pale-yellow solid with 98% purity which was used in the next reaction. HPLC/MS m/z 475.2073 [M+H] + , Rt (AD): 2.16 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.89-8.86 (m, 1 H), 8.15 (dd, J = 8.5, 1.5 Hz, 1 H), 8.04 (t, J = 5.2 Hz, 1 H), 7.98 (d, J = 5.7 Hz, 1 H), 7.85 (d, J = 8.5 Hz, 1 H), 6.97 (d, J = 5.7 Hz, 1 H), 6.91 (s, 1 H), 4.17 (q, J = 7.1 Hz, 2H), 4.08-4.03 (m, 2H), 3.78- 3.69 (m, 6H), 2.70 (s, 3H), 2.44 (s, 3H), 1.26 (t, J = 7.1 Hz, 3H).

Example 55: 6-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-2- propoxy-5H-pyrrolo[3,4-b]pyridin-7-one

Example 55.1 : Methyl 6-chloro-3-methyl-pyridine-2-carboxylate (1000.00 mg, 5.3876 mmol) was dissolved in anhydrous DCE (53.88 mL, 0.1000 M) under argon. AIBN (88.47 mg, 0.5388 mmol) and NBS (958.89 mg, 5.3876 mmol) were added, and the reaction mixture was heated to reflux for 2 h. The reaction mixture was cooled to ambient temperature, evaporated onto silica gel and purified by silica gel normal phase chromatography (0-30% EtOAc in cyclohexane) to give methyl 3- (bromomethyl)-6-chloro-pyridine-2-carboxylate (1.12 g, 79%, 4.2457 mmol) as a colourless, crystalline solid. HPLC/MS m/z 287.9862 [M+Na] + , Rt (AJ): 2.07 min. 1 H NMR (600 MHz, DMSO-cfe): 6 8.14 (d, J = 8.3 Hz, 1 H). 7.78 (d, J = 8.3 Hz, 1 H), 4.94 (s, 2H), 3.91 (s, 3H).

Example 55.2: Methyl 3-(bromomethyl)-6-chloro-pyridine-2-carboxylate (1.12 g, 3.3875 mmol), DIPEA (1.18 mL, 6.775 mmol) and 1-Boc-ethylenediamine (0.56 mL, 3.5569 mmol) were mixed in anhydrous MeCN (33.88 mL, 0.1000 M) under argon and heated at reflux for 1.5 h. The reaction mixture was cooled to ambient temperature, evaporated onto silica gel and purified by silica gel normal phase flash chromatography (0-100% EtOAc in cyclohexane) to give tert-butyl N-[2-(2-chloro-7- oxo-5H-pyrrolo[3,4-b]pyridin-6-yl)ethyl]carbamate (748 mg, 71%, 2.3993 mmol) as a colourless, crystalline solid. HPLC/MS m/z 334.1697 [M+Na] + , Rt (AJ): 1.99 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.15 (d, J = 8.1 Hz, 1 H), 7.68 (d, J = 8.1 Hz, 1 H), 6.94 (t, J = 6.1 Hz, 1 H), 4.51 (s, 2H), 3.58 (t, J = 5.9 Hz, 2H), 3.18-3.23 (m, 2H), 1.27 (s, 9H). Example 55.3: tert-Butyl N-[2-(2-chloro-7-oxo-5H-pyrrolo[3,4-b]pyridin-6- yl)ethyl]carbamate (150.00 mg, 0.4811 mmol), cesium carbonate (220.83 mg, 0.6736 mmol), tBuBrettPhos Pd G3 (17.13 mg, 0.0192 mmol), and 1-propanol (0.05 mL, 0.6255 mmol) were suspended in 1,4-dioxane (1.20 mL, 0.4000 M) under N2. The mixture was then evacuated and backfilled with N2 (x 5, 15 sec evacuation). The mixture was then heated to 80 °C and stirred (1200 RPM) under N2. The reaction was left to stir overnight. The reaction mixture was dried under reduced pressure and subjected to silica gel normal phase column chromatography (0-100% EtOAc:CycHex). Fractions were combined, evaporated to afford tert-butyl N-[2-(7- oxo-2-propoxy-5H-pyrrolo[3,4-b]pyridin-6-yl)ethyl]carbamate (137 mg, 85%, 0.4085 mmol) as a clear viscous oil. HPLC/MS m/z 358.1727 [M+H] + , Rt (AI): 1.42 min. 1 H NMR (600 MHz, Chloroform-d): 5 7.68 (d, J = 8.4 Hz, 1H), 6.90 (d, J = 8.4 Hz, 1 H), 4.99 (s, 1 H), 4.44 (t, J = 6.8 Hz, 2H), 4.40 (s, 2H), 3.79 (t, J = 6.0 Hz, 2H), 3.47 (q, J = 6.0 Hz, 2H), 1.83 (h, J = 7.2 Hz, 2H), 1.36 (s, 9H), 1.05 (t, J = 7.4 Hz, 3H). Example 55.4: tert-Butyl /V-[2-(7-oxo-2-propoxy-5H-pyrrolo[3,4-b]pyridin-6- yl)ethyl]carbamate (130.00 mg, 0.3876 mmol) was mixed with 4 M HCI in dioxane (9.69 mL, 38.76 mmol) and anyhydrous 1,4-dioxane (10.00 mL, 0.0400 M) at room temperature under argon and stirred for overnight. Volatiles were removed under reduced pressure. The crude was dissolved in MeOH/water and filtered through a 1 g SCX column. The product was released with 2N ammonia in MeOH to give 6-(2- aminoethyl)-2-propoxy-5H-pyrrolo[3,4-b]pyridin-7-one (74.1 mg, 81%, 0.3149 mmol) as an off-white solid. HPLC/MS mlz 258.1211 [M+H] + , Rt (Al): 0.79 min.

Example 55.5: 6-(2-Aminoethyl)-2-propoxy-5H-pyrrolo[3,4-b]pyridin-7-one (30.00 mg, 0.1275 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1,2,4-oxadiazole [Intermediate A5] (34.77 mg, 0.1530 mmol), PyBrop (71.33 mg, 0.1530 mmol), N,N- diisopropylethylamine (0.08 mL, 0.4782 mmol) and anhydrous DCM (0.64 mL, 0.2000 M) in a microwave vial at room temperature under nitrogen. The reaction mixture was heated at 60 °C by microwave irradiation for 1 h. Volatiles were removed under reduced pressure. The crude was purified directly by reverse phase flash chromatography (10-60% MeOH in water). Fractions containing product were filtered through a 1g SCX column. The product was released with ammonia in MeOH to give 6-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino ]ethyl]-2- propoxy-5H-pyrrolo[3,4-b]pyridin-7-one (15.3 mg, 27%, 0.0344 mmol) as an amorphous solid. HPLC/MS mlz 445.1993 [M+H] + , Rt (AJ): 2.14 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.83 (d, J = 1.6 Hz, 1H), 8.14 (dd, J = 8.4, 1.5 Hz, 1H), 8.03 (t, J = 5.6 Hz, 1H), 7.93 (dd, J = 13.3, 7.0 Hz, 2H), 7.84 (d, J = 8.5 Hz, 1 H), 6.98-6.93 (m, 2H), 4.48 (s, 2H), 4.23 (t, J = 6.7 Hz, 2H), 3.85 (t, J = 5.8 Hz, 2H), 3.79 (q, J = 5.7 Hz, 2H), 2.68 (s, 3H), 1.72 (h, J = 7.1 Hz, 2H), 0.96 (t, J = 7.4 Hz, 3H).

Example 56: 6-(5-M ethyl- 1 ,3,4-oxadiazol-2-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)-1-isoquinolyl]amino]ethyl]isoindolin-1-one

Example 56.1: A solution of methyl 5-bromo-2-(bromomethyl)benzoate (3000.00 mg, 9.7412 mmol) and 1-Boc-ethylenediamine (1794.73 mg, 11.202 mmol), triethylamine (2.06 mL, 14.612 mmol) in MeOH (50.00 mL, 0.1900 M) was refluxed under nitrogen overnight. After being cooled down to room temperature, the reaction mixture was concentrated in vacuo to give the crude product, which was purified by silica gel normal phase column chromatography (eluting with 20-60% EtOAc:CycHex) to give tert-butyl N-[2-(6-bromo-1-oxo-isoindolin-2-yl)ethyl]carbamate (2372.5 mg, 69%, 6.6788 mmol) as a white solid. HPLC/MS m/z 377.037, 379.035 [M+Na] + , Rt (Al): 1.40 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 7.88 (d, J = 1.9 Hz, 1H), 7.74 (dd, J = 8.1 , 1.9 Hz, 1 H), 7.50 (d, J = 8.0 Hz, 1 H), 4.54 (s, 2H), 3.69 (dd, J = 6.5, 5.1 Hz, 2H), 3.35 (dd, J = 6.5, 5.1 Hz, 2H), 1.31 (s, 9H).

Example 56.2: tert-Butyl N-[2-(6-bromo-1-oxo-isoindolin-2-yl)ethyl]carbamate (400.00 mg, 1.126 mmol), Xantphos Palladacycle Gen. 4 (21.70 mg, 0.0225 mmol) were mixed in a 5 mL microwave vial. The vial was capped, flushed with argon and evacuated before being fitted with a balloon filled with carbon monoxide (63.08 mg, 2.2521 mmol). Anyhydrous 1,4-dioxane (2.25 mL, 0.5000 M), and acethydrazide (150.15 mg, 2.0269 mmol) were added and the reaction mixture was heated at 90 °C overnight. To this crude product was added /V,/V-diisopropylethylamine (0.20 mL, 1.126 mmol) and p-toluene-sulfonyl-chloride (214.67 mg, 1.126 mmol). After 2 h another portion of p-toluene-sulfonyl-chloride (214.67 mg, 1.126 mmol) was added, and the reaction was stirred overnight. The crude mixture was subjected to silica gel normal phase column chromatography (0-100% EtOAc:CycHex -> 10% MeOH:EtOAc) to give tert-butyl N-[2-[6-(5-methyl-1,3,4-oxadiazol-2-yl)-1-oxo- isoindolin-2-yl]ethyl]carbamate (195.6 mg, 48%, 0.5458 mmol) as a fine cream coloured solid. HPLC/MS mlz 381.1517 [M+Na] + , Rt (Al): 1.22 min. 1 H NMR (600 MHz, Chloroform-d): 5 8.42 (d, J = 1.5 Hz, 1H), 8.29 (dd, J = 8.0, 1.6 Hz, 1H), 7.60 (dd, J = 7.8, 1.0 Hz, 1 H), 4.88 (d, J = 7.1 Hz, 1H), 4.57 (s, 2H), 3.77 (t, J = 6.0 Hz, 2H), 3.46 (q, J = 6.0 Hz, 2H), 2.64 (s, 3H), 1.31 (s, 9H).

Example 56.3: tert-Butyl N-[2-[6-(5-methyl-1 ,3,4-oxadiazol-2-yl)-1-oxo-isoindolin-2- yl]ethyl]carbamate (150.00 mg, 0.4185 mmol) was mixed with 4 M HCI in dioxane (10.46 mL, 41.854 mmol) and anyhydrous 1,4-dioxane (10.00 mL, 0.0400 M) at room temperature under argon and stirred for 2 h. Volatiles were removed under reduced pressure. The crude was dissolved in MeOH/water and filtered through a 2 g SCX column. The product was released with 2M ammonia in MeOH to give 2-(2- aminoethyl)-6-(5-methyl-1,3,4-oxadiazol-2-yl)isoindolin-1-on e (93 mg, 86%, 0.3601 mmol) as an off-white solid. HPLC/MS mlz 259.1211 [M+H] + , Rt (AI): 0.61 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.19 (dd, J = 7.9, 1.6 Hz, 1H), 8.14 (d, J = 1.5 Hz, 1 H), 7.86-7.80 (m, 1 H), 4.65 (s, 2H), 3.54 (t, J = 6.4 Hz, 2H), 2.82 (t, J = 6.4 Hz, 2H), 2.61 (s, 3H).

Example 56.4: 2-(2-Aminoethyl)-6-(5-methyl-1,3,4-oxadiazol-2-yl)isoindolin -1-one (45.00 mg, 0.1742 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4- oxadiazole [Intermediate A5] (47.51 mg, 0.2091 mmol), PyBrop (97.47 mg, 0.2091 mmol), /V,/V-diisopropylethylamine (0.11 mL, 0.6534 mmol) and anhydrous DCM (0.87 mL, 0.2000 M) in a microwave vial at room temperature under nitrogen. The reaction mixture was heated at 60 °C by microwave irradiation for 2 h. Volatiles were removed under reduced pressure. The crude was purified directly by reverse phase flash chromatography (10-60% MeOH in water). All fractions contained an impurity so all fractions containing product were filtered through a 2 g SCX column, the product was released with ammonia in MeOH, and the solvent evaporated in vacuo. The crude mixture was then subjected to silica gel normal phase column chromatography (20-100%EtOAc:CycHex -> 10% MeOH:EtOAc). The fractions containing pure product were collected and evaporated in vacuo to afford 6-(5- methyl-1,3,4-oxadiazol-2-yl)-2-[2-[[7-(5-methyl-1,2,4-oxadia zol-3-yl)-1- isoquinolyl]amino]ethyl]isoindolin-1-one (19.3 mg, 24%, 0.0413 mmol) as a fine white solid. HPLC/MS m/z 468.1758 [M+H] + , Rt (AJ): 1.84 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.85-8.81 (m, 1H), 8.18 (dd, J = 7.9, 1.6 Hz, 1 H), 8.14 (dd, J= 8.5, 1.5 Hz, 1H), 8.10 (dd, J = 1.5, 0.7 Hz, 1 H), 8.04 (t, J = 5.6 Hz, 1 H), 7.95 (d, J = 5.7 Hz, 1 H), 7.87-7.80 (m, 2H), 6.95 (dd, J = 5.9, 0.8 Hz, 1H), 4.70 (s, 2H), 3.88 (dd, J = 6.6, 5.1 Hz, 2H), 3.82 (q, J = 5.6 Hz, 2H), 2.67 (s, 3H), 2.59 (s, 3H).

Example 57: Propan-2-yl 6-methyl-2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-1-oxo-3,4-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate

Example 57.1: /V-iodosuccinimide (3493.46 mg, 15.668 mmol) was slowly added portion-wise to a solution of ethyl 5-methyl-1H-pyrrole-2-carboxylate (2400.00 mg, 15.668 mmol) in anhydrous DCM (31.34 mL, 0.5000 M) at 0 °C. The reaction was stirred for 2 h at 0 °C and then allowed to warm to room temperature and stirred overnight. The reaction mixture was concentrated, and the crude mixture was separated by silica gel normal phase column chromatography (0-20% EtOAc:CycHex) to afford ethyl 4-iodo-5-methyl-1H-pyrrole-2-carboxylate (4.12 g, 94%, 14.766 mmol) as a fine white solid. HPLC/MS mlz 251.9519 [M+H-CH 2 CH 3 ] + , Rt (AC): 1.36 min. 1 H NMR (600 MHz, Chloroform-d): 5 9.29 (s, 1H), 6.93 (d, J = 2.6 Hz, 1H), 4.30 (q, J = 7.1 Hz, 2H), 2.30 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

Example 57.2: A solution of ethyl 4-iodo-5-methyl-1 H-pyrrole-2-carboxylate (4000.00 mg, 14.333 mmol), tert-Butyl-/V-hydroxyethyl carbamate (3.99 mL, 25.8 mmol), and triphenylphosphine (6015.06 mg, 22.933 mmol) in anhydrous THF (35.83 mL, 0.4000 M) was cooled to 0 °C. diisopropyl azodicarboxylate (8.47 mL, 43 mmol) was added dropwise over 20 min, and the resulting mixture was warmed to room temperature and stirred for 18 h. The crude product was evaporated under reduced pressure and subjected to normal phase column chromatography (0-10% EtOAc:Cychex) to afford an impure ethyl 1-[2-(tert-butoxycarbonylamino)ethyl]-4- iodo-5-methyl-pyrrole-2-carboxylate (3.65 g, 57%, 8.2127 mmol) as a cream coloured solid which was used in the next step without further purification. HPLC/MS mlz 445.0591 [M+Na] + , Rt (Al): 1.69 min. 1 H NMR (500 MHz, Chloroform-d): 5 7.07 (s, 1H), 4.43 (t, J = Q.2 Hz, 2H), 4.26 (dq, J = 21.4, 7.1 Hz, 2H), 3.40 (q, J= 6.1 Hz, 2H), 2.30 (d, J = 7.2 Hz, 3H), 1.41 (s, 9H), 1.37-1.26 (m, 3H). Example 57.3: Ethyl 1-[2-(tert-butoxycarbonylamino)ethyl]-4-iodo-5-methyl-pyrrol e- 2-carboxylate (2.85 g, 6.7494 mmol) was mixed with 4 N HCI in 1,4-dioxane (67.49 mL, 269.98 mmol) and anhydrous 1,4-dioxane (67.53 mL, 0.0700 M) at room temperature under argon and stirred for 2 h. Volatiles were removed under reduced pressure. The crude was dissolved in EtOH (26.87 mL, 0.0700 M) and triethylamine (4.75 mL, 33.747 mmol) and the mixture was stirred for 72 h at 80 °C. The crude mixture was evaporated under reduced pressure, absorbed onto silica and subjected to a silica gel normal phase column chromatography (0-10% MeOH:DCM). The fractions were collected and evaporated to afford 7-iodo-6- methyl-3,4-dihydro-2H-pyrrolo[1,2-a]pyrazin-1-one (1.34 g, 72%, 4.8372 mmol) as a cream coloured solid. HPLC/MS m/z 276.9833 [M+H] + , Rt (Al): 1.14 min.

Example 57.4: 7-lodo-6-methyl-3,4-dihydro-2H-pyrrolo[1 ,2-a]pyrazin-1-one (700.00 mg, 2.5356 mmol), Xantphos Palladacycle Gen. 4 (146.56 mg, 0.1521 mmol) were mixed in a 20 mL microwave vial. The vial was capped, flushed with argon and evacuated before being fitted with a balloon filled with carbon monoxide (717.39 mg, 25.356 mmol), anyhydrous 1,4-dioxane (8.45 mL, 0.3000 M), 2-propanol (9.71 mL, 126.78 mmol) were added, and the reaction mixture was heated at 80 °C overnight. The reaction was evaporated under reduced pressure and the crude was subjected to silica gel normal phase column chromatography (0-10% DCM:MeOH) to afford a semi-pure propan-2-yl 6-methyl-1-oxo-3,4-dihydro-2H-pyrrolo[1 ,2-a]pyrazine-7- carboxylate (300 mg, 50%, 1.2697 mmol) as a dark brown amorphous solid.

HPLC/MS m/z 237.1241 [M+H] + , Rt (Al): 1.13 min. The product was used in the next step without further purification.

Example 57.5: To a solution of propan-2-yl 6-methyl-1-oxo-3,4-dihydro-2H- pyrrolo[1,2-a]pyrazine-7-carboxylate (360.00 mg, 1.5237 mmol) in THF (15.24 mL, 0.1000 M) at 0 °C was added NaH (134.08 mg, 3.3521 mmol) and the mixture was stirred and allowed to warm to room temperature over 1 h. Bromoacetonitrile (0.35 mL, 5.0281 mmol) was then added and the reaction was allowed to stir at room temperature overnight. The crude was evaporated and subjected to silica gel normal phase column chromatography (0-10% MeOH:DCM). The pure fractions were collected and evaporated to afford propan-2-yl 2-(cyanomethyl)-6-methyl-1-oxo-3,4- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (165.4 mg, 39%, 0.6008 mmol) as a pale-yellow solid. HPLC/MS m/z 276.1347 [M+H] + , Rt (Al): 1.17 min. 1 H NMR (500 MHz, DMSO-d 6 ): 5 7.00 (s, 1 H), 5.03 (p, J = 6.2 Hz, 1H), 4.55 (s, 2H), 4.19-4.13 (m, 2H), 3.83-3.74 (m, 2H), 2.47 (s, 3H), 1.26 (d, J = 6.3 Hz, 6H). Example 57.6: To a cold solution (0 °C) of propan-2-yl 2-(cyanomethyl)-6-methyl-1- oxo-3, 4-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (150.00 mg, 0.5449 mmol) and cobalt(ll) chloride hexahydrate (259.28 mg, 1.0897 mmol) in MeOH (10.90 mL, 0.0500 M) was added NaBH4 (288.07 mg, 5.4486 mmol) (portion-wise during 10 min). The mixture was poured into hydrochloric acid (1 N, 15 mL) and stirred until the black precipitate had dissolved. The aqueous layer was made alkaline with ammonia, extracted with CHCI3, dried, filtered and concentrated to give propan-2-yl 2-(2-aminoethyl)-6-methyl-1-oxo-3,4-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (135.5 mg, 89%, 0.4851 mmol) as a yellow oil. HPLC/MS m/z 280.1659 [M+H] + , Rt (Al): 0.90 min.

Example 57.7: 5-Methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole [Intermediate A5] (87.85 mg, 0.3866 mmol), bromotri(pyrrolidino)phosphonium hexafluorophosphate (180.24 mg, 0.3866 mmol) and DI PEA (0.21 mL, 1.2082 mmol) in DCM (1.91 mL, 0.1200 M) were stirred under a nitrogen atmosphere for 30 mins at 40 °C in a microwave vial. Propan-2-yl 2-(2-aminoethyl)-6-methyl-1-oxo-3,4- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (90.00 mg, 0.3222 mmol) was then added in DCM (0.77 mL, 0.1200 M) and the reaction was heated at 60 °C by microwave irradiation for 2 h. Volatiles were removed under reduced pressure. The crude was purified directly by reverse phase flash chromatography (10-80% MeOH in water). Fractions combined, run through an SCX-II column and subjected to another reverse phase flash chromatography (10-80% MeOH in water). Fractions still remain impure, SCX-II, evaporated and then subjected for a 3rd time to reverse phase flash chromatography (10-80% MeOH in water). The pure fractions were run through an SCX-II column, released with NH3 in water and evaporated to afford clean product propan-2-yl 6-methyl-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-1-oxo-3,4-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (15.01 mg, 9%, 0.0305 mmol) as a pale-yellow solid. (99.3% purity by UV).

HPLC/MS m/z 489.2237 [M+H] + , Rt (AJ): 2.19 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.90-8.85 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.03 (t, J = 5.2 Hz, 1 H), 7.97 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.96 (d, J = 5.7 Hz, 1 H), 6.89 (s, 1 H), 5.00 (hept, J = 6.3 Hz, 1 H), 4.07-3.99 (m, 2H), 3.78-3.68 (m, 6H), 2.69 (s, 3H), 2.43 (s, 3H), 1.24 (d, J = 6.2 Hz, 6H).

Example 58: N-[2-[2-(Ethoxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridin-6 -yl]ethyl]-

7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-amine

Example 58.1: 1M Lithium aluminium hydride solution in THF (0.97 mL, 0.9706 mmol) was added dropwise to a solution of 6-(tert-butoxycarbonyl)-4, 5,6,7- tetrahydrothieno[2,3-c]pyridine-2-carboxylic acid (250.00 mg, 0.8823 mmol) in THF (3.68 mL) under a nitrogen atmosphere. The reaction was cooled to 0 °C and stirred for 15 min before warming room temperature and stirred for an additional 1 h. The reaction mixture was carefully quenched with 40 uL of water, followed by 40 uL of 15% NaOH aq. sol and 120 uL of water. After the quenching procedure, the reaction mixture was diluted with diethyl ether (~3 mL) and some anhydrous sodium sulfate was added. This mixture was stirred for 15 min before it was filtered off through a Telso phase separator and washed off with diethyl ether and DCM. The obtained filtrate was evaporated to dryness to give the crude product. The crude has been dissolved in EtOAc (10 mL) and washed 2 times with HCI 0.5M (5 mL). Organic layer has been dried on Na2SO4 and evaporated to give tert-butyl 2- (hydroxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-6-carbo xylate (133 mg, 45%, 0.3950 mmol) as slightly yellow oil. HPLC/MS mlz 292.0849 [M+Na] + , Rt (Al): 1.38 min. 1 H NMR (600 MHz, Chloroform-d): 56.69 (d, J = 5.6 Hz, 1H), 4.74 (d, J = 5.5 Hz, 2H), 4.57 (d, J = 5.3 Hz, 2H), 3.65 (t, J = 7.0 Hz, 2H), 2.67-2.62 (m, 2H), 1.47 (d, J = 3.1 Hz, 9H).

Example 58.2: Sodium hydride (21.24 mg, 0.5309 mmol) was added to a solution of tert-butyl 2-(hydroxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-6-car boxylate (144.44 mg, 0.4826 mmol) in DMF (0.90 mL) under ice cooling (colourless solution turned yellow). After stirring at 0 °C for 5 min, iodoethane (0.04 mL, 0.5309 mmol) was added and stirring was continued for a further 4 h. The reaction mixture was partitioned between water (20 mL) and ethyl acetate (30 mL). After phase separation the aqueous layer was further extracted with ethyl acetate (2x30 mL). The combined organic layers were washed with brine (40 mL), dried (MgSC>4), filtered and evaporated to dryness. The obtained crude (79.8 mg) was purified by normal phase silica column chromatography using a 10 g KP-silica column and a gradient of 0-50% EtOAc in cyclohexane as eluant. Fractions were combined and evaporated to dryness to give the required product tert-butyl 2-(ethoxymethyl)-5,7- dihydro-4H-thieno[2,3-c]pyridine-6-carboxylate (87 mg, 61%, 0.2925 mmol) as clear oil. HPLC/MS m/z 320.1118 [M+Na] + , Rt (AI): 1.59 min. 1 H N MR (600 MHz, Chloroform-d): 5 6.67 (s, 1 H), 4.56 (d, J = 5.6 Hz, 4H), 3.69-3.61 (m, 2H), 3.53 (q, J = 7.0 Hz, 2H), 2.64 (t, J = 6.1 Hz, 2H), 1.47 (s, 9H), 1.22 (t, J = 7.0 Hz, 3H). Example 58.3: tert-Butyl 2-(ethoxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-6- carboxylate (87.00 mg, 0.2925 mmol) was dissolved in dry 1 ,4-dioxane (1.54 mL). Hydrogen chloride (4 M in 1,4-dioxane) (1.10 mL, 4.3879 mmol) was added, stirred at room temperature overnight. The volatiles were evaporated to dryness. The obtained salt 2-(ethoxymethyl)-4,5,6,7-tetrahydrothieno[2,3-c]pyridine hydrochloride (67 mg, 49%, 0.1439 mmol) (beige powder) was taken through to the next step without further purification. HPLC/MS m/z 198.0871 [M+H] + , Rt (Al): 0.52 min. Example 58.4: 2-(Ethoxymethyl)-4,5,6,7-tetrahydrothieno[2,3-c]pyridine hydrochloride (81.00 mg, 0.3465 mmol) was dissolved in CHCI3 (1.73 mL).

Triethylamine (0.05 mL, 0.3465 mmol) was added, and the reaction mixture was cooled to 0 °C. A 1M solution of 1 -nitroethene in xylene (0.35 mL, 0.3465 mmol) was added and the reaction was stirred at 0 °C for 10 min. The reaction was then stirred at room temperature for 1h 40 min. The reaction mixture was concentrated in vacuo. The obtained crude (53.5 mg) was purified by silica gel column chromatography using a Biotage KP-NH 12 g column (eluent: 20-80% EtOAc in cyclohexane). Fractions were combined and evaporated to dryness to give colourless oil 2-(ethoxymethyl)-6-(2-nitroethyl)-5,7-dihydro-4H-thieno[2,3- c]pyridine (36 mg, 37%, 0.1265 mmol) as a white solid. HPLC/MS m/z 271.1031 [M+H] + , Rt (Al): 0.63 min. 1 H NMR (500 MHz, Chloroform-d): 5 6.66 (s, 1 H), 4.56-4.52 (m, 4H), 3.75 (t, J = 1.7 Hz, 2H), 3.52 (q, J = 7.0 Hz, 2H), 3.24-3.18 (m, 2H), 2.85 (t, J = 5.8 Hz, 2H), 2.69-2.61 (m, 2H), 1.21 (t, J = 7.0 Hz, 3H).

Example 58.5: 2-(Ethoxymethyl)-6-(2-nitroethyl)-5,7-dihydro-4H-thieno[2,3- c]pyridine (27.00 mg, 0.0999 mmol), Zn powder (65.30 mg, 0.9987 mmol) and ammonium chloride (53.42 mg, 0.9987 mmol) were suspended in a mixture of THF/water/EtOH (0.9 mL/0.12 mL/0.12 mL). The reaction mixture was stirred at room temperature for 1h 40 min. The mixture was filtered through celite, rinsed with a 1:1 mixture of DCM:EtOH. The filtrate was passed through a 2 g Biotage NH2 ion exachange column and rinsed with a 1:1 mixture of DCM:EtOH. The filtrate was concentrated to give a pale-yellow oil 2-[2-(ethoxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridin-6- yl]ethanamine (25 mg, 92%, 0.0915 mmol). HPLC/MS m/z 241.1235 [M+H] + , Rt (Al): 0.30 min. 1 H NMR (600 MHz, Chloroform-d): 5 6.67 (s, 1 H), 4.55 (s, 2H), 3.76 (t, J = 1.6 Hz, 2H), 3.52 (q, J = 7.0 Hz, 2H), 2.93 (dd, J = 6.7, 5.1 Hz, 2H), 2.85 (t, J = 5.8 Hz, 2H), 2.75-2.69 (m, 5H), 1.21 (t, J = 7.0 Hz, 3H).

Example 58.6: 2-[2-(Ethoxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridin-6- yl]ethanamine (33.00 mg, 0.1373 mmol) was dissolved in dry DCM (1.06 mL, 0.1300 M) then DI PEA (95.65 uL, 0.5492 mmol) was added followed by 5-methyl-3-(2- oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole [Intermediate A5] (37.43 mg, 0.1647 mmol) and PyBrop (96.00 mg, 0.2059 mmol). The mixture was stirred on at room temperature under argon overnight. Volatiles were removed under reduced pressure and the crude was purified by prep. HPLC (AccqPrep, gradient ACN/water pH 11) to give N-[2-[2-(ethoxymethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridin-6 -yl]ethyl]-7-(5- methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-amine (5 mg, 8%, 0.0111 mmol) as an off- white solid. HPLC/MS m/z 450.1955 [M+H] + , Rt (AJ): 2.08 min. 1 H NMR (600 MHz, Chloroform-d): 5 8.55-8.53 (m, 1 H), 8.21 (dd, J = 8.5, 1.5 Hz, 1 H), 8.06 (d, J = 5.8 Hz, 1 H), 7.74 (d, J = 8.5 Hz, 1 H), 6.94 (dd, J = 5.9, 0.9 Hz, 1 H), 6.69 (s, 1 H), 6.16 (s, 1 H), 4.56 (s, 2H), 3.82-3.75 (m, 4H), 3.53 (q, J = 7.0 Hz, 2H), 2.96 (t, J = 6.0 Hz, 2H), 2.90 (t, J = 5.8 Hz, 2H), 2.76-2.71 (m, 2H), 2.67 (s, 3H), 1.21 (t, J = 7.0 Hz, 3H).

Example 59: 6-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)-1-isoquinolyl]amino]ethyl]isoindolin-1-one

Example 59.1 : A solution of 3-(1-chloro-7-isoquinolyl)-5-methyl-1 ,2,4-oxadiazole [Intermediate A4] (1000.00 mg, 4.0707 mmol) and ethylenediamine (5.44 mL, 81.413 mmol) in NMP (5.00 mL) in a 10-20 mL microwave vial was heated under microwave irradiation for 1 h at 160 °C. A solution of saturated NaHCO 3 was added (100 mL) and the product was extracted with EtOAc (3 x 50 mL). The organic layer was combined, dried over MgSC>4 and concentrated under reduced pressure to give N'-[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]ethane-1 ,2-diamine (982 mg, 90%, 3.6465 mmol) as a yellow solid. HPLC/MS m/z 270.1356 [M+H] + , Rt (AJ): 0.95 min. 1 H NMR (600 MHz, DMSO-d 6 ): 6 8.91-8.92 (m, 1 H), 8.13 (dd, J = 8.5, 1.6 Hz, 1H), 7.94 (d, J = 5.7 Hz, 1 H), 7.83 (d, J = 8.5 Hz, 1H), 7.79 (t, J = 5.4 Hz, 1H), 6.92 (dd, J = 5.9, 0.8 Hz, 1 H), 3.50 (td, J = 6.4, 5.2 Hz, 2H), 2.81 (t, J = 6.5 Hz, 2H), 2.70 (s, 3H), 1.67 (br s, 2H).

Example 59.2: A solution of methyl 2-(bromomethyl)-5-cyano-benzoate (215.00 mg, 0.8462 mmol) and /V'-[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]ethane-1,2- diamine (250.67 mg, 0.9308 mmol), triethylamine (0.18 mL, 1.2693 mmol) in MeOH (4.23 mL, 0.2000 M) was refluxed under N2 overnight. After being cooled down to room temperature, the reaction mixture was filtered, and the filtrate was concentrated in vacuo to give the crude product, which was washed several times with EtOAc to afford 2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-3-oxo-isoindoline-5-carbonitrile (310.8 mg, 89%, 0.7573 mmol) as a white solid. HPLC/MS m/z 411.1486 [M+H] + , Rt (Al): 1.05 min.

Example 59.3: 2-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-3- oxo-isoindoline-5-carbonitrile (100.00 mg, 0.2436 mmol), hydroxylamine hydrochloride (33.86 mg, 0.4873 mmol) and TEA (0.07 mL, 0.4873 mmol) were mixed in [bmim]OAc (0.24 mL, 1 M) under argon. The reaction mixture was heated at 80 °C for 1.5 h. The reaction mixture was cooled to ambient temperature, mixed with EtOAc (30 mL). The organic layer was mixed with saturated NaCI (50 mL) and the organic layer was extracted. The aqueous layer was extracted further with EtOAc (2 x 30 mL). The combined organic layer was washed with saturated NaCI (3 x 20 mL) and subsequently concentrated under reduced pressure to afford N- hydroxy-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-3-oxo- isoindoline-5-carboxamidine (95 mg, 62%, 0.1500 mmol) which was used for the next step without further purification. HPLC/MS m/z 444.1725 [M+H] + , Rt (Al): 0.76 min.

Example 59.4: /V-Hydroxy-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-3-oxo-isoindoline-5-carboxamidine (80.00 mg, 0.1263 mmol), acetic anhydride (0.02 mL, 0.2526 mmol) and anhydrous MeCN (1.50 mL, 0.0800 M) were mixed in a microwave vial under argon. The reaction mixture was heated at 160 °C for 10 min by microwave irradiation. Volatiles were removed under reduced pressure. The crude was directly purified by prep. HPLC (AccqPrep, 30- 43% MeOH in water, pH3, 26 min). The fractions containing product were filtered through a 1 g SCX2 column. The product was released with 2 N ammonia in MeOH to give 6-(5-methyl-1,2,4-oxadiazol-3-yl)-2-[2-[[7-(5-methyl-1,2,4-o xadiazol-3-yl)-1- isoquinolyl]amino]ethyl]isoindolin-1-one (23 mg, 39%, 0.0487 mmol) as a white powder. HPLC/MS m/z 468.1787 [M+H] + , Rt (AJ): 2.02 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.81-8.85 (m, 1H), 8.19 (dd, J = 7.9, 1.6 Hz, 1 H), 8.15-8.16 (m, 1H), 8.14 (dd, J = 8.5, 1.5 Hz, 1H), 8.04 (t, J = 5.6 Hz, 1 H), 7.95 (d, J = 5.7 Hz, 1H), 7.84 (d, J = 8.5 Hz, 1 H), 7.79 (dd, J = 7.8, 0.9 Hz, 1 H), 6.95 (dd, J = 5.8, 0.8 Hz, 1 H), 4.68 (s, 2H), 3.85-3.90 (m, 2H), 3.78-3.84 (m, 2H), 2.68 (s, 3H), 2.67 (s, 3H).

Example 60: 6-(3-M ethyl- 1 ,2,4-oxadiazol-5-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)-1-isoquinolyl]amino]ethyl]isoindolin-1-one

Example 60.1: tert-Butyl N-[2-(6-bromo-1-oxo-isoindolin-2-yl)ethyl]carbamate (350.00 mg, 0.9853 mmol), acetamide oxime (364.95 mg, 4.9264 mmol) and XantPhos Pd G4 (18.98 mg, 0.0197 mmol) were mixed in a microwave vial. The vial was capped, evacuated before being fitted with a balloon filled with carbon monoxide. Anyhydrous 1,4-dioxane (1.97 mL, 0.5000 M) and DIPEA (0.34 mL, 1.9706 mmol) were added, and the reaction mixture was heated at 90 °C for 20 h. Additional XantPhos Pd G4 (18.98 mg, 0.0197 mmol) was added and the reaction mixture was continued to be heated at 90 °C for 5 h. The reaction mixture was evaporated onto silica gel and directly purified by silica gel normal phase flash chromatography (30-100% EtOAc:CycHex) to give tert-butyl N-[2-[6-(3-methyl- 1,2,4- oxadiazol-5-yl)-1-oxo-isoindolin-2-yl]ethyl]carbamate (93 mg, 26%, 0.2595 mmol) as a white solid. HPLC/MS m/z 381.1517 [M+Na] + , Rt (AJ): 2.34 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.29 (dd, J = 7.9, 1.6 Hz, 1 H), 8.23 (d, J = 1.6 Hz, 1 H), 7.87 (d, J = 7.9 Hz, 1H), 6.96 (t, J = 6.1 Hz, 1H), 4.63 (s, 2H), 3.59 (t, J = 6.0 Hz, 2H), 3.21 (q, J = 6.1 Hz, 2H), 2.44 (s, 3H), 1.28 (s, 9H). Example 60.2: tert-Butyl N-[2-[6-(3-methyl-1 ,2,4-oxadiazol-5-yl)-1-oxo-isoindolin-2- yl]ethyl]carbamate (90.00 mg, 0.2511 mmol) was mixed with 4N HCI in 1,4-dioxane (6.28 mL, 25.112 mmol) and anyhydrous 1,4-dioxane (6.00 mL) at room temperature under argon and stirred for 3 d. Volatiles were removed under reduced pressure. The crude was dissolved in MeOH/water and filtered through a 1 g SCX column. The product was released with 2M ammonia in MeOH to give 2-(2- aminoethyl)-6-(3-methyl-1,2,4-oxadiazol-5-yl)isoindolin-1-on e (64 mg, 99%, 0.2478 mmol) as an off-white solid. HPLC/MS m/z 259.1191 [M+H] + , Rt (AJ): 1.07 min. 1 H NMR (600 MHz, DMSO-d 6 ): 6 8.29 (dd, J = 7.9, 1.6 Hz, 1 H), 8.23 (d, J = 1.5 Hz, 1H), 7.85 (dd, J = 7.9, 0.9 Hz, 1 H), 4.66 (s, 2H), 3.53 (t, J = 6.4 Hz, 2H), 2.80 (t, J = 6.4 Hz, 2H), 2.44 (s, 3H), 1.69 (s, 2H).

Example 60.3: 2-(2-Aminoethyl)-6-(3-methyl-1,2,4-oxadiazol-5-yl)isoindolin -1-one (36.42 mg, 0.1410 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4- oxadiazole [Intermediate A5] (35.24 mg, 0.1551 mmol), PyBroP (72.30 mg, 0.1551 mmol), DIPEA (68.33 mg, 0.5287 mmol) and anhydrous DCM (0.50 mL, 0.2500 M) in a microwave vial at room temperature under argon. The reaction mixture was heated at 60 °C by microwave irradiation for 1 h. Volatiles were removed, and the crude was directly purified by reverse phase flash chromatography (30-60% MeOH in water). Fractions containing product were filtered through a 1 g SCX-2 column. The product was released by 2M ammonia in MeOH to give 6-(3-methyl-1,2,4- oxadiazol-5-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]isoindolin-1-one (19 mg, 52%, 0.0404 mmol) as an off-white solid. HPLC/MS m/z 468.1786 [M+H] + , Rt (AJ): 2.01 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.82 (s, 1 H), 8.28 (dd, J = 7.9, 1.6 Hz, 1H), 8.19-8.21 (m, 1 H), 8.14 (dd, J = 8.4, 1.5 Hz, 1 H), 8.04 (t, J = 5.5 Hz, 1H), 7.94 (d, J = 5.7 Hz, 1H), 7.83-7.87 (m, 2H), 6.95 (d, J = 5.7 Hz, 1H), 4.71 (s, 2H), 3.86-3.90 (m, 2H), 3.80-3.84 (m, 2H), 2.67 (s, 3H), 2.43 (s, 3H).

Example 61: Ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1- oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate

Example 61.1 : To a solution of ethyl 1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine- 7-carboxylate (0.300 g, 1.44 mmol) in tetrahydrofuran (5 mL) was added sodium hydride (115 mg, 2.88 mmol, 60% purity) at 0 °C. The mixture was stirred at 0 °C for 0.5 h. Then to the mixture was added 2-bromoacetonitrile (518 mg, 4.32 mmol, 3.00 eq). The mixture was stirred at 20 °C for 12 h. The mixture was diluted with saturated ammonium chloride aqueous solution (10 mL) at 0 °C and extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with brine (3 x 10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 10/1 to 0/1) to afford ethyl 2- (cyanomethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (0.130 g, 526 umol, 36% yield) as a yellow solid. HPLC/MS m/z: 248.2 [M+H] + , Rt (V): 0.79 min. 1 H NMR (400 MHz, DMSO-d 6 ): 57.69 (d, J = 1.6 Hz, 1 H), 7.02 (d, J = 1.6 Hz, 1 H), 4.56 (s, 2H), 4.33-4.28 (m, 2H), 4.21 (q, J = 7.0 Hz, 2H), 3.84-3.77 (m, 2H), 1.27 (t, J = 7.1 Hz, 3H).

Example 61.2: To a solution of ethyl 2-(cyanomethyl)-1-oxo-1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (0.100 g, 404 umol) in a mixture solvent of methanol (10 mL) and ammonium hydroxide (1 mL) was added Raney- Nickel (10.0 mg, 10% purity). The mixture was stirred at 20 °C for 12 h under hydrogen atmosphere (45 Psi). The mixture was filtered and concentrated under reduced pressure to afford ethyl 2-(2-aminoethyl)-1-oxo-1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (80.0 mg, crude) as yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ): 67.60 (d, J = 1.6 Hz, 1 H), 6.88 (d, J = 1.6 Hz, 1 H), 4.19 (q, J = 7.0 Hz, 4H), 3.70 (br dd, J = 5.1 , 6.7 Hz, 2H), 3.42-3.38 (m, 2H), 2.76-2.65 (m, 2H), 1.25 (t, J = 7.1 Hz, 3H).

Example 61.3: To a solution of ethyl 2-(2-aminoethyl)-1-oxo-1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (40.0 mg, 159 umol) and 7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)isoquinoline 2-oxide (36.2 mg, 159 umol) in dichloromethane (5 mL) were added /V,/V-diisopropylethylamine (61.7 mg, 478 umol) and bromotri(pyrrolidin-1-yl)phosphonium hexafluorophosphate(V) (148 mg, 318 umol). The mixture was stirred at 20 °C for 12 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (ethyl acetate), prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um;mobile phase: [water(10mM NH 4 HCO 3 )-ACN]; B%: 26%-56%,8min) and prep-HPLC (column: llnisil 3-100 C18 Ultra 150*50mm*3 um;mobile phase: [water(0.225%FA)- ACN]; B%: 18%-38%,10min) to afford ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7- carboxylate (10.51 mg, 20.54 umol, 12% yield, 99% purity, formate) as a white solid. HPLC/MS m/z: 461.2 [M+H] + , Rt (T): 0.78 min. 1 H NMR (400 MHz, DMSO-d 6 ): 5 8.87 (s, 1 H), 8.34 (s, 1 H), 8.14 (dd, J = 1.1, 8.5 Hz, 1 H), 8.07-8.01 (m, 1 H), 7.96 (d, J = 5.6 Hz, 1 H), 7.85 (d, J = 8.5 Hz, 1 H), 7.58 (d, J = 1.5 Hz, 1 H), 6.96 (d, J = 5.8 Hz, 1 H), 6.90 (d, J = 1.6 Hz, 1 H), 4.18 (q, J = 6.9 Hz, 4H), 3.81-3.65 (m, 6H), 2.69 (s, 3H), 1.24 (t, J = 7.1 Hz, 3H).

Example 62: Propan-2-yl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate

Example 62.1 : To a solution of ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-1-oxo- 1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7- carboxylate (0.120 g, 261 umol) [Example 61.3] in a mixture solvent of tetra hydrofuran (2 mL) and methanol (2 mL) was added a solution of lithium hydroxide monohydrate (32.8 mg, 782 umol) in water (1 mL). The mixture was stirred at 70 °C for 12 h. After cooling to room temperature, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um;mobile phase: [water(10mM NH 4 HCO 3 )-ACN]; B%: 4%-34%,8min) to afford 2-(2-((7-(5-methyl- 1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-1-oxo-1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid (70.0 mg, 162 umol, 62% yield) as a yellow oil. HPLC/MS m/z: 433.4 [M+H] + , Rt (V): 0.81 min. Example 62.2: To a solution of 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid (50.0 mg, 116 umol) and potassium carbonate (16.0 mg, 116 umol) in DMF (2 mL) was added 2-bromopropane (42.7 mg, 347 umol). The mixture was stirred at 80 °C for 2 h. After cooling to room temperature, the reaction was filtered. The filtrate was purified by prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um;mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 30%-60%,8min), prep-HPLC (column: Phenomenex Synergi C18 150*25mm* 10um;mobile phase: [water(0.225%FA)-ACN]; B%: 12%-42%,10min) and lyophilized to afford propan-2-yl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-1-oxo-1 ,2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (18.39 mg, 35.0 umol, 30% yield, 99% purity, formate) as a white solid. HPLC/MS m/z: 475.2 [M+H] + , Rt (T): 0.79 min. 1 H NMR (400 MHz, DMSO-d 6 ): 68.87 (s, 1 H), 8.24 (s, 1 H), 8.14 (dd, J = 1.5, 8.5 Hz, 1 H), 8.07-8.01 (m, 1 H), 7.96 (d, J = 5.6 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.54 (d, J = 1.6 Hz, 1 H), 6.96 (d, J = 5.8 Hz, 1 H), 6.88 (d, J = 1.8 Hz, 1 H), 5.01 (sept, J = 6.3 Hz, 1 H), 4.22-4.12 (m, 2H), 3.76-3.69 (m, 6H), 2.69 (s, 3H), 1.24 (d, J = 6.3 Hz, 6H).

Example 63: Ethyl 5-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4-oxo-4,5,6,7-tetrahydrothieno[3,2-c]pyridin e-2-carboxylate

Example 63.1 : To a mixture of 2-bromo-6,7-dihydro-5/7-thieno[3,2-c]pyridin-4-one (800 mg, 3.45 mmol) in ethanol (20 mL) were added palladium acetate (77.4 mg, 345 umol), triethylamine (698 mg, 6.89 mmol), 4,5-bis(diphenylphosphino)-9,9- dimethylxanthene (598 mg, 1.03 mmol) and DMF (4 mL). The mixture was stirred at 70 °C for 12 h under carbon monoxide atmosphere (40 Psi). After the reaction was completed, the mixture was cooled to 25 °C and quenched by water (5 mL). The suspension was filtered, and the filtrate was concentrated to remove ethanol. The residue was extracted with ethyl acetate (3 * 10 mL). The combined organic layers were washed with brine (3 x 20 mL), dried over anhydrous sodium sulfate and concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 5/1 to 1/1) to afford ethyl 4-oxo-4, 5,6,7- tetrahydrothieno[3,2-c]pyridine-2-carboxylate (750 mg, 3.33 mmol, 97% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ): 57.79 (s, 2H), 4.28 (q, J = 7.2 Hz, 2H), 3.47 (dt, J = 2.8, 6.8 Hz, 2H), 3.06 (t, J = 6.8 Hz, 2H), 1.29 (t, J = 7.2 Hz, 3H). Example 63.2: To a suspension of sodium hydride (249 mg, 6.21 mmol, 60% purity) in tetrahydrofuran (7 mL) was added ethyl 4-oxo-4,5,6,7-tetrahydrothieno[3,2- c]pyridine-2-carboxylate (700 mg, 3.11 mmol). The mixture was stirred at 0 °C for 0.5 h. Then to the mixture was added 2-bromoacetonitrile (1.12 g, 9.32 mmol). The mixture was stirred at 25 °C for 2 h. The mixture was quenched with saturated ammonium chloride aqueous solution (5 mL). The mixture was concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 5/1 to 1/1) to afford ethyl 5-(cyanomethyl)-4-oxo-4, 5,6,7- tetrahydrothieno[3,2-c]pyridine-2-carboxylate (450 mg, 1.70 mmol, 54% yield) as a yellow solid. HPLC/MS m/z: 265.1 [M+H] + , Rt (T): 0.86 min. 1 H NMR (400 MHz, Chloroform-d): 58.08 (s, 1 H), 4.51 (s, 2H), 4.36 (q, J = 7.2 Hz, 2H), 3.82 (t, J = 6.8 Hz, 2H), 3.24 (t, J = 6.8 Hz, 2H), 1.38 (t, J = 7.2 Hz, 3H).

Example 63.3: A mixture of ethyl 5-(cyanomethyl)-4-oxo-4, 5,6,7- tetrahydrothieno[3,2-c]pyridine-2-carboxylate (250 mg, 946 umol), Raney-Nickel (81.0 mg, 94.6 umol, 10% purity) and ammonium hydroxide (1 mL) in ethanol (10 mL) was stirred at 25 °C for 12 h under hydrogen atmosphere (45 Psi). After the reaction was completed, the mixture was filtered, and the filtrate was concentrated to give a residue. The residue was purified by prep-TLC (ethyl acetate/methanol/ammonium hydroxide (25% purity) = 50/10 /1) to afford ethyl 5-(2- aminoethyl)-4-oxo-4,5,6,7-tetrahydrothieno[3,2-c]pyridine-2- carboxylate (75.0 mg, 280 umol, 29% yield) as a colorless solid. 1 H NMR (400 MHz, Chloroform-d): 58.00 (s, 1H), 4.34 (q, J = 7.2 Hz, 2H), 3.75 (t, J = 6.8 Hz, 2H), 3.71 (br t, J = 6.0 Hz, 2H), 3.17-3.10 (m, 4H), 1.37 (t, J = 7.2 Hz, 3H).

Example 63.4: A mixture of 7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinoline 2-oxide (50.0 mg, 220 umol), ethyl 5-(2-aminoethyl)-4-oxo-4,5,6,7-tetrahydrothieno[3,2- c]pyridine-2-carboxylate (59.0 mg, 220 umol), diisopropylethylamine (114 mg, 880 umol) and bromotri(pyrrolidin-1-yl)phosphonium hexafluorophosphate(V) (185 mg, 396 umol) in dichloromethane (5 mL) was stirred at 25 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC(column: Waters Xbridge 150*25mm* 5um; mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 46%-76%,10 min), prep-HPLC(column: Phenomenex Synergi C18 150*25mm* 10um; mobile phase: [water(0.225%FA)-ACN]; B%: 20%-47%,9 min) and lyophilized to afford ethyl 5-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-4- oxo-4,5,6,7-tetrahydrothieno[3,2-c]pyridine-2-carboxylate (16.99 mg, 32.1 umol, 14% yield, 99% purity, formate) as a colorless solid. HPLC/MS m/z: 478.2 [M+H] + , Rt (T): 0.84 min. 1 H NMR 1 H N MR (400 MHz, DMSO-d 6 ): 68.87 (s, 1 H), 8.20-8.11 (m, 1 H), 8.02 (br s, 1 H), 7.96 (d, J = 5.6 Hz, 1 H), 7.84 (d, J = 8.4 Hz, 1 H), 7.79 (s, 1 H), 6.95 (d, J = 5.6 Hz, 1 H), 4.28 (q, J = 7.2 Hz, 2H), 3.76-3.66 (m, 6H), 3.09 (t, J = 6.8 Hz, 2H), 2.69 (s, 3H), 1.28 (t, J = 7.2 Hz, 3H).

Example 64: 2-(2-((7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-7-

(5-methyl-1 ,3,4-oxadiazol-2-yl)-3,4-dihydropyrrolo[1 ,2-a]pyrazin-1(2/-/)-one

A mixture of 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-1-oxo- 1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid (40.0 mg, 92.5 umol) [Example 62.1] and acetohydrazide (8.91 mg, 120 umol) in polyphosphoric acid (0.5 mL) was stirred at 90 °C for 2 h. The reaction was diluted with ethyl acetate (10 mL). Then the pH of the mixture was adjusted to 8 with saturated sodium carbonate aqueous solution. The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25mm* 5um; mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 20%-50%, 9 min) and lyophilized to afford 2-(2-((7-(5- methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-7-(5-methyl-1 ,3,4-oxadiazol-2-yl)-3,4-dihydropyrrolo[1 ,2- a]pyrazin-1 (2/-/)-one (10.78 mg, 22.7 umol, 24% yield, 99% purity) as a colorless solid. HPLC/MS m/z: 471.3 [M+H] + , Rt (V): 0.90 min. 1 H NMR (400 MHz, DMSO-d 6 ): 68.92 (br s, 1 H), 8.21 (br s, 1 H), 7.94 (br d, J = 5.7 Hz, 1 H), 7.93-7.84 (m, 1 H), 7.68 (d, J = 1.7 Hz, 1 H), 7.02 (br d, J = 2.1 Hz, 1 H), 6.97 (d, J = 1.6 Hz, 1 H), 4.23 (br t, J = 5.6 Hz, 2H), 3.77 (br s, 6H), 2.69 (s, 3H), 2.50 (s, 3H).

Example 65: 2-(2-((7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-7-

(3-methyl-1 ,2,4-oxadiazol-5 -yl)-3,4-dihydropyrrolo[1 ,2-a]pyrazin-1(2/7)-one

Example 65.1 : To a solution of 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid (80.0 mg, 185 umol) [Example 62.1], 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (70.9 mg, 370 umol), 1 -hydroxybenzotriazole (37.5 mg, 278 umol) and /V,/V-diisopropylethylamine (71.7 mg, 555 umol, 96.7 uL) in acetonitrile (5 mL) was added /V-hydroxyacetamidine (27.4 mg, 370 umol). The mixture was stirred at 20 °C for 12 h. The mixture was concentrated to give a residue. The residue was triturated with water (3 mL) to afford /V'-((2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7- carbonyl)oxy)acetimidamide (90.0 mg, crude) as a yellow solid. HPLC/MS m/z: 489.3 [M+H] + , Rt (V): 0.89 min. 1 H NMR (400 MHz, DMSO-d 6 ): 58.88 (s, 1 H), 8.20- 8.11 (m, 1 H), 8.06 (br s, 1 H), 7.97 (d, J = 5.8 Hz, 1 H), 7.85 (d, J = 8.5 Hz, 1 H), 7.75 (d, J = 1.3 Hz, 1 H), 7.16 (d, J = 1.4 Hz, 1 H), 6.96 (d, J = 5.6 Hz, 1 H), 6.38 (br s, 3H), 4.16 (br t, J = 5.4 Hz, 2H), 3.74 (br s, 6H), 2.69 (s, 3H), 2.50 (br s, 3H).

Example 65.2: To a solution of /V'-((2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7- carbonyl)oxy)acetimidamide (60.0 mg, 123 umol) in tetrahydrofuran (3 mL) was added tetrabutylammonium fluoride (1 M in tetrahydrofuran, 368 uL, 368 umol). The mixture was stirred at 40 °C for 12 h. The mixture was diluted with saturated ammonium chloride aqueous solution (10 mL) and extracted with ethyl acetate (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (ethyl acetate), prep-HPLC (column: Waters Xbridge 150*25mm* 5um; mobile phase: [water(10mM NH4HCO3)- ACN]; B%: 29%-59%, 8 min) and lyophilized to afford 2-(2-((7-(5-methyl-1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-7-(3-methyl-1 ,2,4-oxadiazol-5 -yl)-3,4- dihydropyrrolo[1 ,2-a]pyrazin-1 (2/-/)-one (13.02 mg, 27.40 umol, 22% yield, 99% purity) as a colorless solid. HPLC/MS m/z: 471.3 [M+H] + , Rt (T): 0.77 min. 1 H NMR (400 MHz, DMSO-d 6 ): 68.87 (s, 1 H), 8.15 (dd, J = 1.3, 8.6 Hz, 1 H), 8.09-8.02 (m, 1 H), 7.97 (d, J = 5.7 Hz, 1 H), 7.88-7.80 (m, 2H), 7.06 (d, J = 1.7 Hz, 1 H), 6.96 (d, J = 5.7 Hz, 1H), 4.24 (br t, J = 5.7 Hz, 2H), 3.76 (br s, 6H), 2.68 (s, 3H), 2.33 (s, 3H).

Example 66: Ethyl 1-methyl-5-(2- ((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4,5,6,7-tetrahydro-1/7-pyrrolo[3,2-c]pyridin e-2-carboxylate

Example 66.1: To a mixture of DMF (11.4 g, 156 mmol, 12 mL) and dichloromethane (40 mL) was added phosphorus oxychloride (13.2 g, 86.3 mmol) at 0 °C slowly. The mixture was stirred at 0 °C for 15 min. To the mixture was added a solution of benzyl 4-oxopiperidine-1 -carboxylate (10.0 g, 42.9 mmol) in dichloromethane (40 mL) slowly. The resulting mixture was stirred at 25 °C for 2 h. After the reaction was completed, the reaction mixture was poured into a solution of sodium acetate (10.0 g) in ice water (100 mL). The reaction mixture was extracted with dichloromethane (3 x 40 mL), washed with water, dried over anhydrous sodium sulfate, concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 20/1 to 8/1) to afford benzyl 4-chloro-3-formyl-5,6-dihydropyridine-1(2/7)-carboxylate (5.40 g, 19.3 mmol, 45% yield) as a light-yellow oil. 1 H NMR (400 MHz, Chloroform-d): 5 10.14 (s, 1 H), 7.40-7.33 (m, 5H), 5.16 (s, 2H), 4.23 (br s, 2H), 3.70 (t, J = 5.6 Hz, 2H), 2.70 (br s, 2H).

Example 66.2: To a solution of benzyl 4-chloro-3-formyl-5,6-dihydropyridine-1(2/7)- carboxylate (5.40 g, 19.3 mmol) in dichloromethane (100 mL) was added ethyl 2- (triphenylphosphoranylidene)acetate (6.73 g, 19.3 mmol). The mixture was stirred at 25 °C for 12 h. After the reaction was completed, the mixture was concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 50/1 to 5/1) to afford benzyl 4-chloro-3-(3-ethoxy-3- oxoprop-1-en-1-yl)-5,6- dihydropyridine-1(2/7)-carboxylate (5.92 g, 16.9 mmol, 87% yield) as a yellow oil. HPLC/MS m/z: 350.1 [M+H] + , Rt (T): 1.02 min. 1 H NMR (400 MHz, Chloroform-d): 57.84 (d, J = 16.4 Hz, 1 H), 7.41-7.35 (m, 5H), 6.06-5.78 (m, 1 H), 5.19-5.15 (m, 2H), 4.25 (q, J = 7.2 Hz, 4H), 3.69 (t, J = 6.0 Hz, 2H), 2.76-2.53 (m, 2H), 1.32 (t, J = 7.2 Hz, 3H).

Example 66.3: A mixture of benzyl 4-chloro-3-(3-ethoxy-3-oxoprop-1-en-1-yl)-5,6- dihydropyridine-1 (2/-/)-carboxylate (3.90 g, 11.15 mmol) and sodium azide (1.96 g, 30.15 mmol, 2.70 eq) in DMF (40 mL) was stirred at 25 °C for 8 h. The mixture was poured into water (40 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organic layer was washed with brine (3 x 20 mL), dried over anhydrous sodium sulfate and concentrated to afford benzyl 4-azido-3-(3-ethoxy-3-oxoprop-1- en-1-yl)-5,6-dihydropyridine-1(2/-/)-carboxylate (4.6 g, crude) as a yellow oil. It was used for next step directly. HPLC/MS m/z: 379.1 [M+Na] + , Rt (T): 1.09 min.

Example 66.4: A solution of benzyl 4-azido-3-(3-ethoxy-3-oxoprop-1-en-1-yl)-5,6- dihydropyridine-1 (2/-/)-carboxylate (4.60 g, 12.9 mmol) in toluene (45 mL) was stirred at 60 °C for 8 h. After the reaction was completed, the mixture was concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 10/1 to 3/1) to afford 5-benzyl 2-ethyl 6,7- dihydro-1/7-pyrrolo[3,2-c]pyridine-2,5(4/-/)-dicarboxylate (700 mg, crude) as a yellow oil. 1 H NMR (400 MHz, Chloroform-d): 58.86 (br s, 1 H), 7.45-7.30 (m, 5H), 6.67 (s, 1 H), 5.17 (s, 2H), 4.50 (s, 2H), 4.30 (q, J = 7.2 Hz, 2H), 3.85-3.75 (m, 2H), 2.80-2.68 (m, 2H), 1.35 (t, J = 7.2 Hz, 3H).

Example 66.5: To a suspension of cesium carbonate (1.39 g, 4.26 mmol) in acetonitrile (10 mL) was added 5-benzyl 2-ethyl 6,7-dihydro-1/7-pyrrolo[3,2- c]pyridine-2,5(4/-/)-dicarboxylate (700 mg, 2.13 mmol) and iodomethane (1.56 g, 11.0 mmol, 5.16 eq). The mixture was stirred at 50 °C for 12 h. After the reaction was completed, the mixture was poured into water (5 mL). The mixture was extracted with ethyl acetate (3 x 5 mL), the combined organic layer was concentrated to afford 5-benzyl 2-ethyl 1-methyl-6,7-dihydro-1/7-pyrrolo[3,2- c]pyridine-2,5(4/-/)-dicarboxylate (690 mg, crude) as a yellow oil. HPLC/MS m/z: 343.2 [M+H] + , Rt (U): 1.12 min. 1 H NMR (400 MHz, Chloroform-d): 57.39-7.34 (m, 5H), 6.75 (br s, 1 H), 5.16 (s, 2H), 4.48 (s, 2H), 4.27 (q, J = 7.2 Hz, 2H), 3.86-3.77 (m, 5H), 2.67 (br s, 2H), 1 .34 (t, J = 7.2 Hz, 3H).

Example 66.6: A mixture of 5-benzyl 2-ethyl 1-methyl-6,7-dihydro-1/7-pyrrolo[3,2- c]pyridine-2,5(4/-/)-dicarboxylate (0.7 g, 2.04 mmol) and palladium on activated carbon (0.100 g, 5% purity) in methanol (20 mL) was stirred at 25 °C for 12 h under hydrogen atmosphere (15 Psi). After the reaction was completed, the mixture was filtered. The filtrate was concentrated to afford ethyl 1-methyl-4,5,6,7-tetrahydro-1/7- pyrrolo[3,2- c]pyridine-2-carboxylate (0.22 g, crude) as a colorless oil. 1 H NMR (400 MHz, Chloroform-d): 56.76 (s, 1 H), 4.30-4.25 (m, 2H), 4.11 (s, 2H), 3.80-3.78 (m, 3H), 3.44 (br t, J = 6.0 Hz, 2H), 2.94 (br t, J = 5.6 Hz, 2H), 1.34 (t, J = 7.2 Hz, 3H). Example 66.7: A mixture of ethyl 1-methyl-4,5,6,7-tetrahydro-1/7-pyrrolo[3,2- c]pyridine-2-carboxylate (200 mg, 960 umol), tert-butyl /\/-(2-bromoethyl)carbamate (258 mg, 1.15 mmol), sodium iodide (288 mg, 1.92 mmol) and cesium carbonate (626 mg, 1.92 mmol) in acetonitrile (0.5 mL) was stirred at 60 °C for 2 h. After the reaction was completed, the mixture was filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 8/1 to 3/1) to afford ethyl 5-(2-((terf- butoxycarbonyl)amino)ethyl)-1-methyl-4,5,6,7-tetrahydro-1/7- pyrrolo[3,2-c]pyridine- 2-carboxylate (260 mg, 739 umol, 77% yield) as a colorless oil. 1 H NMR (400 MHz, Chloroform-d): 56.76 (s, 1 H), 4.30-4.25 (m, 2H), 3.92 (s, 1 H), 3.81 (s, 3H), 3.78- 3.70 (m, 1 H), 3.68-3.57 (m, 2H), 3.50 (s, 1 H), 3.49-3.27 (m, 2H), 3.31-3.23 (m, 1 H), 3.16-3.11 (m, 1 H), 3.05-2.91 (m, 2H), 1.45 (s, 9H), 1.36-1.32 (m, 3H).

Example 66.8: A mixture of ethyl 5-(2-((tert-butoxycarbonyl)amino)ethyl)-1-methyl- 4,5,6,7-tetrahydro-1/7-pyrrolo[3,2-c] pyridine-2-carboxylate (50.0 mg, 142 umol) in hydrochloric acid/ethyl acetate (4 M, 0.5 mL) was stirred at 25 °C for 1 h. The mixture was concentrated to afford ethyl 5-(2-aminoethyl)-1-methyl-4, 5,6,7- tetrahydro-1/7-pyrrolo[3,2-c]pyridine-2-carboxylate (41.0 mg, crude, hydrochlorde) as a gray solid. HPLC/MS m/z: 252.5 [M+H] + , Rt (T): 0.22 min.

Example 66.9: A mixture of ethyl 5-(2-aminoethyl)-1-methyl-6,7-dihydro-4/7- pyrrolo[3,2-c]pyridine-2-carboxylate (41.0 mg, 142 umol, hydrochloride), diisopropylethylamine (73.6 mg, 570 umol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7- yl)-1 ,2,4-oxadiazole [Intermediate A5] (32.4 mg, 142 umol) and bromo(tripyrrolidin-1- yl)phosphonium;hexafluorophosphate (120 mg, 256 umol) in DMF (2.50 mL) was stirred at 25 °C for 12 h. After the reaction was completed, the pH of the mixture was adjusted to around 7 by adding hydrochloric acid (1 M). The mixture was purified by prep-HPLC(column: Phenomenex Gemini-NX C18 75*30mm*3um; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B%: 44%-74%, 11.5 min) and lyophilized to afford ethyl 1-methyl-5-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-4,5,6,7-tetrahydro-1/7-pyrr olo[3,2-c]pyridine-2- carboxylate (17.89 mg, 38.8 umol, 27% yield) as a yellow solid. HPLC/MS m/z: 461.2 [M+H] + , Rt (V): 1.03 min. 1 H NMR (400 MHz, DMSO-d 6 ): 58.86 (s, 1 H), 8.13 (dd, J = 1.2, 8.4 Hz, 1 H), 7.96 (d, J = 5.6 Hz, 1 H), 7.88-7.78 (m, 2H), 6.93 (d, J = 5.6 Hz, 1 H), 6.62 (s, 1 H), 4.16 (q, J = 7.2 Hz, 2H), 3.73-3.65 (m, 5H), 3.44 (s, 2H), 2.85- 2.75 (m, 4H), 2.70 (s, 3H), 2.67-2.61 (m, 2H), 1.24 (t, J = 7.2 Hz, 3H).

Example 67: Ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1-oxo-1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate

Example 67.1 : To a solution of 7-bromo-3,4-dihydroisoquinolin-1(2/7)-one (1.50 g, 6.64 mmol), triethylamine (1.34 g, 13.3 mmol) in a mixture solvent of ethanol (20 mL) and DMF (4 mL) were added palladium(ll) acetate (149 mg, 664 umol) and (5- diphenylphosphanyl-9,9-dimethyl-xanthen-4-yl)-diphenyl-phosp hane (1.15 g, 1.99 mmol). The mixture was stirred at 70 °C for 24 h under carbon monoxide atmosphere (45 Psi). The mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 10/1 to 0/1) to afford ethyl 1-oxo-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (0.900 g, 4.11 mmol, 61 % yield) as a yellow solid. HPLC/MS m/z: 220.0 [M+H] + , Rt (V): 0.81 min. 1 H NMR (400 MHz, DMSO-d 6 ): 68.41 (d, J = 1.6 Hz, 1 H), 8.11 (br s, 1 H), 8.03 (dd, J = 1.7, 7.8 Hz, 1 H), 7.47 (d, J = 7.9 Hz, 1 H), 4.33 (q, J = 7.1 Hz, 2H), 3.39 (td, J = 3.2, 6.5 Hz, 2H), 2.99 (t, J = 6.5 Hz, 2H), 1.33 (t, J = 7.1 Hz, 3H).

Example 67.2: To a solution of ethyl 1-oxo-1 ,2,3,4-tetrahydroisoquinoline-7- carboxylate (0.800 g, 3.65 mmol) in tetrahydrofuran (10 mL) was added sodium hydride (204 mg, 5.11 mmol, 60% purity) at 0 °C. The mixture was stirred at 0 °C for 0.5 h. Then to the mixture was added 2-bromoacetonitrile (1 .31 g, 11.0 mmol) was added. The mixture was stirred at 20 °C for 12 h. The mixture was diluted with saturated ammonium chloride aqueous solution (20 mL) at 0 °C and extracted with ethyl acetate (3 x 20 mL). The combined organic layer was washed with brine (3 x 20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 10/1 to 2/1) to afford ethyl 2- (cyanomethyl)-1-oxo-1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate (0.600 g, 2.32 mmol, 63% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ): 68.46 (d, J = 1.5 Hz, 1H), 8.09 (dd, J = 1.7, 7.9 Hz, 1 H), 7.51 (d, J = 8.1 Hz, 1H), 4.63 (s, 2H), 4.34 (q, J = 7.0 Hz, 2H), 3.71 (t, J = 6.6 Hz, 2H), 3.18-3.10 (m, 2H), 1.34 (t, J = 7.1 Hz, 3H).

Example 67.3: To a solution of ethyl 2-(cyanomethyl)-1-oxo-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (600 mg, 2.32 mmol) in a mixture solvent of methanol (20 mL) and ammonium hydroxide (4 mL) was added Raney-nickel (60.0 mg, 10% purity). The mixture was stirred at 20 °C for 12 h under hydrogen atmosphere (45 Psi). The mixture was filtered and concentrated to give a residue. The residue was purified by reverse phase column chromatography (C18, 40 g; condition: water/acetonitrile = 1/0 to 0/1 , 0.1% formic acid) and prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um; mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 8%-38%, 8 min) to afford ethyl 2-(2-aminoethyl)-1-oxo- 1 ,2,3,4- tetrahydroisoquinoline-7-carboxylate (0.250 g, 953 umol, 41% yield) as a yellow oil. 1 H NMR (400 MHz, Methanol-d 4 ) 58.64 (d, J = 1.6 Hz, 1H), 8.15 (dd, J = 2.0, 7.9 Hz, 1 H), 7.46 (d, J = 8.1 Hz, 1 H), 4.41 (q, J = 7.2 Hz, 2H), 3.87 (t, J = 5.9 Hz, 2H), 3.73 (t, J = 6.7 Hz, 2H), 3.26 (t, J = 5.9 Hz, 2H), 3.18 (t, J = 6.6 Hz, 2H), 1.42 (t, J = 7.2 Hz, 3H).

Example 67.4: To a solution of ethyl 2-(2-aminoethyl)-1-oxo-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (60.0 mg, 201 umol, hydrochloride), N,N- diisopropylethylamine (123 mg, 953 umol) in dichloromethane (5 mL) were added 5- methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1,2,4-oxadiazole [Intermediate A5] (43.3 mg, 191 umol) and bromotripyrrolidinophosphonium hexafluorophosphate (178 mg, 381 umol). The mixture was stirred at 25 °C for 12 h. The mixture was concentrated under reduced pressure. The residue was purified by prep-TLC (petroleum ether/ethyl acetate = 1/1), prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um; mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 36%-66%, 8 min) and lyophilized to afford ethyl 2-(2-((7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-1-oxo-1,2,3,4-tetrahydroiso quinoline-7-carboxylate (10.94 mg, 22.97 umol, 12% yield, 99% purity) as a yellow solid. HPLC/MS m/z: 472.2 [M+H] + , Rt (V): 1.01 min. 1 H NMR (400 MHz, DMSO-d 6 ): 58.87 (s, 1 H), 8.40 (d, J = 1.7 Hz, 1H), 8.15 (dd, J = 1.4, 8.5 Hz, 1H), 8.06 (br t, J = 5.1 Hz, 1H), 8.01 (dd, J = 1.9, 7.9 Hz, 1 H), 7.96 (d, J = 5.7 Hz, 1H), 7.85 (d, J = 8.4 Hz, 1 H), 7.43 (d, J = 7.8 Hz, 1H), 6.95 (d, J = 5.7 Hz, 1 H), 4.32 (q, J = 7.1 Hz, 2H), 3.83-3.73 (m, 4H), 3.62 (t, J = 6.5 Hz, 2H), 3.01 (t, J = 6.5 Hz, 2H), 2.68 (s, 3H), 1.32 (t, J = 7.1 Hz, 3H). Example 68: Propan-2-yl 2-(2-((7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1,2,3,4-tetrahydropyrrolo[1,2-a]pyrazine-7-c arboxylate

Example 68.1: To a solution of ethyl 2-oxopropanoate (20.0 g, 172 mmol) in acetonitrile (150 mL) was added ethane-1,2-diamine (10.4 g, 172 mmol). The mixture was stirred at 20 °C for 0.5 h. To the mixture was added a solution of ethyl 3-bromo-2-oxo-propanoate (33.6 g, 172 mmol) in acetonitrile (60 mL) and iron(lll) chloride (5.59 g, 34.5 mmol) at 0 °C. The mixture was stirred at 80 °C for 12 h. After cooling to room temperature, the mixture was diluted with saturated ammonium chloride aqueous solution (150 mL) and extracted with ethyl acetate (3 x 150 mL). The combined organic phase was washed with brine (3 x 100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 5/1 to ethyl acetate/methanol = 10/1) to afford ethyl 1-oxo- 1 ,2,3,4-tetrahydropyrrolo[1,2-a]pyrazine-7-carboxylate (4.00 g, 19.2 mmol, 11% yield) as a brown solid. 1 H NMR (400 MHz, DMSO-d 6 ): 67.92 (br s, 1H), 7.64 (s, 1 H), 6.91 (s, 1 H), 4.25-4.18 (m, 2H), 4.18-4.12 (m, 2H), 3.51 (br s, 2H), 1.26 (br t, J = 7.0 Hz, 3H).

Example 68.2: To a solution of ethyl 1-oxo-1 ,2,3,4-tetrahydropyrrolo[1,2-a]pyrazine- 7-carboxylate (4.00 g, 19.2 mmol) and borontrifluoridediethyletherate (46.0 g, 324 mmol, 40.0 mL) in tetrahydrofuran (300 mL) was added sodium borohydride (8.00 g, 211 mmol) in portions at 0 °C. The mixture was stirred at 20 °C for 24 h. The mixture was quenched with ethyl alcohol (40 mL) and stirred for 12 h. The mixture was concentrated to give a residue. The residue was diluted with hydrochloric acid aqueous solution (10% 30 mL) and the solution was stirred for 2 h. The resulting solution was neutralized with saturated sodium bicarbonate aqueous solution and the pH of the solution was 8. The mixture was diluted with ethyl acetate (200 mL). After separation, the aqueous layer was extracted with ethyl acetate (2 x 200 mL). The combined organic layers were concentrated to give a residue. The residue was purified by reverse phase column chromatography (C18, 40 g; condition: water/acetonitrile = 1/0 to 0/1 , 0.1% trifluoroacetic acid) to afford ethyl 1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (1.00 g, 3.24 mmol, 16% yield, trifluoroacetate) as a yellow solid. 1 H NMR (400 MHz, DMSO-de): 69.36 (br s, 2H), 7.48 (d, J = 1.6 Hz, 1 H), 6.36 (d, J = 0.8 Hz, 1 H), 4.31 (s, 2H), 4.24-4.18 (m, 2H), 4.18-4.12 (m, 2H), 3.56 (br t, J = 5.8 Hz, 2H), 1.23 (t, J = 7.1 Hz, 3H).

Example 68.3: To a suspension of ethyl 1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7- carboxylate (0.400 g, 1.30 mmol, trifluoroacetate), tert-butyl /\/-(2- bromoethyl)carbamate (1.15 g, 5.13 mmol) in DMF (5 mL) was added N,N- diisopropylethylamine (798 mg, 6.18 mmol) and potassiumiodide (34.2 mg, 206 umol). The mixture was stirred at 90 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 10/1 to 0/1) to afford ethyl 2-(2-((tert- butoxycarbonyl)amino)ethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (0.350 g, 1.04 mmol, 80% yield) as yellow oil. HPLC/MS m/z: 338.1 [M+H] + , Rt (V): 0.92 min. 1 H NMR (400 MHz, DMSO-d 6 ): 67.28 (d, J = 1.6 Hz, 1 H), 6.73 (br t, J = 5.4 Hz, 1 H), 6.10 (s, 1 H), 4.13 (q, J = 7.1 Hz, 2H), 3.94 (br t, J = 5.3 Hz, 2H), 3.54 (s, 2H), 3.13-3.05 (m, 2H), 2.77 (br t, J = 5.4 Hz, 2H), 2.47 (br s, 2H), 1.24-1.20 (m, 3H).

Example 68.4: A solution of ethyl 2-(2-((terf-butoxycarbonyl)amino)ethyl)-1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (0.350 g, 1.04 mmol) in hydrochloric acid/ethyl acetate (4 M, 5 mL) was stirred at 20 °C for 2 h. The mixture was concentrated under reduced pressure to afford ethyl 2-(2-aminoethyl)-1 , 2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (0.25 g, crude, hydrochloride) as a yellow solid. 1 H NMR (400 MHz, Methanol-d 4 ): 57.43 (d, J = 1.6 Hz, 1 H), 6.46 (s, 1 H), 4.44 (s, 2H), 4.38 (t, J = 5.8 Hz, 2H), 4.24 (q, J = 7.1 Hz, 2H), 3.74-3.69 (m, 2H), 3.56-3.42 (m, 4H), 1.31 (t, J = 7.2 Hz, 3H).

Example 68.5: To a solution of ethyl 2-(2-aminoethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2- a]pyrazine-7-carboxylate (0.150 g, 548 umol, hydrochloride), 5-methyl-3-(2- oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole [Intermediate A5] (125 mg, 548 umol) and /V,/V-diisopropylethylamine (354 mg, 2.74 mmol) in dichloromethane (2 mL) was added bromotripyrrolidinophosphonium hexafluorophosphate (511 mg, 1.10 mmol). The mixture was stirred at 35 °C for 12 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (ethyl acetate) and reverse phase column chromatography (C18, 40 g; condition: water/acetonitrile = 1/0 to 0/1 , 0.1% ammonium hydroxide) to afford ethyl 2-(2-((7- (5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-1 ,2,3,4- tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate (50.0 mg, 112 umol, 20% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ): 68.87 (s, 1 H), 8.14 (dd, J = 1.5, 8.4 Hz, 1 H), 7.96 (d, J = 5.7 Hz, 1 H), 7.90-7.78 (m, 2H), 7.29 (d, J = 1.7 Hz, 1 H), 6.94 (d, J = 5.6 Hz, 1 H), 6.12 (d, J = 1.5 Hz, 1 H), 4.13 (q, J = 7.1 Hz, 2H), 3.97 (t, J = 5.5 Hz, 2H), 3.70 (q, J = 6.2 Hz, 2H), 3.65 (s, 2H), 2.88 (t, J = 5.5 Hz, 2H), 2.79 (t, J = 6.7 Hz, 2H), 2.70 (s, 3H), 1.22 (t, J = 7.1 Hz, 3H).

Example 68.6: To a solution of ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7- carboxylate (45.0 mg, 101 umol) in a mixture solvent of tetrahydrofuran (2 mL) and methanol (2 mL) was added a solution of lithium hydroxide monohydrate (12.7 mg, 302 umol) in water (1 mL). The mixture was stirred at 70 °C for 12 h. Then the mixture was concentrated under reduced pressure to afford 2-(2-((7-(5-methyl-1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine- 7-carboxylic acid (50 mg, crude, lithium salt) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ): 68.88 (s, 1 H), 8.15-8.13 (m, 1 H), 7.97 (d, J = 5.6 Hz, 1 H), 7.87-7.82 (m, 1 H), 6.94 (d, J = 5.7 Hz, 1 H), 6.69 (s, 1 H), 5.84 (s, 1 H), 3.89-3.83 (m, 2H), 3.71 (m, 2H), 3.02 (dt, J = 3.7, 6.4 Hz, 2H), 2.82 (br d, J = 4.4 Hz, 2H), 2.77 (br d, J = 6.5 Hz, 2H), 2.70 (s, 3H).

Example 68.7: To a solution of 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid (50 mg, crude, lithium salt) in DMF (2.5 mL) was added a solution of 2-bromopropane (18.8 mg, 153 umol) in DMF (0.5 mL). The mixture was stirred at 50 °C for 4 h. Then the mixture was filtered. The filtrate was purified by prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um; mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 40%-70%, 8 min) and lyophilized to afford propan-2-yl 2-(2-((7-(5-methyl-1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine- 7-carboxylate (15.64 mg, 33.6 umol, 28% yield, 99% purity) as a yellow solid. HPLC/MS m/z: 461.2 [M+H] + , Rt (V): 1.03 min. 1 H NMR (400 MHz, DMSO-d 6 ): 6 8.87 (s, 1 H), 8.14 (dd, J = 1.5, 8.6 Hz, 1 H), 7.96 (d, J = 5.7 Hz, 1 H), 7.89-7.85 (m, 1 H), 7.84 (d, J = 8.6 Hz, 1 H), 7.25 (d, J = 1.7 Hz, 1 H), 6.94 (d, J = 5.6 Hz, 1 H), 6.10 (d, J = 1.6 Hz, 1 H), 4.98 (quin, J = 6.2 Hz, 1 H), 3.97 (t, J = 5.4 Hz, 2H), 3.70 (q, J = 6.3 Hz, 2H), 3.65 (s, 2H), 2.88 (br t, J = 5.5 Hz, 2H), 2.78 (t, J = 6.7 Hz, 2H), 2.70 (s, 3H), 1.22 (d, J = 6.2 Hz, 6H). Example 69: Ethyl 6-[2-[[3-(5-methyl-1,2,4-oxadiazol-3-yl)benzoyl]amino]ethyl] -5,7- dihydro-4H-thieno[2,3-c]pyridine-2-carboxylate

Example 69.1 : 6-(tert-Butoxycarbonyl)-4,5,6,7-tetrahydrothieno[2,3-c]pyrid ine-2- carboxylic acid (100.00 mg, 0.3529 mmol), 4-dimethylaminopyridine (12.94 mg, 0.1059 mmol) and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (101.49 mg, 0.5294 mmol) were dissolved in a mixture of DMF (1.01 mL) and EtOH (1.00 mL). The reaction mixture was stirred at 70 °C for 45 min. The mixture was cooled down to room temperature, water and EtOAc were added, the product was extracted with EtOAc, dried over MgSO4 and concentrated in vacuo on to silica. The product was purified using Isco Combi-flash purification system, eluent cyclohexane/EtOAc 20% to 80% over 8 min to afford 06-tert-butyl 02-ethyl 5,7- dihydro-4H-thieno[2,3-c]pyridine-2,6-dicarboxylate (80 mg, 73%, 0.2569 mmol) as a colorless oil. HPLC/MS m/z: 212.07 [M+H-Boc] + , Rt (AE): 1.54 min.

Example 69.2: 06-tert-butyl 02-ethyl 5,7-dihydro-4H-thieno[2,3-c]pyridine-2,6- dicarboxylate (70.00 mg, 0.2248 mmol) was dissolved in dry 1,4-dioxane (1.12 mL). Hydrogen chloride (4 M in 1,4-dioxane) (0.56 mL, 2.2479 mmol) was added, the reaction mixture was stirred at room temperature for 18 h. Additional hydrogen chloride (4 M in 1 ,4-dioxane) (0.56 mL, 2.2479 mmol) was added, stirred for another 6 h. The solvent was removed in vacuo to give ethyl 4,5,6,7-tetrahydrothieno[2,3- c]pyridine-2-carboxylate hydrochloride (48 mg, 86%, 0.1938 mmol) as a colorless powder. HPLC/MS m/z: 212.07 [M+H] + , Rt (AE): 0.69 min.

Example 69.3: Ethyl 4,5,6,7-tetrahydrothieno[2,3-c]pyridine-2-carboxylate hydrochloride (48.00 mg, 0.1938 mmol) and N-Boc-2-chloroethylamine (88.79 mg, 0.4844 mmol) were dissolved in dry DMF (0.97 mL). Triethylamine (81.83 uL, 0.5813 mmol) was added and the reaction mixture was stirred at 70 °C for 18 h. Water and EtOAc were added. The product was extracted with EtOAc, dried over MgSO4 and concentrated in vacuo. Purification by silica gel column chromatography (eluent: 20-60% EtOAc in cyclohexane) to yield ethyl 6-[2-(tert- butoxycarbonylamino)ethyl]-5,7-dihydro-4H-thieno[2,3-c]pyrid ine-2-carboxylate (26 mg, 38%, 0.0734 mmol) as an orange oil. HPLC/MS m/z: 355.107 [M+H] + , Rt (AE): 1.03 min.

Example 69.4: Ethyl 6-[2-(tert-butoxycarbonylamino)ethyl]-5,7-dihydro-4H- thieno[2,3-c]pyridine-2-carboxylate (26.00 mg, 0.0734 mmol) was dissolved in dry 1 ,4-dioxane (0.37 mL). Hydrogen chloride (4 M in dioxane) (0.18 mL, 0.7335 mmol) was added, and the reaction mixture was stirred at room temperature for 2 h. The solvent was removed in vacuo to give ethyl 6-(2-aminoethyl)-5,7-dihydro-4H- thieno[2,3-c]pyridine-2-carboxylate hydrochloride (21 mg, 98%, 0.0722 mmol) as a pale-red powder. Ethyl 6-(2-aminoethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-2- carboxylate hydrochloride (21.00 mg, 0.0722 mmol) and 3-(5-Methyl-1 ,2,4- oxadiazol-3-yl)benzoic acid (22.12 mg, 0.1083 mmol) were dissolved in dry DMF (0.36 mL). 1-Propanephosphonic anhydride (63.82 uL, 0.1083 mmol) and triethylamine (15.25 uL, 0.1083 mmol) were added and the reaction mixture was stirred at room temperature for 18 h. 1-Propanephosphonic anhydride (63.82 uL, 0.1083 mmol) and 3-(5-Methyl-1 ,2,4-oxadiazol-3-yl)benzoic acid (22.12 mg, 0.1083 mmol) were added, stirred at room temperature for 3 h. Purification by reverse flash chromatography (eluent: 30-80% MeOH/water + 0.1% formic acid) followed by ion exchange SCX-2 chromatography eluting with 2 M of NH3 in MeOH to afford ethyl 6-[2-[[3-(5-methyl-1 ,2,4-oxadiazol-3-yl)benzoyl]amino]ethyl]-5,7-dihydro-4H- thieno[2,3-c]pyridine-2-carboxylate (3 mg, 9%, 0.0066 mmol) as an off-white solid. HPLC/MS m/z: 441.16 [M+H] + , Rt (AF): 2.13 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.52 (t, J = 1.8 Hz, 1 H), 8.19 (dt, J = 7.8, 1.4 Hz, 1 H), 8.01-7.96 (m, 1 H), 7.61 (t, J = 7.8 Hz, 1 H), 7.51 (s, 1 H), 4.30 (q, J = 7.1 Hz, 2H), 3.87-3.82 (m, 2H), 3.66 (t, J = 6.6 Hz, 2H), 2.92 (t, J = 5.9 Hz, 2H), 2.86 (t, J = 6.7 Hz, 2H), 2.82-2.78 (m, 2H), 2.66 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

The following example was obtained in an analogous manner.

Example 70: Propan-2-yl 6-[2-[[3-(5-methyl-1 ,2,4-oxadiazol-3- yl)benzoyl]amino]ethyl]-5,7-dihydro-4H-thieno[2,3-c]pyridine -2-carboxylate

Colorless powder (10 mg, 37%, 0.0220 mmol). HPLC/MS m/z: 455.17 [M+H] + , Rt (AF): 2.26 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.52 (t, J = 1.8 Hz, 1 H), 8.19 (dt, J = 7.8, 1.4 Hz, 1 H), 8.01-7.96 (m, 1 H), 7.61 (t, J = 7.8 Hz, 1 H), 7.49 (s, 1 H), 5.13 (hept, J = 6.3 Hz, 1 H), 3.86-3.82 (m, 2H), 3.65 (t, J = 6.6 Hz, 2H), 2.92 (t, J = 5.9 Hz, 2H), 2.86 (t, J = 6.6 Hz, 2H), 2.80 (t, J = 5.9 Hz, 2H), 2.66 (s, 3H), 1 .33 (d, J = 6.3 Hz, 6H).

Example 71 : tert-Butyl 6-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-5,7-dihydro-4H-thieno[2,3-c]pyridin e-2-carboxylate

Example 71.1 : 6-tert-Butoxycarbonyl-5,7-dihydro-4H-thieno[2,3-c]pyridine-2 - carboxylic acid (100.00 mg, 0.3529 mmol) was dissolved in dry THF (1.47 mL) and cooled to 0 °C. tert-Butyl-2,2,2-trichloroacetamidate (231.36 mg, 1.0588 mmol) in THF (0.2 mL) was added followed by dropwise addition of boron trifluoride diethyl etherate (0.11 mL, 0.8823 mmol). The reaction was warmed to room temperature for 1 h. NaHCOs and EtOAc were added, the organic layer was washed with water, dried over MgSCL . Purification by normal phase silica column chromatography ((0- 35% EtOAc in cyclohexane) afforded di-tert-butyl 5,7-dihydro-4H-thieno[2,3- c]pyridine-2,6-dicarboxylate (112 mg, 93%, 0.3299 mmol) as a yellow oil. HPLC/MS m/z: 362.14 [M+Na] + , Rt (AE): 1.72 min.

Example 71.2: tert-Butyl 6-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-5,7-dihydro-4H-thieno[2,3-c]pyridin e-2-carboxylate was prepared from ditert-butyl 5,7-dihydro-4H-thieno[2,3-c]pyridine-2,6-dicarboxylate in an analogous manner shown in Example 69. Example 71.3: tert-Butyl 6-(2-aminoethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-2- carboxylate (32.81 mg, 0.1162 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)- 1 ,2,4-oxadiazole [Intermediate A5] (22.00 mg, 0.0968 mmol) and PyBroP (55.07 mg, 0.1181 mmol) were dissolved in dry DCM (0.48 mL). DIPEA (80.11 uL, 0.4599 mmol) was added and the reaction mixture was stirred at room temperature for 2 d. Purified by reverse phase column chromatography (eluent: 40-100% MeOH/water + 0.1% formic acid) followed by SCX-2 ion exchange chromatography eluting with ammonia in MeOH 2M to yield tert-butyl 6-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-5,7-dihydro-4H-thieno[2,3-c]pyridin e-2-carboxylate (10 mg, 21%, 0.0203 mmol) as a colorless power. HPLC/MS m/z: 492.21 [M+H] + , Rt (AF): 2.81 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.83-8.79 (m, 1H), 8.18 (dd, J = 8.5, 1.6 Hz, 1 H), 7.90 (d, J = 5.9 Hz, 1H), 7.77 (d, J = 8.5 Hz, 1 H), 7.42 (s, 1 H), 6.95 (dd, J = 6.0, 0.9 Hz, 1H), 3.88-3.83 (m, 2H), 3.80 (t, J = 6.4 Hz, 2H), 2.97 (t, J = 6.4 Hz, 2H), 2.93 (t, J = 5.8 Hz, 2H), 2.82-2.78 (m, 2H), 2.66 (s, 3H), 1.55 (s, 9H).

Example 72: Ethyl 5-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino ]ethyl]- 6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2-carboxylate

Example 72.1: 4,5,6,7-Tetrahydropyrazolo[1 ,5-a]pyrazine-2-carboxylic acid hydrochloride (600.00 mg, 2.9465 mmol) was charged in a flask. Di-tert-butyl dicarbonate (662.97 mg, 2.9465 mmol) in MeCN (11.00 mL) was added followed by triethylamine (0.41 mL, 2.9465 mmol). The reaction mixture was stirred at 40 °C for 40 min. This crude 5-tert-butoxycarbonyl-6,7-dihydro-4H-pyrazolo[1,5-a]pyrazine -2- carboxylic acid (1337 mg) was used without purification in the next step. 5-tert- Butoxycarbonyl-6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2-carboxylic acid (700.00 mg, 1.7287 mmol), 4-dimethylaminopyridine (42.24 mg, 0.3457 mmol) and 1-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (497.09 mg, 2.593 mmol) were dissolved in a mixture of DMF (2.88 mL) and Ethanol (2.88 mL). The reaction mixture was stirred at 50 °C for 4 h. The reaction mixture was concentrated in vacuo. EtOAc and aqueous NH4CI were added, the organic layer washed again with aqueous NH4CI, dried over MgSC>4 and concentrated in vacuo to give 05-tert-butyl 02-ethyl 6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2,5-dicarboxylate (400 mg, 78%, 1.3544 mmol) as a colourless oil. 1 H NMR (500 MHz, Chloroform-d): 5 6.63 (t, J = 0.9 Hz, 1 H), 4.68 (s, 2H), 4.41 (q, J = 7.1 Hz, 2H), 4.27 (t, J = 5.5 Hz, 2H), 3.91 (t, J = 5.5 Hz, 2H), 1.51 (s, 9H), 1.40 (t, J = 7.1 Hz, 3H).

Example 72.2: 05-tert-butyl 02-ethyl 6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2,5- dicarboxylate (400.00 mg, 1.3544 mmol) was dissolved in dry 1 ,4-dioxane (4.51 mL). Hydrogen chloride (4 M in 1 ,4-dioxane) (1.69 mL, 6.7721 mmol) was added, the reaction mixture was stirred at room temperature for 4.5 h. Additional hydrogen chloride (4 M in 1 ,4-dioxane) (1.69 mL, 6.7721 mmol) was added and stirred for 16 h. The solvent was removed in vacuo to give crude ethyl 4, 5,6,7- tetrahydropyrazolo[1 ,5-a]pyrazine-2-carboxylate hydrochloride (360 mg, 115%, 1.5539 mmol) as a colorless powder. HPLC/MS m/z: 196.11 [M+H] + , Rt (Al): 0.27 min.

Example 72.3: Ethyl 4,5,6,7-tetrahydropyrazolo[1 ,5-a]pyrazine-2-carboxylate hydrochloride (150.00 mg, 0.6474 mmol) was dissolved in CHCI3 (3.24 mL). Triethylamine (0.09 mL, 0.6474 mmol) was added, and the reaction mixture was cooled to 0 °C. 1-Nitroethene (1 M in xylene) (0.65 mL, 0.6474 mmol) was added, stirred at 0 °C for 50 min. The mixture was concentrated in vacuo. Purification by column chromatography (eluent: 0-10% MeOH in EtOAc) afforded ethyl 5-(2- nitroethyl)-6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2-carboxylate (117 mg, 67%, 0.4361 mmol) as a colorless oil that solidified on standing. 1 H NMR (500 MHz, DMSO-d 6 ): 5 6.51 (s, 1 H), 4.83-4.75 (m, 2H), 4.24 (q, J = 7.1 Hz, 2H), 4.11 (t, J = 5.6 Hz, 2H), 3.77 (s, 2H), 3.15-3.09 (m, 2H), 3.05-3.00 (m, 2H), 1.26 (t, J = 7.1 Hz, 3H).

Example 72.4: Ethyl 5-(2-nitroethyl)-6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2- carboxylate (90.00 mg, 0.3355 mmol) was dissolved in ethanol (1.68 mL). Palladium on carbon (35.70 mg) was added, the atmosphere was replaced by hydrogen. The reaction mixture was stirred at room temperature for 22 h. The reaction mixture was filtered through celite, rinsed with EtOH. The solvent was removed under reduced pressure. Purification by reverse phase flash chromatography (eluent: 10-60% MeOH/water + 0.1% formic acid) followed by ion exchange SCX-2 chromatography eluting with 2M NH3 in MeOH afforded ethyl 5-(2-aminoethyl)-6,7-dihydro-4H- pyrazolo[1 ,5-a]pyrazine-2-carboxylate (18 mg, 23%). HPLC/MS m/z: 239.15 [M+H] + , Rt (AI): 0.61 min. Example 72.5: Ethyl 5-(2-aminoethyl)-6,7-dihydro-4H-pyrazolo[1,5-a]pyrazine-2- carboxylate (12.00 mg, 0.0504 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)- 1 ,2,4-oxadiazole [Intermediate A5] (22.89 mg, 0.1007 mmol) and Bromotri(pyrrolidino)phosphonium hexafluorophosphate (35.21 mg, 0.0755 mmol) were dissolved in dry DCM (0.50 mL) in a 0.5-2 mL microwave vial. DIPEA (40.35 uL, 0.2317 mmol) was added and the reaction mixture was irradiated at 60 °C for 1 h. The solvent was removed in vacuo. Purification by reverse phase flash chromatography (eluent: 5-60% MeCN/water + 0.1% formic acid) followed by ion exchange SCX-2 chromatography eluting with 2M NH3 in MeOH afforded ethyl 5-[2- [[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethy l]-6,7-dihydro-4H- pyrazolo[1,5-a]pyrazine-2-carboxylate (6 mg, 25%, 0.0127 mmol) as a colorless powder. HPLC/MS m/z: 448.21 [M+H] + , Rt (AD): 2.12 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.81-8.77 (m, 1H), 8.19 (dd, J = 8.5, 1.6 Hz, 1 H), 7.91 (d, J = 5.9 Hz, 1 H), 7.78 (d, J = 8.5 Hz, 1 H), 6.96 (dd, J = 6.0, 0.9 Hz, 1 H), 6.56 (s, 1 H), 4.33 (q, J = 7.1 Hz, 2H), 4.25 (t, J = 5.6 Hz, 2H), 3.88 (s, 2H), 3.80 (t, J = 6.3 Hz, 2H), 3.18-3.12 (m, 2H), 2.98 (t, J = 6.3 Hz, 2H), 2.66 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

Example 73: Propan-2-yl 5-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2-carboxylate

Example 73.1: Propan-2-yl 5-(2-aminoethyl)-6,7-dihydro-4H-pyrazolo[1,5- a]pyrazine-2-carboxylate was prepared by an analogous procedure to Example 72. Example 73.2: Propan-2-yl 5-(2-nitroethyl)-6,7-dihydro-4H-pyrazolo[1,5-a]pyrazine- 2-carboxylate (110.00 mg, 0.3897 mmol), zinc (127.38 mg, 1.9483 mmol) and ammonium chloride (104.21 mg, 1.9483 mmol) were suspended in a mixture of THF (1.50 mL) / water (0.50 mL) / MeOH (0.50 mL). The reaction mixture was stirred at room temperature for 2 h. Additional zinc (127.38 mg, 1.9483 mmol) and ammonium chloride (104.21 mg, 1.9483 mmol) were added, stirred for another 1 h. The mixture was filtered on celite. The filtrate was passed through a 5 g NH2 ion exchange column and rinsed with EtOAc, then a 1:1 mixture of DCM:MeOH. The filtrate was concentrated to give propan-2-yl 5-(2-aminoethyl)-6,7-dihydro-4H-pyrazolo[1 ,5- a]pyrazine-2-carboxylate (60 mg, 61%, 0.2378 mmol) as a colorless oil. HPLC/MS m/z: 253.17 [M+H] + , Rt (Al): 0.74 min.

Example 73.3: Propan-2-yl 5-(2-aminoethyl)-6,7-dihydro-4H-pyrazolo[1 ,5- a]pyrazine-2-carboxylate (30.00 mg, 0.1189 mmol) used in an analogous procedure to Example 72.5 to afford propan-2-yl 5-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-6,7-dihydro-4H-pyrazolo[1 ,5-a]pyrazine-2-carboxylate (5 mg, 9%, 0.0103 mmol) as a colorless powder. HPLC/MS m/z: 462.23 [M+H] + , Rt (AJ): 2.03 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.81-8.78 (m, 1 H), 8.20 (dd, J = 8.5, 1.6 Hz, 1 H), 7.91 (d, J = 5.9 Hz, 1 H), 7.79 (d, J = 8.5 Hz, 1 H), 6.97 (dd, J = 5.9, 0.9 Hz, 1 H), 6.57-6.53 (m, 1 H), 5.19 (hept, J = 6.3 Hz, 1 H), 4.26 (t, J = 5.6 Hz, 2H), 3.88 (s, 2H), 3.80 (t, J = 6.3 Hz, 2H), 3.15 (t, J = 5.6 Hz, 2H), 2.99 (t, J = 6.3 Hz, 2H), 2.66 (s, 3H), 1 .34 (d, J = 6.3 Hz, 6H).

Example 74: 6-(5,5-Dimethyl-4H-oxazol-2-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)-1-isoquinolyl]amino]ethyl]isoindolin-1-one

2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]am ino]ethyl]-3-oxo-isoindoline- 5-carbonitrile [Example 59.2] (42.00 mg, 0.1023 mmol) and zinc chloride (2.79 mg, 0.0205 mmol) were mixed in a microwave vial. The capped vial was evacuated for 10 min. The reaction vessel then was flushed with argon and anhydrous toluene (0.10 mL) and 1-amino-2-methylpropan-2-ol (0.01 mL, 0.1228 mmol) were added. The reaction mixture was heated at 110 °C overnight. More toluene (1 mL) and 1- amino-2-methylpropan-2-ol (10 eq.) were added and the reaction mixture was heated at 150 °C for 10 h by microwave irradiation. The crude was purified by prep-HPLC (Teledyne AccqPrep, 67-74% MeOH in water, pH10, 20 min). Further purification by column chromatography (eluent: 0-15% EtOAc in MeOH) afforded 6- (5,5-dimethyl-4H-oxazol-2-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]isoindolin-1-one (14 mg, 28%, 0.0283 mmol) as a pink solid. HPLC/MS m/z: 483.214 [M+H] + , Rt (AJ): 1.92 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.82 (d, J = 1.5 Hz, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.05-8.01 (m, 2H), 7.99 (d, J = 1.4 Hz, 1H), 7.94 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1H), 7.71-7.66 (m, 1 H), 6.96-6.92 (m, 1H), 4.65 (s, 2H), 3.88-3.84 (m, 2H), 3.82-3.78 (m, 2H), 3.71 (s, 2H), 2.67 (s, 3H), 1.43 (s, 6H).

Example 75: 2-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-6-(5- propyl-1 ,2,4-oxadiazol-3-yl)isoindolin-1-one

Example 75.1: 2-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-3- oxo-isoindoline-5-carbonitrile [Example 59.2] (129.00 mg, 0.3143 mmol), hydroxylamine hydrochloride (24.03 mg, 0.3457 mmol) and triethylamine (0.07 mL, 0.4715 mmol) were mixed in [bmim]OAc (0.63 mL) under argon. The reaction mixture was heated at 80 °C for 1 h. The reaction mixture was cooled to ambient temperature, mixed with EtOAc (30 mL). The organic layer was mixed with saturated NaCI (50 mL) and separated again. The aqueous layer was extracted further with EtOAc (2 x 30 mL). The combined organic layer was washed with saturated NaCI (3 x 20 mL) and subsequently concentrated under reduced pressure without filtering through a pad of anhydrous MgSO4 to retain the undissolved product. The crude N- hydroxy-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-3-oxo- isoindoline-5-carboxamidine (88 mg, 63%, 0.1984 mmol) was used for the next step without further purification. HPLC/MS m/z: 444.175 [M+H]+, Rt (AD): 1.25 min. Example 75.2: N-Hydroxy-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-3-oxo-isoindoline-5-carboxamidine (40.00 mg, 0.0902 mmol), butyric anhydride (0.03 mL, 0.1804 mmol) and anhydrous MeCN (1.00 mL) were mixed in a microwave vial under argon. The reaction mixture was heated at 160 °C for 10 min by microwave irradiation. Volatiles were removed under reduced pressure. The crude was directly purified by prep-HPLC (AccqPrep, 30-43% MeOH in water, pH3, 26 min). Purification by ion exchange SCX-2 chromatography eluting with 2M Nh in MeOH afforded 2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-6-(5-propyl-1,2,4-oxadiazol-3-yl)is oindolin-1-one (28 mg, 62%, 0.0559 mmol) as a colorless powder. HPLC/MS m/z: 496.210 [M+H] + , Rt (AJ): 2.33 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.84-8.82 (m, 1H), 8.20 (dd, J = 7.9, 1.6 Hz, 1H), 8.17 (dd, J= 1.5, 0.8 Hz, 1H), 8.14 (dd, J= 8.5, 1.5 Hz, 1H), 8.04 (t, J= 5.6 Hz, 1H), 7.95 (d, J= 5.7 Hz, 1H), 7.84 (d, J= 8.5 Hz, 1H), 7.79 (dd, J= 7.9, 0.9 Hz, 1H), 6.95 (dd, J= 5.9, 0.9 Hz, 1H), 4.68 (s, 2H), 3.90-3.86 (m, 2H), 3.84-3.79 (m, 2H), 2.99 (t, J= 7.4 Hz, 2H), 2.67 (s, 3H), 1.82 (h, J= 7.4 Hz, 2H), 0.99 (t, J= 7.4 Hz, 3H).

The following example was prepared in a similar manner.

Example 76: 6-(5-Ethyl- 1 ,2,4-oxadiazol-3-yl)-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-

1-isoquinolyl]amino]ethyl]isoindolin-1-one

HPLC/MS m/z: 482.195 [M+H] + , Rt (AJ): 2.18 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.85-8.82 (m, 1H), 8.20 (dd, J= 7.8, 1.6 Hz, 1H), 8.17 (dd, J= 1.5, 0.8 Hz, 1H), 8.14 (dd, J= 8.5, 1.6 Hz, 1H), 8.04 (t, J= 5.6 Hz, 1H), 7.95 (d, J= 5.7 Hz, 1H), 7.84 (d, J = 8.5 Hz, 1H), 7.79 (dd, J= 7.8, 0.9 Hz, 1H), 6.95 (dd, J= 5.9, 0.8 Hz, 1H), 4.68 (s, 2H), 3.90-3.86 (m, 2H), 3.84-3.79 (m, 2H), 3.02 (q, J= 7.6 Hz, 2H), 2.67 (s, 3H), 1.35 (t, J= 7.6 Hz, 3H).

Example 77: Propan-2-yl 2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-1,3-dioxo-isoindoline-5-carboxylate

Example 77.1: A solution of 3-(1-chloro-7-isoquinolyl)-5-methyl-1,2,4-oxadiazole [Intermediate A4] (1000.00 mg, 4.0707 mmol) and ethylenediamine (5.44 mL, 81.413 mmol) in NMP (5.00 mL) in a 10-20 mL microwave vial was heated under microwave irradiation for 1 h at 160 °C. A solution of saturated bicarbonate was added (100 mL) and the product was extracted with EtOAc (3 x 50 mL). The organic layer was combined, dried over magnesium sulfate and concentrated under reduced pressure to give N'-[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]ethane-1 ,2- diamine (982 mg, 90%, 3.6465 mmol) as a yellow solid. HPLC/MS m/z: 270.136 [M+H] + , Rt (AE): 0.95 min.

Example 77.2: N'-[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]ethane-1 ,2- diamine (500.00 mg, 1.8567 mmol) and 4-bromophthalic anhydride (442.56 mg, 1.9495 mmol) were mixed in glacial acetic acid (3.71 mL, 0.5000 M) in a microwave vial under argon. The reaction mixture was heated at 150 °C by microwave irradiation for 1 h. Purification by silica gel normal phase column chromatography (eluent: 40-100% EtOAc in cyclohexane) afforded 5-bromo-2-[2-[[7-(5-methyl-1,2,4- oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]isoindoline-1 , 3-dione (761 mg, 85%, 1.5815 mmol) as a pale-yellow powder. HPLC/MS m/z: 478.050 [M+H] + , Rt (AE): 2.18 min.

Example 77.3: 5-Bromo-2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]isoindoline-1 , 3-dione (50.00 mg, 0.1045 mmol) and XantPhos Pd G4 (1.01 mg, 0.0010 mmol) were mixed in a microwave vial. The capped vial was evacuated before fitted with a balloon filled with carbon monoxide. Anyhydrous 1,4-dioxane (0.52 mL), anhydrous iPrOH (0.40 mL, 5.2268 mmol) and DIPEA (0.04 mL, 0.2091 mmol) were added, and the reaction mixture was heated at 80 °C for 6 h. The reaction mixture was concentrated under reduced pressure, dissolved in DMSO and purified by prep-HPLC (AccqPrep, 55-65% ACN in water, pH10, 20 min) to give propan-2-yl 2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-1,3-dioxo-isoindoline-5-carboxylate (16 mg, 31%, 0.0323 mmol) as a pale-yellow solid. HPLC/MS m/z: 486.178 [M+H] + , Rt (AJ): 2.37 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.70 (d, J = 1.6 Hz, 1 H), 8.33 (dd, J = 7.7, 1.4 Hz, 1 H), 8.16 (d, J = 1.4 Hz, 1 H), 8.13 (dd, J = 8.5, 1.6 Hz, 1H), 7.98 (t, J = 6.0 Hz, 2H), 7.96 (d, J = 7.8 Hz, 1H), 7.82 (d, J = 8.4 Hz, 2H), 7.81 (d, J = 5.8 Hz, 2H), 6.90 (d, J = 5.7 Hz, 1H), 5.18 (hept, J = 6.2 Hz, 1H), 3.95-3.91 (m, 2H), 3.83-3.77 (m, 2H), 2.65 (s, 3H), 1.35 (d, J = 6.3 Hz, 6H).

Example 78: Ethyl 2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino ]ethyl]- 1-oxo-3H-pyrrolo[3,4-c]pyridine-6-carboxylate

Example 78.1: 2-Chloro-5-methyl-isonicotinic acid ethyl ester (500.00 mg, 2.5046 mmol) was dissolved in anhydrous DCE (25.05 mL) under argon. AIBN (41.13 mg, 0.2505 mmol) and NBS (445.77 mg, 2.5046 mmol) were added, and the reaction mixture was heated to reflux for 2 h. The reaction mixture was cooled to ambient temperature. Purification by silica gel normal phase column chromatography (eluent: 0-30% EtOAc in cyclohexane) afforded ethyl 5-(bromomethyl)-2-chloro-pyridine-4- carboxylate (577 mg, 62%, 1.5537 mmol) as a colorless oil. HPLC/MS m/z: 277.958 [M+H] + , Rt (AJ): 2.57 min.

Example 78.2: Ethyl 5-(bromomethyl)-2-chloro-pyridine-4-carboxylate (570.00 mg, 1.5348 mmol), DIPEA (0.53 mL, 3.0697 mmol) and 1-Boc-ethylenediamine (0.26 mL, 1.6116 mmol) were mixed in anhydrous MeCN (18.32 mL) under argon and heated at reflux for 30 min. The reaction mixture was cooled to ambient temperature. Purification by silica gel normal phase column chromatography (eluent: 40-100% EtOAc in cyclohexane) afforded tert-butyl N-[2-(6-chloro-1-oxo-3H- pyrrolo[3,4-c]pyridin-2-yl)ethyl]carbamate (449 mg, 94%, 1.4402 mmol) as a colorless, amorphous solid. HPLC/MS m/z: 312.111 [M+H] + , Rt (AJ): 2.12 min. Example 78.3: tert-Butyl N-[2-(6-chloro-1-oxo-3H-pyrrolo[3,4-c]pyridin-2- yl)ethyl]carbamate (150.00 mg, 0.4811 mmol) and Pd(dppf)Ch ■ DCM (41.32 mg, 0.0481 mmol) were mixed in a microwave vial. The vial was capped and evacuated before being fitted with a balloon filled with carbon monoxide. Anyhydrous DMF (0.50 mL), anhydrous ethanol (0.50 mL, 8.5631 mmol) and triethylamine (0.14 mL, 0.9623 mmol) were added and the reaction mixture was heated at 60 °C for 24 h. Purification by silica gel normal phase column chromatography (eluent: 40-100% EtOAc in cyclohexane) yielded ethyl 2-[2-(tert-butoxycarbonylamino)ethyl]-1-oxo-3H- pyrrolo[3,4-c]pyridine-6-carboxylate (73 mg, 43%, 0.2089 mmol) as an amorphous solid. HPLC/MS m/z: 350.172 [M+H] + , Rt (AJ): 2.13 min.

Example 78.4: Ethyl 2-[2-(tert-butoxycarbonylamino)ethyl]-1-oxo-3H-pyrrolo[3,4- c]pyridine-6-carboxylate (73.00 mg, 0.2089 mmol) was mixed with 4 M HCI in 1,4- dioxane (5.22 mL, 20.894 mmol) and anyhydrous 1 ,4-dioxane (5.00 mL) at room temperature under argon and stirred for 16 h. Volatiles were removed under reduced pressure. The crude was dissolved in MeOH and filtered through a 1 g SCX-2 column. The product was released with ammonia in MeOH/EtOH (7M ammonia in MeOH diluted 1 :4 with ethanol to reduce potential transesterification) to give ethyl 2-(2-aminoethyl)-1-oxo-3H-pyrrolo[3,4-c]pyridine-6-carboxyla te (48 mg, 92%, 0.1926 mmol) as a pale-brown oil. HPLC/MS m/z: 250.119 [M+H] + , Rt (AJ): 0.32 min.

Example 78.5: Ethyl 2-(2-aminoethyl)-1-oxo-3H-pyrrolo[3,4-c]pyridine-6-carboxyla te (48.00 mg, 0.1926 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4- oxadiazole [Intermediate A5] (48.13 mg, 0.2118 mmol), PyBrop (98.75 mg, 0.2118 mmol), DIPEA (0.13 mL, 0.7221 mmol) and anhydrous DCM (1.00 mL) were placed in a microwave vial at room temperature under argon. The reaction mixture was heated at 60 °C by microwave irradiation for 1 h. Volatiles were removed under reduced pressure. Purification by silica gel normal phase column chromatography (eluent: 0-15% MeOH in EtOAc) followed by ion exchange SCX-2 chromatography eluting with NHs in MeOH/EtOH (7M ammonia in MeOH diluted 1 :4 with ethanol to reduce potential transesterification). Further purification by reverse phase flash chromatography (eluent: 20-60% MeOH in water) followed by ion exchange SCX-2 chromatography eluting with ammonia soluition afforded ethyl 2-[2-[[7-(5-methyl- 1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-1-oxo-3H-pyr rolo[3,4-c]pyridine-6- carboxylate (29 mg, 33%, 0.0632 mmol) as an off-white, amorphous solid.

HPLC/MS m/z: 459.178 [M+H] + , Rt (AJ): 1.89 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 9.03 (d, J = 1.1 Hz, 1 H), 8.80-8.79 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.13 (d, J = 1.1 Hz, 1 H), 8.01 (t, J = 5.7 Hz, 1 H), 7.91 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.94 (dd, J = 5.9, 0.8 Hz, 1 H), 4.79 (s, 2H), 4.36 (q, J = 7.1 Hz, 2H), 3.91-3.88 (m, 2H), 3.85-3.80 (m, 2H), 2.67 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

Example 79: 2-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-6- (propylamino)isoindolin-l-one

Example 79.1: tert-Butyl N-[2-(6-bromo-1-oxo-isoindolin-2-yl)ethyl]carbamate [Example 56.1] (200.00 mg, 0.5630 mmol), cesium carbonate (369.15 mg, 1.126 mmol), BrettPhos Pd G3 (5.10 mg, 0.0056 mmol) and BrettPhos (3.02 mg, 0.0056 mmol) were mixed in a microwave vial under argon. 2-Methyl-2-butanol (2.82 mL) and propylamine (0.06 mL, 0.6756 mmol) were added and the reaction mixture was heated at 100 °C for 1.5 h. The reaction mixture was cooled to ambient temperature. Purification by column chromatography (eluent: 40-100% EtOAc in cyclohexane) afforded tert-butyl N-[2-[1-oxo-6-(propylamino)isoindolin-2-yl]ethyl]carbamate (149 mg, 79%, 0.4469 mmol). HPLC/MS m/z: 334.212 [M+H] + , Rt (AJ): 2.30 min.

Example 79.2: Analogous procedures to those used in Example 78.4 and Example 78.5 afforded 2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-6- (propylamino)isoindolin-l-one (29 mg, 0.0633 mmol) as an off-white solid. HPLC/MS m/z: 443.220 [M+H] + , Rt (AJ): 2.04 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.86-8.83 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1H), 8.03 (t, J = 5.5 Hz, 1 H), 7.96 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.22 (d, J = 8.2 Hz, 1H), 6.95 (d, J = 5.7 Hz, 1 H), 6.78 (dd, J = 8.2, 2.3 Hz, 1 H), 6.72 (d, J = 2.2 Hz, 1 H), 5.80 (t, J = 5.5 Hz, 1 H), 4.39 (s, 2H), 3.82-3.78 (m, 2H), 3.78-3.73 (m, 2H), 2.97 (td, J = 7.1, 5.5 Hz, 2H), 2.68 (s, 3H), 1.60-1.50 (m, 2H), 0.93 (t, J = 7.4 Hz, 3H).

Example 80: Ethyl 6-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino ]ethyl]-

7-oxo-5H-pyrrolo[3,4-b]pyridine-2-carboxylate Example 80.1: Methyl 6-chloro-3-methyl-pyridine-2-carboxylate (1000.00 mg, 5.3876 mmol) was dissolved in anhydrous DCE (53.88 mL) under argon. AIBN (88.47 mg, 0.5388 mmol) and NBS (958.89 mg, 5.3876 mmol) were added, and the reaction mixture was heated to reflux for 2 h. The reaction mixture was cooled to ambient temperature. Purification by silica gel normal phase column chromatography (eluent: 0-30% EtOAc in cyclohexane) afforded methyl 3-(bromomethyl)-6-chloro-pyridine-2- carboxylate (1.12 g, 79%, 4.2457 mmol) as a colorless, crystalline solid. HPLC/MS m/z: 287.986 [M+Na] + , Rt (AJ): 2.07 min.

Example 80.2: Methyl 3-(bromomethyl)-6-chloro-pyridine-2-carboxylate (1.12 g, 3.3875 mmol), DIPEA (1.18 mL, 6.775 mmol) and 1-Boc-ethylenediamine (0.56 mL, 3.5569 mmol) were mixed in anhydrous MeCN (33.88 mL) under argon and heated at reflux for 1.5 h. The reaction mixture was cooled to ambient temperature. Purification by silica gel normal phase column chromatography (eluent: 0-100% EtOAc in cyclohexane) afforded tert-butyl N-[2-(2-chloro-7-oxo-5H-pyrrolo[3,4- b]pyridin-6-yl)ethyl]carbamate (748 mg, 71%, 2.3993 mmol) as a colorless, crystalline solid. HPLC/MS m/z: 334.170 [M+H] + , Rt (AJ): 1.99 min.

Example 80.3: tert-Butyl N-[2-(2-chloro-7-oxo-5H-pyrrolo[3,4-b]pyridin-6- yl)ethyl]carbamate (150.00 mg, 0.4811 mmol), palladium acetate (2.18 mg, 0.0096 mmol) and XantPhos (11.14 mg, 0.0192 mmol) were mixed in a microwave vial. The vial was capped, flushed with argon and evacuated before being fitted with a balloon filled with carbon monoxide. Anyhydrous 1 ,4-dioxane (0.96 mL), DI PEA (0.17 mL, 0.9623 mmol) and anhydrous ethanol (0.14 mL, 2.4057 mmol) were added and the reaction mixture was heated at 90 °C for 72 h. Purification by silica gel normal phase column chromatography (eluent: 0-10% MeOH in EtOAc) afforded ethyl 6-[2- (tert-butoxycarbonylamino)ethyl]-7-oxo-5H-pyrrolo[3,4-b]pyri dine-2-carboxylate (41 mg, 24%, 0.1174 mmol) as an amorphous solid. HPLC/MS m/z: 350.247 [M+H] + , Rt (Al): 1.22 min.

Example 80.4: Ethyl 6-[2-(tert-butoxycarbonylamino)ethyl]-7-oxo-5H-pyrrolo[3,4- b]pyridine-2-carboxylate (41.00 mg, 0.1174 mmol) was mixed with 4 M HCI in 1 ,4- dioxane (1.47 mL, 5.8675 mmol) and anyhydrous 1,4-dioxane (3.00 mL) at room temperature under argon and stirred for 2 h. Volatiles were removed under reduced pressure. The crude was dissolved in MeOH and filtered through a 1 g SCX-2 column. The product was released with 2M ammonia in MeOH to give ethyl 6-(2- aminoethyl)-7-oxo-5H-pyrrolo[3,4-b]pyridine-2-carboxylate (32 mg, 88%, 0.1027 mmol) as an off-white solid. HPLC/MS m/z: 250.184 [M+H] + , Rt (Al): 0.43 min. Example 80.5: Ethyl 6-(2-aminoethyl)-7-oxo-5H-pyrrolo[3,4-b]pyridine-2-carboxyla te (30.00 mg, 0.0963 mmol), 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4- oxadiazole [Intermediate A5] (26.25 mg, 0.1155 mmol), PyBrop (53.86 mg, 0.1155 mmol), DIPEA (0.06 mL, 0.3611 mmol) and anhydrous DCM (0.50 mL) in a microwave vial at room temperature under argon. The reaction mixture was heated at 60 °C by microwave irradiation for 1 h. Volatiles were removed under reduced pressure. Purification by reverse phase flash (eluent: 20-60% MeOH/water + 0.1 % formic acid) followed by ion exchange SCX-2 chromatography eluting with MeOH/EtOH (7M ammonia in MeOH diluted 1 :4 with ethanol to reduce potential transesterification). Further purification by column chromatography (eluent: 0-20% MeOH in EtOAc) afforded ethyl 6-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-7-oxo-5H-pyrrolo[3,4-b]pyridine-2-c arboxylate (9.3 mg, 20%, 0.0194 mmol) as an amorphous solid. HPLC/MS m/z: 459.174 [M+H] + , Rt (AJ): 1.84 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.82-8.79 (m, 1 H), 8.25 (d, J = 8.0 Hz, 1 H), 8.20 (d, J = 7.9 Hz, 1 H), 8.14 (dd, J = 8.5, 1.6 Hz, 1 H), 8.03 (t, J = 5.7 Hz, 1 H), 7.92 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.94 (dd, J = 5.9, 0.9 Hz, 1 H), 4.69 (s, 2H), 4.37 (q, J = 7.1 Hz, 2H), 3.91 (t, J = 5.8 Hz, 2H), 3.86-3.80 (m, 2H), 2.67 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

The following example was prepared in a similar manner.

Example 81 : N-Ethyl-6-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-7-oxo-5H-pyrrolo[3,4-b]pyridine-2-c arboxamide

2.6 mg, amorphous solid. HPLC/MS m/z: 458.194 [M+H] + , Rt (AJ): 1.74 min. 1 H NMR (600 MHz, Chloroform-d): 5 8.59-8.55 (m, 1 H), 8.33 (d, J = 7.9 Hz, 1 H), 8.22- 8.15 (m, 2H), 7.94 (d, J = 5.8 Hz, 1 H), 7.91 (dd, J = 7.9, 0.7 Hz, 1 H), 7.70 (d, J = 8.5 Hz, 1 H), 6.90 (dd, J = 5.9, 0.9 Hz, 1 H), 6.33 (s, 1 H), 4.63 (s, 2H), 4.10 (dd, J = 6.7, 4.5 Hz, 2H), 4.07-4.01 (m, 2H), 3.47 (qd, J = 7.3, 6.0 Hz, 2H), 2.64 (s, 3H), 1.22 (t, J = 7.3 Hz, 3H).

Example 82: 2-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-6- nitro-isoindolin-1-one

N'-[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]ethane-1 ,2-diamine [Example 77.1] (200.00 mg, 0.7427 mmol), methyl 2-(bromomethyl)-5-nitrobenzoate (203.54 mg, 0.7427 mmol) and DIPEA (0.26 mL, 1.4853 mmol) were mixed in anhydrous MeCN (7.43 mL) under argon. The reaction mixture was heated at reflux for 1.5 h. Purification by silica gel normal phase column chromatography (eluent: 0-20% MeOH in EtOAc) afforded 2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-6-nitro-isoindolin-1-one (206 mg, 63%, 0.4700 mmol) as a light-brown powder. HPLC/MS m/z: 431.146 [M+H] + , Rt (AJ): 1.90 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 8.82-8.79 (m, 1 H), 8.43 (dd, J = 8.3, 2.2 Hz, 1 H), 8.29 (d, J = 2.2 Hz, 1 H), 8.14 (dd, J = 8.5, 1.6 Hz, 1 H), 8.03 (t, J = 5.6 Hz, 1 H), 7.93 (d, J = 5.7 Hz, 1 H), 7.89 (d, J = 8.3 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.94 (dd, J = 5.9, 0.9 Hz, 1 H), 4.74 (s, 2H), 3.91-3.86 (m, 2H), 3.85-3.79 (m, 2H), 2.67 (s, 3H).

Example 83: N-[2-[2-[[7-(5-Methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-3- oxo-isoindolin-5-yl]propanamide 2-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino ]ethyl]-6-nitro-isoindolin- 1-one [Example 82] (50.00 mg, 0.1141 mmol) was dissolved in anhydrous DMF (0.30 mL) under argon. Zinc powder (29.83 mg, 0.4563 mmol) and glacial acetic acid (0.05 mL, 0.9126 mmol) were added successively, and the reaction mixture was heated at 40 °C for 1 h. The reaction mixture was cooled to ambient temperature. Triethylamine (0.06 mL, 0.4563 mmol) and propionyl chloride (0.01 mL, 0.1255 mmol) were added and the reaction mixture was continued to stir for 1 h. Volatiles were removed under reduced pressure. The crude material was purified by prep-HPLC (AccqPrep, 10-80% MeOH in water, pH3, 15 min) to afford N-[2-[2-[[7- (5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-3 -oxo-isoindolin-5- yl]propanamide (27mg, 51%, 0.0578 mmol). HPLC/MS m/z: 457.198 [M+H] + , Rt (AJ): 1.86 min. 1 H NMR (600 MHz, DMSO-d 6 ): 5 10.03 (s, 1 H), 8.85-8.82 (m, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.04 (t, J = 5.5 Hz, 1 H), 7.98 (d, J = 2.0 Hz, 1 H), 7.95 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.68 (dd, J = 8.2, 2.0 Hz, 1 H), 7.48 (d, J = 8.2 Hz, 1 H), 6.96-6.93 (m, 1 H), 4.51 (s, 2H), 3.86-3.81 (m, 2H), 3.81-3.75 (m, 2H), 2.68 (s, 3H), 2.33 (q, J = 7.5 Hz, 2H), 1.08 (t, J = 7.6 Hz, 3H).

Example 84: Ethyl 6-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4,5,6,7-tetrahydrothieno[2,3-c]pyridine-2-ca rboxylate

Example 84.1 : To a solution of ethyl 4,5,6,7-tetrahydrothieno[2,3-c]pyridine-2- carboxylate hydrochloride (100 mg, 0.4 mmol) in anhydrous DMF (1.4 mL) was added N-Boc-2-chloroethylamine (181.3 mg, 1.0 mmol), potassium carbonate (139.5 mg, 1.0 mmol) and potassium iodide (2.7 mg, 0.02 mmol). The reaction mixture was stirred at 70 °C overnight. The reaction progress was followed by LC-MS and after 19 h showed nearly reaction completion. The reaction mixture was diluted with ethyl acetate (90 mL) and water (30 mL). After phase separation, the organic layer was washed with brine (50 mL), dried over magnesium sulfate, filtered, and evaporate to dryness. The obtained crude was purified by flash silica column chromatography (eluent: 20-70% ethyl acetate in cyclohexane) to afford ethyl 6-[2- (tert-butoxycarbonylamino)ethyl]-5,7-dihydro-4H-thieno[2,3-c ]pyridine-2-carboxylate (63.2 mg, 44%, 0.18 mmol) as a light-yellow oil. HPLC/MS m/z: 355.2 [M+H] + , Rt (Al): 1.194 min.

Example 84.2: Trifluoroacetic acid (0.5 mL, 6.4 mmol) was added drop wise to a solution of ethyl 6-[2-(tert-butoxycarbonylamino)ethyl]-5,7-dihydro-4H-thieno[ 2,3- c]pyridine-2-carboxylate (56.4 mg, 0.16 mmol) in dichloromethane (0.5 mL) and this was stirred for 2 h 15 min. The volatiles were then evaporated to dryness. The obtained crude was purified by SCX-2 cartridge (2 g, 15 mL) using methanol and 2M ammonia solution in methanol as eluents. Basic fractions were combined and evaporated to dryness to give ethyl 6-(2-aminoethyl)-5,7-dihydro-4H-thieno[2,3- c]pyridine-2-carboxylate (36.4 mg, 90%, 0.14 mmol) as a light-orange oil. HPLC/MS m/z: 255.1 [M+H] + , Rt (AC): 0.68 min.

Example 84.3: 6-(2-Aminoethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-2-carb oxylate (33.6 mg, 0.13 mmol) was dissolved in anhydrous dichloromethane (1.32 mL), followed by addition of N,N-diisopropylethylamine (86 uL, 0.50 mmol) followed by 5- methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1,2,4-oxadiazole [Intermediate A5] (30 mg, 0.13 mmol) and PyBroP (80 mg, 0.17 mmol). After 16 h, LC-MS showed good conversion to the desired product. The crude was purified by flash column chromatography (eluent: gradient of 10-100% ethyl acetate in cyclohexane) to give ethyl 6-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino ]ethyl]-5,7-dihydro- 4H-thieno[2,3-c]pyridine-2-carboxylate that wasn't pure. The product was further purified by reverse phase column (eluent: 30-100% of MeOH in water both modified with 0.1% formic acid). Fractions containing the product were combined, evaporated, and filtered through a SCX-II cartridge (2 g, 15 mL) using MeOH and 2M ammonia solution as eluents. Basic fractions were evaporated to dryness in vacuum to give ethyl 6-[2-[[7-(5-methyl-1,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-5,7-dihydro-4H-thieno[2,3-c]pyridin e-2-carboxylate (15 mg, 25%, 0.03 mmol) as a light-yellow powder. HPLC/MS m/z: 464.2 [M+H] + , Rt (AC): 1.19 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.85 (s, 1H), 8.23 (dd, J = 8.5, 1.6 Hz, 1 H), 7.93 (d, J = 5.9 Hz, 1 H), 7.82 (d, J = 8.4 Hz, 1H), 7.54 (s, 1 H), 6.99 (d, J = 6.1 Hz, 1H), 4.32 (q, J = 7.1 Hz, 2H), 3.90 (d, J = 1.6 Hz, 2H), 3.83 (t, J = 6.4 Hz, 2H), 2.99 (m, 4H), 2.87-2.82 (m, 2H), 2.69 (s, 3H), 1.36 (t, J = 7.1 Hz, 3H). Example 85: Methyl 6-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4,5,6,7-tetrahydrothieno[2,3-c]pyridine-2-ca rboxylate

Example 85.1: Triethylamine (71 uL mL, 0.51 mmol) was added to a solution of methyl 4H,5H,6H,7H-thieno[2,3-c]pyridine-2-carboxylate hydrochloride (125 mg, 0.51 mmol) in anhydrous chloroform (2.53 mL). The reaction mixture was cooled to 0 °C and a 1M solution of 1-nitroethene in xylene (0.51 mL, 0.51 mmol) was added. The reaction mixture was stirred at 0 °C for 10 min and then warmed up to room temperature. The reaction mixture was concentrated in vacuum and the obtained crude was purified by flash silica column chromatography (eluent: 20-80% ethyl acetate in cyclohexane) to give methyl 6-(2-nitroethyl)-5,7-dihydro-4H-thieno[2,3- c]pyridine-2-carboxylate (95.9 mg, 70%, 0.35 mmol) as a colorless, amorphous solid. HPLC/MS m/z: 271.1 [M+H] + , Rt (Al): 0.684 min.

Example 85.2: Zinc (185.6 mg, 2.83 mmol), ammonium chloride (151.8 mg, 2.83 mmol) and methyl 6-(2-nitroethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-2-carb oxylate (95.9 mg, 0.35 mmol) were suspended in a mixture of tetrahydrofuran (1.4 mL) / water (0.45 mL) / MeOH (0.5 mL). The obtained reaction mixture was stirred at room temperature. After 2 h, the reaction mixture was filtered on celite and rinsed with a 1 :1 mixture of dichloromethane:methanol (20 mL). The obtained filtrate was passed through a Biotage Isolute NH2 ion exchange column (2 g, 15 mL) and rinsed with a 1 :1 mixture of dichloromethane:methanol. The filtrate was concentrated to give methyl 6-(2-aminoethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-2-carb oxylate (70 mg, 82%, 0.29 mmol) as a sticky clear oil. This compound was taken through to the next step without further purification. HPLC/MS m/z: 241 [M+H] + , Rt (AC): 0.38 min. Example 85.3: Methyl 6-(2-aminoethyl)-5,7-dihydro-4H-thieno[2,3-c]pyridine-2- carboxylate (68.8 mg, 0.29 mmol) was dissolved in anhydrous dichloromethane (2.9 mL) followed by addition of N,N-diisopropylethylamine (187 uL, 1.1 mmol), 5-methyl- 3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole [Intermediate A5] (65 mg, 0.29 mmol) and PyBroP (173 mg, 0.37 mmol). After 19 h 30 min the reaction mixture was evaporated to dryness and the crude was purified with by reverse phase column (eluent: 20-100% gradient of methanol in water both modified with 0.1% formic acid) to give methyl 6-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1-isoquinolyl]amino]ethyl]-5,7- dihydro-4H-thieno[2,3-c]pyridine-2-carboxylate (24.4 mg, 19%, 0.05 mmol) as a beige solid. HPLC/MS m/z: 450.2 [M+H] + , Rt (AC): 1.147 min. 1 H NMR (500 MHz, DMSO-d 6 ): 6 8.87 (s, 1 H), 8.14 (dd, J = 8.4, 1.5 Hz, 1 H), 7.96 (d, J = 5.7 Hz, 1 H), 7.86 (t, J = 5.6 Hz, 1 H), 7.83 (d, J = 8.5 Hz, 1 H), 7.54 (s, 1 H), 6.93 (d, J = 5.8 Hz, 1 H), 3.79-3.77 (m, 5H), 3.71 (q, J = 6.4 Hz, 2H), 2.85-2.78 (m, 4H), 2.69 (s, 3H), 2.69-2.66 (m, 2H).

Example 86: Methyl 6-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4,5,6,7-tetrahydrothieno[2,3-c]pyridine-2-ca rboxylate

Preparation in a similar manner as described for example 55.

39 mg pale-yellow solid. HPLC/MS m/z: 459.15 [M+H] + , Rt (C): 0.96 min. 1 H NMR (300 MHz, DMSO-cfe): 6 8.83 (s, 1 H), 8.14 (dd, J = 8.5, 1.5 Hz, 1 H), 8.03 (s, 1 H), 7.95 (d, J = 5.8 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 7.76 (s, 1 H), 6.95 (d, J = 5.8 Hz, 1 H), 4.44 (s, 2H), 4.26 (t, J = 6.6 Hz, 2H), 3.81 (dd, J = 13.6, 4.9 Hz, 3H), 2.68 (d, J = 1.4 Hz, 3H), 2.21 (s, 3H), 1.83-1.65 (m, 2H), 0.97 (t, J = 7.4 Hz, 3H).

Example 87: Ethyl 1-methyl-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-1 H-1 ,3-benzodiazole-5-carboxylate Example 87.1 : A solution of ethyl 3-amino-4-(methylamino)benzoate (300.0 mg, 1.545 mmol), 3-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino]propanoic acid (307.0 mg, 1.027 mmol) and NMI (190.0 mg, 2.314 mmol) in DMF (5.0 mL) was stirred for 5 min at room temperature. TCFH (261.0 mg, 0.930 mmol) was added, and the resulting mixture was stirred at room temperature overnight. The reaction mixture was evaporated, and the residue was purified by reverse flash chromatography to give ethyl 3-(3-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino]propanamido)-4-(methylamino)benzoate (259 mg, 35%) as a yellow solid. HPLC/MS m/z: 475.25 [M+H] + , Rt (J): 0.75 min.

Example 87.2: A solution of ethyl 3-(3-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin- 1-yl]amino]propanamido)-4-(methylamino)benzoate (85.5 mg, 0.180 mmol) in AcOH (3.0 mL) was stirred at 80 °C overnight. The reaction mixture was cooled to room temperature, diluted with brine (3 mL) and extracted with EtOAc (3 x 3 mL). The combined organic layers were dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by prep-HPLC yielded ethyl 1-methyl-2-(2-[[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino]ethyl)-1 ,3-benzodiazole-5-carboxylate (20.0 mg, 24%) as a colorless solid. HPLC/MS m/z: 457.10 [M+H] + , Rt (K): 1.00 min. 1 H NMR (300 MHz, DMSO-cfe): 6 8.85 (s, 1 H), 8.21-8.08 (m, 3H), 7.99 (d, J = 5.7 Hz, 1 H), 7.90-7.80 (m, 2H), 7.60 (d, J = 8.5 Hz, 1 H), 6.97 (d, J = 5.7 Hz, 1 H), 4.31 (q, J = 7.1 Hz, 2H), 4.02- 3.89 (m, 2H), 3.80 (s, 3H), 3.32 (s, 1 H), 3.27 (s, 1 H), 2.68 (s, 3H), 1.33 (t, J = 7.1 Hz, 3H).

The following compounds were prepared in a similar manner:

Example 88: Propan-2-yl 1-methyl-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-1 H-1 ,3-benzodiazole-5-carboxylate

7 mg colorless solid. HPLC/MS m/z: 471.15 [M+H] + , Rt (B): 0.85 min. 1 H NMR (300 MHz, DMSO-cfe): 6 8.84 (s, 1 H), 8.24-8.07 (m, 3H), 7.98 (d, J = 5.7 Hz, 1 H), 7.90- 7.78 (m, 2H), 7.59 (d, J = 8.5 Hz, 1 H), 6.97 (d, J = 5.8 Hz, 1 H), 5.20-5.04 (m, 1 H), 3.94 (t, J = 6.9 Hz, 2H), 3.80 (s, 3H), 3.27 (s, 2H), 2.68 (s, 3H), 1.32 (d, J = 6.2 Hz, 6H).

Example 89: tert-Butyl 1-methyl-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin- 1-yl]amino}ethyl)-1 H-1 ,3-benzodiazole-5-carboxylate

4 mg colorless solid. HPLC/MS m/z: 485.25 [M+HJ+, Rt (W): 1.95 min. 1 H NMR (300 MHz, DMSO-d 6 ): 6 8.84 (s, 1 H), 8.19-8.08 (m, 3H), 7.98 (d, J = 5.7 Hz, 1 H), 7.85 (d, J = 8.5 Hz, 1 H), 7.82-7.73 (m, 1 H), 7.56 (d, J = 8.4 Hz, 1 H), 6.97 (d, J = 5.7 Hz, 1 H), 4.01-3.89 (m, 2H), 3.79 (s, 3H), 3.28 (d, J = 7.2 Hz, 2H), 2.68 (s, 3H), 1.55 (s, 9H).

Example 90: Ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

Example 90.1 : Preparation as described for Example 87.1 using methyl 5,6- diaminopyridine-3-carboxylate (659.0 mg, 3.942 mmol) and 3-{[7-(5-methyl-1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl]amino}propanoic acid (1.39 g, 4.643 mmol), NMI (2.42 g, 29.507 mmol) and TCFH (2.28 g, 8.126 mmol) in DMF (20.0 mL). Yield: 770 mg (44%) of methyl 5-amino-6-(3-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}propanamido)pyridine-3-carboxylate as a black solid. HPLC/MS m/z: 448.20 [M+H] + , Rt (B): 0.62 min.

Example 90.2: Cyclisation as described for Example 87.2. Yield: 108 mg (44%) of methyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-3H- imidazo[4,5-b]pyridine-6-carboxylate as a black solid. HPLC/MS m/z: 430.10 [M+H] + , Rt (B): 0.72 min. Example 90.3: Saponification with lithium hydroxide in THF and water yielded 50 mg of 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-3H- imidazo[4,5-b]pyridine-6-carboxylic acid as a light-brown solid. HPLC/MS m/z: 416.05 [M+H] + , Rt (B): 0.61 min.

Example 90.4: Esterification with H2SO4 in ethanol provided 5 mg of ethyl 2-(2-((7- (5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-3H-imidazo [4,5- b]pyridine-6-carboxylate as a colorless solid. HPLC/MS m/z: 444.25 [M+H] + , Rt (X): 1.94 min. 1 H NMR (300 MHz, DMSO-cfe): 5 13.42-12.81 (m, 1 H), 8.94-8.78 (m, 2H), 8.43-8.20 (m, 1 H), 8.18-8.04 (m, 2H), 7.98 (d, J = 5.7 Hz, 1 H), 7.84 (d, J = 8.5 Hz, 1 H), 6.97 (d, J = 5.8 Hz, 1 H), 4.34 (q, J = 7.1 Hz, 2H), 3.98 (q, J = 6.5 Hz, 2H), 3.30- 3.25 (s, 2H), 2.67 (s, 3H), 1.33 (t, J = 7.1 Hz, 3H).

The following compounds were prepared in a similar manner:

Example 91 : Propan-2-yl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

7 mg colorless solid. HPLC/MS m/z: 458.25 [M+H] + , Rt (W): 1.58 min. 1 H NMR (300 MHz, DMSO-cfe): 6 8.88-8.80 (m, 2H), 8.31 (s, 1 H), 8.19-8.10 (m, 2H), 7.97 (d, J = 5.8 Hz, 1 H), 7.85 (d, J = 8.6 Hz, 1 H), 6.98 (d, J = 5.8 Hz, 1 H), 5.15 (hept, J = 6.2 Hz, 1 H), 3.98 (q, J = 6.4 Hz, 2H), 3.28 (s, 2H), 2.67 (s, 3H), 1.33 (d, J = 6.2 Hz, 6H).

Example 92: tert-Butyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate 5 mg light-brown solid. HPLC/MS m/z: 472.25 [M+H] + , Rt (W): 1.57 min. 1 H NMR (300 MHz, DMSO-cfe): 613.28 (s, 1H), 8.82 (d, J= 14.1 Hz, 2H), 8.28 (s, 1H), 8.19- 8.05 (m, 2H), 7.98 (d, J= 5.8 Hz, 1H), 7.84 (d, J= 8.6 Hz, 1H), 6.97 (d, J= 5.8 Hz, 1H), 4.02-3.92 (m, 2H), 3.32-3.28 (m, 2H), 2.67 (s, 3H), 1.56 (s, 9H).

Example 93: Ethyl 3-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1- yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

22 mg colorless solid. HPLC/MS m/z: 458.05 [M+H] + , Rt (B): 0.71 min. 1 H NMR (400 MHz, DMSO-cfe): 68.89 (d, J= 1.9 Hz, 1H), 8.87-8.81 (m, 1H), 8.42 (d, J= 1.9 Hz, 1H), 8.19-8.09 (m, 2H), 7.97 (d, J= 5.7 Hz, 1H), 7.86 (d, J= 8.5 Hz, 1H), 6.98 (d, J = 5.7 Hz, 1H), 4.40-4.32 (m, 2H), 4.03-3.96 (m, 2H), 3.83 (s, 3H), 3.50-3.25 (m, 2H), 2.69 (s, 3H), 1.35 (t, J= 7.1 Hz, 3H).

Example 94: Propan-2-yl 3-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6 -carboxylate

18 mg colorless solid. HPLC/MS m/z: 472.10 [M+H] + , Rt (B): 0.75 min. 1 H NMR (400 MHz, DMSO-cfe): 68.89-8.83 (m, 2H), 8.40 (d, J= 1.9 Hz, 1H), 8.18-8.08 (m, 2H), 7.97 (d, J= 5.7 Hz, 1H), 7.86 (d, J= 8.5 Hz, 1H), 6.98 (d, J= 5.8 Hz, 1H), 5.23-5.14 (m, 1H), 4.03-3.96 (m, 2H), 3.84 (s, 3H), 3.50-3.25 (m, 2H), 2.69 (s, 3H), 1.35 (d, J = 6.2 Hz, 6H).

Example 95: tert-Butyl 3-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin - 1-yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

11 mg colorless solid. HPLC/MS m/z: 486.20 [M+H] + , Rt (B): 0.80 min. 1 H NMR (400 MHz, DMSO-cfe): 6 8.86-8.82 (m, 2H), 8.36 (d, J = 2.0 Hz, 1 H), 8.16 (d, J = 8.5, 1.5 Hz, 1 H), 8.11 (t, J = 5.6 Hz, 1 H), 7.97 (d, J = 5.7 Hz, 1 H), 7.86 (d, J = 8.5 Hz, 1 H), 6.98 (d, J = 5.7 Hz, 1 H), 4.00 (d, J = 6.3 Hz, 2H), 3.83 (s, 3H), 3.50-3.25 (m, 2H), 2.69 (s, 3H), 1.58 (s, 9H).

Example 96: Ethyl 5-methyl-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

Example 96.1 : To a stirred solution of 5-bromo-6-methylpyridine-2,3-diamine (1.57 g, 7.770 mmol) and 3-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}propanoic acid (2.17 g, 7.275 mmol) in DMF (20.0 mL) were added 1- methyl-1 H-imidazole (1.52 g, 18.513 mmol) and TCFH (3.04 g, 10.835 mmol). The resulting mixture was stirred at room temperature for 16 h. The resulting mixture was concentrated under vacuum and the residue purified by silica gel column chromatography (eluent: MeOH/DCM - 5:95) to afford 1.3 g (35%) of N-(2-amino-5- bromo-6-methylpyridin-3-yl)-3-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}propanamide as a brown solid. HPLC/MS m/z: 484.15 [M+H] + , Rt (B): 0.61 min.

Example 96.2: A mixture of N-(2-amino-5-bromo-6-methylpyridin-3-yl)-3-[[7-(5- methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino]propanamide (180.0 mg, 0.373 mmol) in AcOH (2.0 mL) was stirred at 150 °C for 20 min. The resulting mixture was codes to room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: Dichloromethane/MeOH - 10:1) to afford 115 mg (66%) of N-(2-[6-bromo-5-methyl-3H-imidazo[4,5-b]pyridin-2- yl]ethyl)-7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-amine as a brown solid. HPLC/MS m/z: 466.00 [M+H] + , Rt (R): 0.72 min.

Example 96.3: Carbonylation using the described conditions yielded 8 mg (7%) of ethyl 5-methyl-2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)- 3H-imidazo[4,5-b]pyridine-6-carboxylate as a colorless solid. HPLC/MS m/z: 458.10 [M+H] + , Rt (K): 0.85 min. 1 H NMR (300 MHz, DMSO-d 6 ): 5 12.93 (s, 1 H), 8.85 (d, J =

1.5 Hz, 1 H), 8.27 (s, 1 H), 8.18-8.06 (m, 2H), 7.98 (d, J = 5.7 Hz, 1 H), 7.84 (d, J =

8.5 Hz, 1 H), 6.97 (d, J = 5.8 Hz, 1 H), 4.29 (q, J = 7.1 Hz, 2H), 3.96 (q, J = 6.7 Hz, 2H), 3.24 (d, J = 7.0 Hz, 2H), 2.71 (d, J = 22.4 Hz, 6H), 1.32 (t, J = 7.1 Hz, 3H).

The following compounds were prepared in a similar manner:

Example 97: Ethyl 3,5-dimethyl-2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

46 mg colorless solid. HPLC/MS m/z: 472.10 [M+H] + , Rt (B): 0.74 min. 1 H NMR (400 MHz, DMSO-cfe): 6 8.85 (d, J = 1.4 Hz, 1 H), 8.37 (s, 1 H), 8.17 (dd, J = 8.5, 1.5 Hz, 1 H), 8.14-8.08 (m, 1 H), 7.99 (d, J = 5.7 Hz, 1 H), 7.87 (d, J = 8.5 Hz, 1 H), 6.99 (d, J = 5.7 Hz, 1 H), 4.37-4.28 (m, 2H), 4.03-3.94 (m, 2H), 3.79 (s, 3H), 3.45-3.25 (m, 2H), 2.82 (s, 3H), 2.70 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

Example 98: Propan-2-yl 3,5-dimethyl-2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-3H-imidazo[4,5-b]pyridine-6 -carboxylate 21 mg colorless solid. HPLC/MS m/z: 486.15 [M+H] + , Rt (B): 0.78 min. 1 H NMR (400 MHz, DMSO-cfe): 68.86 (d, J=1.5 Hz, 1H), 8.34 (s, 1H), 8.17 (dd, J = 8.5, 1.6 Hz, 1H), 8.15-8.11 (m, 1H), 7.99 (d, J= 5.7 Hz, 1H), 7.88 (d, J= 8.5 Hz, 1H), 6.99 (d, J = 5.7 Hz, 1H), 5.20-5.10 (m, 1H), 4.04-3.94 (m, 2H), 3.79 (s, 3H), 3.37-3.34 (m, 2H), 2.81 (s, 3H), 2.70 (s, 3H), 1.35 (d, J= 6.2 Hz, 6H).

Example 99: tert-Butyl 3,5-dimethyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-3H-imidazo[4,5-b]pyridine-6 -carboxylate

34 mg colorless solid. HPLC/MS m/z: 500.15 [M+H] + , Rt (B): 0.81 min. 1 H NMR (400 MHz, DMSO-cfe): 68.85 (s, 1H), 8.30 (s, 1H), 8.17 (dd, J= 8.5, 1.6 Hz, 1H), 8.13- 8.09 (m, 1H), 7.99 (d, J= 5.7 Hz, 1H), 7.87 (d, J= 8.5 Hz, 1H), 6.99 (d, J= 5.7 Hz, 1H), 4.04-3.94 (m, 2H), 3.79 (s, 3H), 3.39-3.33 (m, 2H), 2.79 (s, 3H), 2.70 (s, 3H), 1.58 (s, 9H).

Example 100: Ethyl 6-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3-yl)isoquinolin -1- yl]amino}ethyl)-1H-imidazo[4,5-b]pyridine-5-carboxylate

3 mg colorless solid. HPLC/MS m/z: 458.10 [M+H] + , Rt (L): 0.60 min. 1 H NMR (300 MHz, DMSO-cfe): 612.89 (s, 1H), 8.85 (d, J= 1.6 Hz, 1H), 8.18-8.04 (m, 2H), 7.98 (d, J= 5.8 Hz, 1H), 7.89-7.77 (m, 2H), 6.97 (d, J= 5.8 Hz, 1H), 4.31 (q, J= 7.1 Hz, 2H), 3.96 (q, J = 6.6 Hz, 2H), 3.45-3.25 (m, 2H), 2.67 (s, 3H), 2.53 (s, 3H), 1.31 (t, J = 7.1 Hz, 3H).

Example 101: Propan-2-yl 6-methyl-2-(2-{[7-(5-methyl-1,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-1H-imidazo[4,5-b]pyridine-5 -carboxylate

4 mg pale-yellow solid. HPLC/MS m/z: 472.10 [M+H] + , Rt (L): 0.62 min. 1 H NMR (300 MHz, DMSO-cfe): 6 12.86 (s, 1H), 8.84 (d, J = 1.5 Hz, 1 H), 8.18-8.04 (m, 2H), 7.98 (d, J = 5.8 Hz, 1H), 7.89-7.73 (m, 2H), 6.97 (d, J = 5.8 Hz, 1H), 5.15 (hept, J = 6.3 Hz, 1 H), 3.96 (q, J = 6.6 Hz, 2H), 3.40-3.20 (m, 2H), 2.67 (s, 3H), 2.51 (s, 3H), 1.31 (d, J = 6.2 Hz, 6H).

Example 102: Ethyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-1,3-benzothiazole-5-carboxylate

Example 102.1: A solution of 2-amino-4-bromobenzenethiol (117.0 mg, 0.573 mmol) and 3-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino]propanenitrile (160.0 mg, 0.573 mmol) in AcOH (3.0 mL) and EtOH (3.00 mL) was stirred at 80 °C under nitrogen atmosphere overnight. The resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was diluted with water (5 mL). The aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried with sodium sulfate, filtered, and concentrated in vacuum. The residue was purified by silica gel column chromatography (eluent: Dichloromethane/MeOH - 10:1) to afford 210 mg (79%) of N-[2-(5-bromo-1,3- benzothiazol-2-yl)ethyl]-7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-amine as a brown solid. HPLC/MS m/z: 468.00 [M+H] + , Rt (B): 0.98 min.

Example 102.2: Carbonylation using the described conditions yielded 21 mg (11%) of ethyl 2-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl]amino}ethyl)-1 ,3- benzothiazole-5-carboxylate as an off-white solid. HPLC/MS m/z: 460.20 [M+H] + , Rt (S): 1.90 min. 1 H NMR (400 MHz, DMSO-cfe): 6 8.92-8.87 (m, 1 H), 8.47 (d, J = 1.7 Hz, 1 H), 8.23-8.12 (m, 3H), 8.01 (d, J = 5.7 Hz, 1 H), 7.96 (dd, J = 8.4, 1.6 Hz, 1 H), 7.88 (d, J = 8.5 Hz, 1 H), 7.04-6.98 (m, 1 H), 4.36 (q, J = 7.1 Hz, 2H), 3.99 (q, J = 6.4 Hz, 2H), 3.58 (t, J = 6.7 Hz, 2H), 2.69 (s, 3H), 1.36 (t, J = 7.1 Hz, 3H).

Example 103: Ethyl 7-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-8-oxo-1 ,2,3,4,7,8-hexahydro-2,7-naphthyridine-2-carboxylate

Example 103.1: 2H-[2,7]Naphthyridin-1-one (500.0 mg, 3.421 mmol), 2-(Boc- amino)ethyl bromide (920.0 mg, 4.105 mmol) and potassium carbonate (1.42 g, 10.264 mmol) were dissolved in dry DMF (13.3 mL). The suspension was heated to 80 °C and stirred for 4 h. After 1-2 h, the reaction mixture turned red. All volatiles were evaporated, and the crude product was purified by RP-flash chromatography to yield 691 mg (70 %) of tert-butyl N-[2-(1-oxo-1 ,2-dihydro-2,7-naphthyridin-2- yl)ethyl]carbamate as a red solid. HPLC/MS m/z: 289.9 [M+H] + , Rt (G): 1.07 min. Example 103.2: tert-Butyl N-[2-(1-oxo-1 ,2-dihydro-2,7-naphthyridin-2- yl)ethyl]carbamate (691.0 mg, 2.388 mmol) were hydrogenated in THF (10 mL) and acetic acid (2 mL) in presence of PtC^xFLO (130 mg) at room temperature for 6 h at 1 bar. The catalyst was filtered off and the filtrate was evaporated. The residue was used in the next step without further purification.

Example 103.3: To a solution of tert-butyl N-[2-(1-oxo-1 ,2,5,6,7,8-hexahydro-2,7- naphthyridin-2-yl)ethyl]carbamate (250.0 mg, 0.579 mmol) and dry pyridine (468 pl, 5.795 mmol) in dry dichloromethane (3.7 mL) was added ethyl chloroformate (61 pl, 0.637 mmol). The reaction mixture was stirred at room temperature for 1 h, concentrated in vacuum and the residue was purified by flash column chromatography to give 79 mg (37 %) of ethyl 7-(2-{[(tert- butoxy)carbonyl]amino}ethyl)-8-oxo-1 ,2,3,4,7,8-hexahydro-2,7-naphthyridine-2- carboxylate as a beige solid. HPLC/MS m/z: 265.9 [M+H-Boc] + , Rt (G): 1.38 min. Example 103.4: Ethyl 7-(2-{[(tert-butoxy)carbonyl]amino}ethyl)-8-oxo-1 ,2,3,4,7,8- hexahydro-2,7-naphthyridine-2-carboxylate (79.0 mg, 0.216 mmol) was dissolved in a solution of HCI in 1 ,4-dioxane (4.0 M, 3.8 mL) and stirred 15 min at room temperature. All volatiles were removed in vacuum and the resulting oil (75 mg) was used without further purification.

Example 103.5: 7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-2-ium-2olate (20.0 mg, 0.088 mmol) and 2-[7-(ethoxycarbonyl)-1-oxo-1 ,2,5,6,7,8-hexahydro-2,7- naphthyridin-2-yl]ethan-1-amine hydrochloride (33.2 mg, 0.110 mmol) were dissolved in dry dichloromethane (1 mL) under an argon atmosphere. N- Ethyldiisopropylamine for synthesis (101 pL, 0.594 mmol) and bromo- tripyrrolidinophosphonium-hexafluorphosphate (53.1 mg, 0.114 mmol) were added and the reaction mixture was stirred at room temperature for 3 d. All volatiles were removed under reduced pressure and the residue was directly purified by RP flash chromatography to afford 8 mg (19%) of ethyl 7-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl]amino}ethyl)-8-oxo-1 ,2,3,4,7,8-hexahydro-2,7-naphthyridine-2- carboxylate as a colorless solid. HPLC/MS m/z: 475.20 [M+H] + , Rt (N): 1.34 min. 1 H NMR (700 MHz, Methanol-d 4 ): 5 8.76-8.73 (m, 1 H), 8.23-8.20 (m, 1 H), 7.89 (d, J = 5.8 Hz, 1 H), 7.82-7.79 (m, 1 H), 7.32 (d, J = 6.8 Hz, 1 H), 7.00-6.97 (m, 1 H), 6.10 (d, J = 6.8 Hz, 1 H), 4.40 (s, 2H), 4.36 (t, J = 5.8 Hz, 2H), 4.20-4.15 (m, 2H), 3.96-3.92 (m, 2H), 3.71-3.59 (m, 2H), 2.69 (s, 3H), 2.66-2.62 (m, 2H), 1.33-1.26 (m, 3H).

The following compounds were prepared in a similar manner:

Example 104: Propyl 7-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-8-oxo-1 ,2,3,4,7,8-hexahydro-2,7-naphthyridine-2-carboxylate

17 mg colorless solid. HPLC/MS m/z: 488.8 [M] + , Rt (M): 1.78 min. 1 H NMR (700 MHz, Methanol-ck): 6 8.74-8.73 (m, 1 H), 8.20 (dd, J = 8.4, 1.6 Hz, 1 H), 7.90 (d, J =

5.8 Hz, 1 H), 7.80 (d, J = 8.5 Hz, 1 H), 7.32 (d, J = 6.9 Hz, 1 H), 6.98-6.96 (m, 1 H), 6.10 (d, J = 6.9 Hz, 1 H), 4.42-4.40 (m, 2H), 4.36 (t, J = 5.8 Hz, 2H), 4.08 (t, J = 6.6 Hz, 2H), 3.94 (t, J = 5.8 Hz, 2H), 3.67-3.61 (m, 2H), 2.69 (s, 3H), 2.64 (t, J = 5.7 Hz, 2H), 1.73-1.66 (m, 2H), 1.00-0.96 (m, 3H).

Example 105: Propan-2-yl 7-(2-{[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl]amino}ethyl)-8-oxo-1 ,2,3,4,7,8-hexahydro-2,7-naphthyridine-2-carboxylate

25 mg beige solid. HPLC/MS m/z: 488.9 [M] + , Rt (M): 1.77 min. 1 H NMR (400 MHz, DMSO-cfe): 6 8.84-8.81 (m, 1 H), 8.15 (dd, J = 8.5, 1.5 Hz, 1 H), 7.99 (t, J = 5.4 Hz, 1 H), 7.96 (d, J = 5.7 Hz, 1 H), 7.85 (d, J = 8.5 Hz, 1 H), 7.28 (d, J = 6.9 Hz, 1 H), 6.97 (d, J = 5.4 Hz, 1 H), 5.98 (d, J = 6.9 Hz, 1 H), 4.87-4.78 (m, 1 H), 4.24-4.18 (m, 4H), 3.78 (q, J = 5.6 Hz, 2H), 3.52 (t, J = 5.8 Hz, 2H), 2.69 (s, 3H), 2.57-2.52 (m, 2H), 1.21 (d, J = 6.2 Hz, 6H).

Example 106: Propan-2-yl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate

Example 106.1: To a solution of 2-bromopropane (865 mg, 7.03 mmol, 1.50 eq) in dimethyl formamide (10 mL) was added potassium carbonate (1.30 g, 9.38 mmol, 2.00 eq). The mixture was stirred at 60 °C for 0.5 h. To the mixture was added 2- (terf-butoxycarbonyl)-1,2,3,4-tetrahydroisoquinoline-7-carbo xylic acid (1.30 g, 4.69 mmol, 1.00 eq). The mixture was stirred at 60 °C for 3 h. The mixture was diluted with saturated ammonium chloride aqueous solution (20 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organic layers was washed with brine (3 x 20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 2-terf-butyl 7-isopropyl 3,4-dihydroisoquinoline-2,7(1/7)- dicarboxylate (1.40 g, 4.38 mmol, 93% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-cfe): 67.75-7.73 (m, 2H), 7.30 (d, J = 8.2 Hz, 1H), 5.12 (quin, J = 6.2 Hz, 1H), 4.56 (br s, 2H), 3.56 (t, J = 5.9 Hz, 2H), 2.84 (t, J = 5.9 Hz, 2H), 1.43 (s, 9H), 1.31 (d, J = 6.2 Hz, 6H).

Example 106.2: A solution of 2-terf-butyl 7-isopropyl 3,4-dihydroisoquinoline- 2,7(1 /-/)-dicarboxylate (0.600 g, 1.95 mmol, 1.00 eq) in hydrochloric acid/ethyl acetate (4 M, 10 mL) was stirred at 20 °C for 2 h. The mixture was concentrated under reduced pressure to afford isopropyl 1,2,3,4-tetrahydroisoquinoline-7- carboxylate (0.450 g, crude, hydrochloride) as a brown solid. 1 H NMR (400 MHz, Methanol-d 4 ): 67.92 (d, J = 7.9 Hz, 1H), 7.88 (s, 1 H), 7.38 (d, J = 7.9 Hz, 1H), 5.22 (td, J = 6.3, 12.5 Hz, 1H), 4.43 (s, 2H), 3.54 (t, J = 6.4 Hz, 2H), 3.19 (t, J = 6.4 Hz, 2H), 1.37 (d, J = 6.4 Hz, 6H).

Example 106.3: To a solution of isopropyl 1 ,2,3,4-tetrahydroisoquinoline-7- carboxylate (1.00 g, 3.91 mmol, 1.00 eq, hydrochloride) and N,N- diisopropylethylamine (2.53 g, 19.6 mmol, 5.00 eq) in dimethyl formamide (10 mL) were added potassium iodide (649 mg, 3.91 mmol, 1.00 eq) and tert-butyl /\/-(2- bromoethyl)carbamate (2.63 g, 11.7 mmol, 3.00 eq). The mixture was stirred at 90 °C for 12 h. The mixture was diluted with saturated ammonium chloride aqueous solution (20 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organic layer was washed with brine (3 x 20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reversed phase column chromatography (C18, 40 g; condition: water/acetonitrile = 1/0 to 0/1 , 0.1% ammonium hydroxide) to afford isopropyl 2-(2-((terf-butoxycarbonyl)amino)ethyl)-1,2,3,4-tetrahydrois oquinoline-7- carboxylate (0.600 g, 1.66 mmol, 42% yield) as a yellow solid. HPLC/MS m/z: 363.1 [M+H] + , Rt (V): 1.04 min. 1 H NMR (400 MHz, DMSO-d 6 ): 67.69 (br d, J = 7.9 Hz, 1H), 7.64 (s, 1 H), 7.24 (d, J = 7.9 Hz, 1H), 6.72 (br t, J = 5.0 Hz, 1 H), 5.11 (td, J = 6.2, 12.5 Hz, 1 H), 3.63 (s, 2H), 3.16-3.09 (m, 2H), 2.86 (br t, J = 5.3 Hz, 2H), 2.69 (br t, J = 5.7 Hz, 2H), 1.37 (s, 9H), 1 .31 (d, J = 6.2 Hz, 6H).

Example 106.4: A solution of ethyl 5-(2-((tert-butoxycarbonyl)amino)ethyl)-1-methyl- 4,5,6,7-tetrahydro-1/7-pyrrolo[3,2- c]pyridine-3-carboxylate (0.250 g, 711 umol, 1.00 eq) in hydrochloric acid/ethyl acetate (4 M, 5 mL) was stirred at 20 °C for 2 h. The mixture was concentrated under reduced pressure to afford isopropyl 2-(2- aminoethyl)-1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate (0.200 g, crude, hydrochloride) as a yellow solid. 1 H NMR (400 MHz, Methanol-d^: 67.94 (d, J = 8.1 Hz, 1 H), 7.90 (s, 1 H), 7.41 (d, J = 8.1 Hz, 1 H), 5.22 (td, J = 6.2, 12.5 Hz, 1 H), 3.75- 3.66 (m, 2H), 3.62 (br d, J = 6.7 Hz, 2H), 3.58-3.53 (m, 2H), 3.38-3.31 (m, 4H), 1.37 (d, J = 6.2 Hz, 6H).

Example 106.5: To a solution of isopropyl 2-(2-aminoethyl)-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (65.8 mg, 220 umol, 1.25 eq, hydrochloride), /V,/V-diisopropylethylamine (114 mg, 880 umol, 5.00 eq) in dichloromethane (5 mL) were added 7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinoline 2-oxide (40.0 mg, 176 umol, 1.00 eq) and bromotripyrrolidinophosphonium hexafluorophosphate (164 mg, 352 umol, 2.00 eq) at 20 °C. The mixture was stirred at 20 °C for 12 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (petroleum ether/ethyl acetate = 1/1), prep-HPLC (column: Phenomenex Gemini-NX C18 75*30mm*3um; mobile phase: [water(10 mM NH4HCO3)-ACN]; B%: 50%-80%, 8min) and lyophilized to afford isopropyl 2-(2-((7- (5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (30.02 mg, 63.03 umol, 35% yield, 99% purity) as a yellow solid. HPLC/MS m/z: 472.3 [M+H] + , Rt (V): 1.09 min. 1 H NMR (400 MHz, DMSO-cfe): 68.87 (s, 1 H), 8.14 (dd, J = 1.2, 8.5 Hz, 1 H), 7.97 (d, J = 5.7 Hz, 1 H), 7.91-7.80 (m, 2H), 7.71-7.64 (m, 2H), 7.24 (d, J = 7.8 Hz, 1 H), 6.94 (d, J = 5.7 Hz, 1 H), 5.11 (quin, J = 6.2 Hz, 1 H), 3.80-3.68 (m, 4H), 2.92-2.85 (m, 2H), 2.83-2.74 (m, 4H), 2.69 (s, 3H), 1 .30 (d, J = 6.2 Hz, 6H).

Example 107: Ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate Example 107.1: A mixture of 2-terf-butoxycarbonyl-3,4-dihydro-1/7-isoquinoline-7- carboxylic acid (900 mg, 3.25 mmol, 1.00 eq), iodoethane (607. mg, 3.89 mmol, 1.20 eq) and potassium carbonate (897 mg, 6.49 mmol, 2.00 eq) in acetonitrile (20 mL) was stirred at 60 °C for 12 h. After the reaction was completed, the mixture was filtered. The filtrate was concentrated to afford 2-terf-butyl 7-ethyl 3,4- dihydroisoquinoline-2,7(1/-/)- dicarboxylate (0.998 g, crude) as a yellow oil. 1 H NMR (400 MHz, CDCI3) 67.84 (d, J = 7.8 Hz, 1 H), 7.81 (s, 1 H), 7.21 (d, J = 7.8 Hz, 1 H), 4.62 (s, 2H), 4.38 (q, J = 7.2 Hz, 2H), 3.67 (t, J = 6.0 Hz, 2H), 2.89 (t, J = 6.0 Hz, 2H), 1.50 (s, 9H), 1.40 (t, J = 7.2 Hz, 3H).

Example 107.2: A mixture of 2-terf-butyl 7-ethyl 3,4-dihydroisoquinoline-2,7(1/7)- dicarboxylate (300 mg, 982 umol, 1.00 eq) in hydrochloric acid/ethyl acetate (4 M, 10 mL) was stirred at 25 °C for 0.5 h. After the reaction was completed, the mixture was concentrated to afford ethyl 1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate (230 mg, crude, hydrochloride) as a white solid. 1 H NMR (400 MHz, DMSO-de): 69.57 (br s, 2H), 7.91-7.77 (m, 2H), 7.37 (d, J = 8.0 Hz, 1 H), 4.36-4.28 (m, 4H), 3.41-3.39 (m, 2H), 3.08 (br t, J = 6.0 Hz, 2H), 1.35-1.29 (m, 3H). HPLC/MS m/z: 206.2 [M+H] + , Rt (U): 0.66 min.

Example 107.3: A mixture of ethyl 1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate (230 mg, 951 umol, 1.00 eq, hydrochloride), cesium carbonate (775 mg, 2.38 mmol, 2.50 eq) and terf-butyl /\/-(2-bromoethyl)carbamate (235 mg, 1.05 mmol, 1.10 eq) in acetonitrile (5 mL) was stirred at 60 °C for 12 h. After the reaction was completed, the mixture was filtered, and the filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 10/1 to 3/1) to afford ethyl 2-(2-((terf-butoxycarbonyl)amino)ethyl)-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (268 mg, crude) as a yellow oil. 1 H NMR (400 MHz, Chloroform-d): 67.88 (d, J = 8.0 Hz, 1 H), 7.77 (s, 1 H), 7.23 (d, J = 8.0 Hz, 1 H), 4.37 (q, J = 7.2 Hz, 2H), 4.10-3.68 (m, 2H), 3.58-3.40 (m, 2H), 3.23-2.87 (m, 6H), 1.45 (s, 9H), 1.41-1.38 (m, 3H).

Example 107.4: A mixture of ethyl 2-(2-((terf-butoxycarbonyl)amino)ethyl)-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (260 m g, 746umol, 1.00 eq) in hydrochloric acid/ethyl acetate (4 M, 10 mL) was stirred at 25 °C for 1 h. After the reaction was completed, the mixture was concentrated to afford ethyl 2-(2-aminoethyl)-1 , 2,3,4- tetrahydroisoquinoline-7-carboxylate (220 mg, crude, hydrochloride) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ): 68.45-8.25 (m, 3H), 7.89-7.79 (m, 2H), 7.41 (d, J = 8.0 Hz, 1 H), 4.86-4.41 (m, 2H), 4.39-4.18 (m, 4H), 3.92-3.59 (m, 2H), 3.46 (br s, 2H), 3.24-3.03 (m, 2H), 1.32 (t, J = 7.2 Hz, 3H).

Example 107.5: A mixture of ethyl 2-(2-aminoethyl)-1 ,2,3,4-tetrahydroisoquinoline-7- carboxylate (100 mg, 351 umol, 1.00 eq, hydrochloric acid salt), 5-methyl-3-(2- oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole (79.7 mg, 351umol, 1 eq), diisopropylethylamine (181 mg, 1.40 mmol, 4.00 eq) and bromo(tripyrrolidin-1- yl)phosphonium; hexafluorophosphate (295 mg, 632 umol, 1.80 eq) in dichloromethane (5 mL) was stirred at 25 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC(column: Waters Xbridge 150*25mm* 5um;mobile phase: [water(10mM NH4HCC>3)-ACN];B%: 56%- 86%,10min), prep-HPLC(column: Phenomenex luna C18 150*25mm* 10um;mobile phase: [water(0.225%FA)-ACN];B%: 15%-45%,11min) and lyophilized to afford ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-1 ,2,3,4- tetrahydroisoquinoline-7-carboxylate (33.77 mg, 66.3 umol, 18% yield, 99% purity, formate) as a yellow gum. 1 H NMR (400 MHz, DMSO-cfe): 58.87 (s, 1 H), 8.16-8.11 (m, 2H), 7.96 (d, J = 6.0 Hz, 1 H), 7.92-7.86 (m, 1 H), 7.84 (d, J = 8.8 Hz, 1 H), 7.74- 7.68 (m, 2H), 7.26 (d, J = 7.6 Hz, 1 H), 6.94 (d, J = 6.0 Hz, 1 H), 4.28 (q, J = 7.2 Hz, 2H), 3.82 (s, 2H), 3.79-3.72(m, 2H), 2.93-2.84 (m, 6H), 2.69 (s, 3H), 1.30 (t, J = 7.2 Hz, 3H). HPLC/MS m/z: 458.1 [M+H] + , Rt (T): 0.70 min.

Example 108: Methyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate

Example 108.1: Methyl 1 ,2,3,4-tetrahydroisoquinoline-7-carboxylate HCL (100.00 mg, 0.4392 mmol) , N-Boc-2-chloroethylamine (201.27 mg, 1.098 mmol), Potassium carbonate (212.44 mg, 1.5372 mmol) and potassium iodide (7.29 mg, 0.0439 mmol) were dissolved in dry DMF (2.20 mL, 0.2000 M ). The reaction mixture was stirred at 70 °C for 16 h. Purification by NP column chromatography (eluent: 0-20% MeOH in DCM) afforded methyl 2-[2-(tert-butoxycarbonylamino)ethyl]-3,4-dihydro-1 H- isoquinoline-7-carboxylate (75.7 mg, 52%, 0.2264 mmol). HPLC/MS m/z: 335.198 [M+H] + , Rt (AG): 1.10 min.

Example 108.2: Methyl 2-[2-(tert-butoxycarbonylamino)ethyl]-3,4-dihydro-1 H- isoquinoline-7-carboxylate (80.00 mg, 0.2392 mmol) was dissolved in dry 1 ,4- dioxane (1.20 mL, 0.2000 M). 4N hydrogen chloride in dioxane (0.60 mL, 2.3923 mmol) was added and the reaction mixture was stirred overnight at room temperature. Concentration of the solution under reduced pressure gave methyl 2- (2-aminoethyl)-3,4-dihydro-1 H-isoquinoline-7-carboxylate hydrochloride (61.22 mg, 95%, 0.2261 mmol) which was used without further purification, methyl 2-(2- aminoethyl)-3,4-dihydro-1 H-isoquinoline-7-carboxylate hydrochloride (59.58 mg, 0.2201 mmol) and 5-methyl-3-(2-oxidoisoquinolin-2-ium-7-yl)-1 ,2,4-oxadiazole (40.00 mg, 0.1760 mmol) were mixed in DCM (0.88 mL, 0.2000 M) at room temperature under argon. N,N-Diisopropylethylamine (0.15 mL, 0.8362 mmol) and Bromotri(pyrrolidino)phosphonium hexafluorophosphate (106.69 mg, 0.2289 mmol) were added successively and the reaction mixture was continued to stir at room temperature for 3 d. The crude product was subjected to RP column chromatography to give methyl 2-[2-[[7-(5-methyl-1 ,2,4-oxadiazol-3-yl)-1- isoquinolyl]amino]ethyl]-3,4-dihydro-1 H-isoquinoline-7-carboxylate (10.8 mg, 13%, 0.0233 mmol) as an amorphous solid. HPLC/MS m/z: 444.202 [M+H] + , Rt (AC): 0.99 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.83 (dt, J = 1.6, 0.7 Hz, 1 H), 8.20 (dd, J = 8.5, 1.6 Hz, 1 H), 7.92 (d, J = 5.9 Hz, 1 H), 7.82-7.75 (m, 3H), 7.25 (d, J = 8.0 Hz, 1 H), 6.97 (dd, J = 6.0, 0.9 Hz, 1 H), 3.87 (s, 3H), 3.86-3.82 (m, 4H), 3.03 (t, J = 6.0 Hz, 2H), 2.96 (t, J = 6.4 Hz, 2H), 2.93 (t, J = 6.0 Hz, 2H), 2.65 (s, 3H).

Example 109: Ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxylate

Using ethyl 1 H,2H,3H,4H-pyrrolo[1 ,2-a]pyrazine-7-carboxylate hydrochloride in a similar procedure to Example 108 afforded ethyl 2-(2-((7-(5-methyl-1 ,2,4-oxadiazol-

3-yl)isoquinolin-1-yl)amino)ethyl)-1,2,3,4-tetrahydropyrr olo[1,2-a]pyrazine-7- carboxylate (6.1 mg, 0.014mmol). HPLC/MS m/z: 447.213 [M+H] + , Rt (AJ): 2.15 min. 1 H N MR (600 MHz, Methanol-d 4 ): 5 8.84 (dt, J = 1.7, 0.8 Hz, 1 H), 8.22 (dd, J = 8.5, 1.6 Hz, 1 H), 7.93 (d, J = 5.9 Hz, 1 H), 7.82 (d, J = 8.5 Hz, 1 H), 7.29 (d, J = 1.8 Hz, 1 H), 6.99 (dd, J = 6.0, 1.0 Hz, 1 H), 6.26 (dt, J = 2.0, 1.1 Hz, 1 H), 4.24 (q, J = 7.1 Hz, 2H), 4.11 (dd, J = 6.4, 4.8 Hz, 2H), 3.88-3.74 (m, 4H), 3.10-3.01 (m, 2H), 2.96 (t, J = 6.4 Hz, 2H), 2.69 (s, 3H), 1.33 (t, J = 7.1 Hz, 3H).

Example 110: Ethyl 6-methyl-5-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4,5,6,7-tetrahydropyrazolo[1 ,5-a]pyrazine-2-carboxylate

Example 110.1: Propargyl amine (273.39 mg, 4.9635 mmol) and triethylamine (0.70 mL, 4.9635 mmol) were mixed with DCM (4.51 mL) and cooled to 0 °C. 2- Nitrobenzenesulfonyl chloride (1.00 g, 4.5122 mmol) was added in one portion, the reaction mixture was warmed to room temperature and stirred for 3 h. The reaction was diluted with DCM, washed twice with 1 M HCI and brine. The organic layer was dried over MgSO 4 and cone, in vacuo to give 2-nitro-N-prop-2-ynyl- benzenesulfonamide(1034 mg, 95%, 4.304 mmol) as an off-white powder. 1 H NMR (500 MHz, Chloroform-d): 5 8.23-8.18 (m, 1 H), 7.95-7.90 (m, 1 H), 7.78-7.73 (m, 2H), 5.70 (s, 1 H), 4.02 (dd, J = 6.3, 2.5 Hz, 2H), 1.97 (t, J = 2.5 Hz, 1 H).

Example 110.2: 2-Nitro-N-prop-2-ynyl-benzenesulfonamide (150.00 mg, 0.6244 mmol), triphenylphosphine (245.65 mg, 0.9366 mmol) and 1-chloro-2-propanol (88.54 mg, 0.9366 mmol) were dissolved in dry THF (3.12 mL). Di-tert- butylazodicarboxylate (215.65 mg, 0.9366 mmol) was added and stirred for 1 h.45. Further triphenylphosphine (245.65 mg, 0.9366 mmol), di-tert-butylazodicarboxylate (215.65 mg, 0.9366 mmol) and 1-chloro-2-propanol (88.54 mg, 0.9366 mmol) were added. Stirred at room temperature for 1 h 30 min. The solvent was removed in vacuo. Purification by NP column chromatography (eluent: 10-40% EtOAc in cyclohexane) afforded N-(2-chloro-1-methyl-ethyl)-2-nitro-N-prop-2-ynyl- benzenesulfonamide (206 mg, 104%, 0.6503 mmol) as a white gum. HPLC/MS m/z: 339.017 [M+Na] + , Rt (AG): 1.27 min.

Example 110.3: N-(2-Chloro-1-methyl-ethyl)-2-nitro-N-prop-2-ynyl- benzenesulfonamide (186.00 mg, 0.5872 mmol) was dissolved in dry toluene (5.87 mL). Ethyldiazoacetate (0.52 mL, 0.6459 mmol) was added, and the reaction mixture was stirred at 120 °C overnight. The solvent was removed in vacuo.

Purification by NP column chromatography (eluent: 10-50% EtOAc in cyclohexane) afforded ethyl 6-methyl-5-(2-nitrophenyl)sulfonyl-6,7-dihydro-4H-pyrazolo[1 ,5- a]pyrazine-2-carboxylate (120 mg, 52%, 0.3043 mmol) as an orange oil. HPLC/MS m/z: 395.140 [M+H] + , Rt (AG): 1.41 min.

Example 110.4: Ethyl 6-methyl-5-(2-nitrophenyl)sulfonyl-6,7-dihydro-4H- pyrazolo[1,5-a]pyrazine-2-carboxylate (120.00 mg, 0.3043 mmol) and cesium carbonate (198.27 mg, 0.6085 mmol) were dissolved in dry MeCN (1.52 mL). Benzenethiol (62.49 uL, 0.6085 mmol) was added and the reaction mixture was stirred at room temperature for 2 h. The solvent was removed in vacuo. Purification by NP column chromatography (Eluent: 0-15% MeOH in DCM) afforded ethyl 6- methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine-2-carboxyla te (43 mg, 68%, 0.2055 mmol). HPLC/MS m/z: 210.117 [M+H] + , Rt (Al): 0.27 min.

Example 110.5: Ethyl 6-methyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine-2- carboxylate (40.00 mg, 0.1912 mmol) was dissolved in CHCh (0.96 mL). 1- Nitroethene (1 M in xylene) (0.19 mL, 0.1912 mmol) was added, and the reaction mixture was cooled to 0 °C, stirred at 0 °C for 30 min. Further 0.5 eq of 1- nitroethene was added, stirred for an extra 10 min. The mixture was concentrated in vacuo. Purification by NP column chromatography (Eluent: 50-100% EtOAc in cyclohexane) afforded ethyl 6-methyl-5-(2-nitroethyl)-6,7-dihydro-4H-pyrazolo[1,5- a]pyrazine-2-carboxylate (36 mg, 67%, 0.1275 mmol) as a pale orange oil.

HPLC/MS m/z: 283.140 [M+H] + , Rt (Al): 1.11 min.

Example 110.6: Ethyl 6-methyl-5-(2-nitroethyl)-6,7-dihydro-4H-pyrazolo[1,5- a]pyrazine-2-carboxylate (36.00 mg, 0.1275 mmol), zinc (83.38 mg, 1.2752 mmol) and ammonium chloride (68.21 mg, 1.2752 mmol) were suspended in a mixture of THF (0.77 mL, 0.1300 M ) / water (0.10 mL) I MeOH (0.10 mL). The reaction mixture was stirred at room temperature for 2 h 20 min. The mixture was filtered on celite, rinsed with a 1 :1 mixture of DCM:MeOH. The filtrate was passed through a 2 g NH2 ion exchange column and rinsed with a 1:1 mixture of DCM:MeOH. The filtrate was concentrated to give ethyl 5-(2-aminoethyl)-6-methyl-6,7-dihydro-4H-pyrazolo[1 ,5- a]pyrazine-2-carboxylate (30 mg, 93%, 0.1189 mmol) as a pale- yellow oil. HPLC/MS m/z: 275.148 [M+H] + , Rt (Al): 0.69 min.

Example 110.7: Ethyl 5-(2-aminoethyl)-6-methyl-6,7-dihydro-4H-pyrazolo[1 ,5- a]pyrazine-2-carboxylate (30.00 mg, 0.1189 mmol), 5-methyl-3-(2-oxidoisoquinolin- 2-ium-7-yl)-1 ,2,4-oxadiazole (27.02 mg, 0.1189 mmol) and bromotri(pyrrolidino)phosphonium hexafluorophosphate (83.14 mg, 0.1784 mmol) were dissolved in dry DCM (0.79 mL) in a 0.5-2 mL microwave vial. DIPEA (95.27 uL, 0.5469 mmol) was added and the reaction mixture was irradiated at 60 °C for 1 h. The solvent was removed in vacuo. Purification by reverse phase flash chromatography (eluent: 30-80% MeOH/H 2 O + 0.1% formic acid) followed by ion exchange SCX-2 chromatography eluting with 2M NH3 in MeOH. Further purification by Biotage KP-NH silica column chromatography (eluent: 30-80% EtOAc in cyclohexane) afforded ethyl 6-methyl-5-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3- yl)isoquinolin-1-yl)amino)ethyl)-4,5,6,7-tetrahydropyrazolo[ 1,5-a]pyrazine-2- carboxylate (20 mg, 35%, 0.0420 mmol). HPLC/MS m/z: 462.226 [M+H] + , Rt (AJ): 1.93 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.80-8.76 (m, 1 H), 8.20 (dd, J = 8.5, 1.6 Hz, 1 H), 7.91 (d, J = 5.9 Hz, 1 H), 7.79 (d, J = 8.5 Hz, 1 H), 6.97 (dd, J = 6.0, 0.9 Hz, 1 H), 6.58 (s, 1 H), 4.33 (q, J = 7.1 Hz, 2H), 4.28 (dd, J = 12.8, 4.4 Hz, 1 H), 4.09 (d, J = 15.7 Hz, 2H), 3.98 (d, J = 15.9 Hz, 1 H), 3.94 (dd, J = 12.7, 6.4 Hz, 1 H), 3.81- 3.71 (m, 2H), 3.47-3.40 (m, 1 H), 3.04-2.94 (m, 2H), 2.67 (s, 3H), 1.36 (t, J = 7.1 Hz, 3H), 1.22 (d, J = 6.7 Hz, 3H).

Example 111 : Ethyl 4-methyl-5-(2-((7-(5-methyl-1 ,2,4-oxadiazol-3-yl)isoquinolin-1- yl)amino)ethyl)-4,5,6,7-tetrahydropyrazolo[1 ,5-a]pyrazine-2-carboxylate

Using 1-Methyl-prop-2-ynylamine hydrochloride and 2-chloroethanol in a similar procedure to Example 110 afforded ethyl 4-methyl-5-(2-((7-(5-methyl-1 ,2,4- oxadiazol-3-yl)isoquinolin-1-yl)amino)ethyl)-4,5,6,7-tetrahy dropyrazolo[1 ,5- a]pyrazine-2-carboxylate (3 mg, 0.0062 mmol). HPLC/MS m/z: 462.225 [M+H] + , Rt (AJ): 2.02 min. 1 H NMR (600 MHz, Methanol-d 4 ): 5 8.79 (dt, J = 1.6, 0.7 Hz, 1 H), 8.20 (dd, J = 8.5, 1.6 Hz, 1H), 7.92 (d, J = 5.9 Hz, 1 H), 7.80 (d, J = 8.5 Hz, 1H), 6.97 (dd, J = 6.0, 0.9 Hz, 1 H), 6.60 (d, J = 0.9 Hz, 1H), 4.33 (q, J = 7.1 Hz, 2H), 4.27-4.18 (m, 2H), 3.98 (q, J = 6.6 Hz, 1 H), 3.86-3.79 (m, 1 H), 3.77-3.71 (m, 1 H), 3.50 (dt, J = 12.9, 4.7 Hz, 1H), 3.21-3.14 (m, 1H), 3.09-3.02 (m, 1 H), 2.90-2.84 (m, 1 H), 2.66 (s, 3H), 1.45 (d, J = Q.Q Hz, 3H), 1.36 (t, J= 7.1 Hz, 3H).

Example 112: Ethyl 5-methyl-2-(2-(3-(5-methyl-1,2,4-oxadiazol-3- yl)benzamido)ethyl)-3H-imidazo[4,5-b]pyridine-6-carboxylate

Example 112.1: To methyl 3-aminopropanoate hydrochloride (400.00 mg, 2.8657 mmol), 3-(5-methyl-1,2,4-oxadiazol-3-yl)benzoic acid (585.13 mg, 2.8657 mmol) in DMF (14.33 mL) was added DIPEA (2.00 mL, 11.463 mmol) followed by HATU (1011.31 mg, 4.2986 mmol). The reaction was stirred for 18 h at room temperature. The mixture was diluted with EtOAc (75 mL) and washed with water (100 mL). The water was extracted with fresh EtOAc (75 mL). The organics were combined and washed with aqeous saturated bicarbonate solution (100 mL), and brine (100 mL) before drying over MgSO4. After filtering and concentrating in vacuo the methyl 3- [[3-(5-methyl-1,2,4-oxadiazol-3-yl)benzoyl]amino]propanoate (705 mg, 85%, 2.437 mmol) was isolated as an off-white powder. HPLC/MS m/z: 290.125 [M+H] + , Rt (AA): 1.20 min.

Example 112.2: To methyl 3-[[3-(5-methyl-1 ,2,4-oxadiazol-3- yl)benzoyl]amino]propanoate (500.00 mg, 1.7284 mmol) in THF (8.64 mL) was added water (8.64 mL) followed by lithium hydroxide hydrate (290.09 mg, 6.9135 mmol). After stirring for 2.5 h water (20 mL) was added and the THF removed in vacuo. The solution was acidified with 1M citric acid solution and extracted with EtOAc (3 x 25ml). The organics were combined, washed with brine, and dried over MgSO4to afford 3-[[3-(5-methyl-1,2,4-oxadiazol-3-yl)benzoyl]amino]propanoic acid (423 mg, 89%, 1.5367 mmol) as a colorless solid. HPLC/MS m/z: 187.057 [M+H] + , Rt (AA): 1.09 min.

Example 112.3: To ethyl 6-amino-2-methyl-5-nitro-pyridine-3-carboxylate (41.00 mg, 0.1821 mmol) in THF (3.00 mL) and EtOH (1.00 mL) under nitrogen was very carefully added 10% Pd on carbon (3.87 mg, 0.0364 mmol). A hydrogen atmosphere was introduced and stirred at rt for 18 h. Filtered through Celite which in turn was washed with fresh THF and EtOH and organics combined. Concentration in vacuo gave ethyl 5,6-diamino-2-methyl-pyridine-3-carboxylate (24 mg, 68%, 0.1229 mmol) as a light tan coloured powder that was kept in the freezer. HPLC/MS m/z: 196.106 [M+H] + , Rt (AA): 0.33 min.

Example 112.4: To 3-[[3-(5-methyl-1,2,4-oxadiazol-3-yl)benzoyl]amino]propanoic acid (35.00 mg, 0.1272 mmol) in dry Toluene (0.50 mL, 0.2500 M) was added thionyl chloride (0.04 mL, 0.5086 mmol). The suspension was heated to 80 °C for 2 h. Volatiles were removed in vacuo and the crude 3-[[3-(5-methyl-1,2,4-oxadiazol-3- yl)benzoyl]amino]propanoyl chloride taken straight on to next step without further purification. Ethyl 5,6-diamino-2-methyl-pyridine-3-carboxylate (24.00 mg, 0.1229 mmol) was suspended in anhydrous toluene (0.5 mL) and 3-[[3-(5-methyl-1,2,4- oxadiazol-3-yl)benzoyl]amino]propanoyl chloride (36.11 mg, 0.1229 mmol) in anhydrous toluene (0.5 mL) was added. The suspension was stirred for 5 min before an extra 1 mL anhydrous toluene was added to try and dissolve material. Heated to 85 °C for 15 min before 1 drop of cone. HCI was added. Mixture was heated at this temperature for 19 h. Volatiles were removed in vacuo. The crude was dissolved in glacial acetic acid (2 mL) and heated at 95 °C for 23 h and then 150 °C for 3 h in a microwave. The volatiles were removed in vacuo and the crude purified by reverse phase column chromatography (eluent: 20-90% MeOH/H2O + 0.1% formic acid) to afford ethyl 5-methyl-2-[2-[[3-(5-methyl-1 ,2,4-oxadiazol-3-yl)benzoyl]amino]ethyl]- 3H-imidazo[4,5-b]pyridine-6-carboxylate (4.4 mg, 8%, 0.0101 mmol) as a tan powder. HPLC/MS m/z: 435.174 [M+H] + , Rt (AC): 1.26 min. 1 H NMR (500 MHz, DMSO-cfe): 6 13.07 (s, 1H), 8.91 (s, 1H), 8.45 (t, J = 1.7 Hz, 1 H), 8.31 (s, 1H), 8.12 (dt, J = 7.7, 1.4 Hz, 1 H), 8.01 (dt, J = 7.9, 1.4 Hz, 1 H), 7.65 (t, J = 7.8 Hz, 1 H), 4.31 (q, J = 7.1 Hz, 2H), 3.76 (td, J = 7.1, 5.5 Hz, 2H), 3.22-3.08 (m, 2H), 2.76 (s, 3H), 2.68 (s, 3H), 1.34 (t, J = 7.1 Hz, 3H).

Example 113: Testing compounds of the present invention for inhibitory activities against HSET - HSET ADP-Glo Assay S - 3 M ATP

Reagents: (+4 °C Storage) - Paclitaxel Prod.No.TXD.01 2 mM in DMSO (from Universal Biologicals Cambridge).

- Tubulin Protein (Pre-formed Microtubules): Bovine Brain Prod.No.MT001-XL Lot.025 10 mg/mL (from Universal Biologicals Cambridge).

Reagents: (-80 °C Storage)

- MT001-XL - Tubulin Protein (Pre-formed Microtubules): Bovine Brain - reconstituted in buffer - 15 mM PIPES pH7, 1 mM MgCI2, 20 pM paclitaxel (aliquots at 10 mg/mL) from Universal Biologicals Cambridge.

- HSET full length protein - Current batch is FL HSET Prepl (SEQ-000096_002- 01_01)

- Buffer is 20 mM Hepes pH 7.5, 200 mM NaCI, 2 mM TCEP, 5% glycerol (5 pL aliquots at 10.2 pM)

Reagents: (-20°C Storage)

- ADP-Glo Kinase Assay kit (Promega Prod.No.V9102) 10,000 assay points

- ADP-Glo reagent (50 mL), Kinase Detection Reagent (100 mL), and Ultrapure ATP (10 mM) aliquoted

Buffer Stocks (filtered and stored at rt for up to 1 month)

- HEPES acid, 4-(2-Hydroxyethyl)piperazine-1 -ethanesulfonic acid MW:238, 238.3 mg/mL = 1M

- 7149 mg/30 mL = 1M (pH to 6.8 with 5 M NaOH)

- PIPES, 1,4-Piperazinediethanesulfonic acid MW: 302.4, 30.24 mg/mL = 100 mM;

907.2 mg/30 mL = 100 mM (pH to 7 with 5 M NaOH, white cloudy suspension until the pH changes)

- EGTA, Ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid MW: 380.35, 38.035 mg/mL = 100 mM

- 1901.75 mg/50 mL = 100 mM (pH to ~7 with 5 M NaOH) - takes a long time to get into solution and for pH to stabilize

- Triton-X-100, MW: 625, 62.5 mg/mL = 100 mM (6.25% w/v) (viscous, pipette Triton X-100 with a Gilson using a trimmed pipette tip)

ECHO Protocol - Create an ECHO intermediate plate by adding 24.5 pL DMSO to columns 1 & 2, and 40 pL DMSO to columns 23 & 24 of an ECHO 384PP plate

- Add 100 nL of compound/DMSO per well in 384-well Proxi-Plate Plus (white) - Perkin Elmer Cat# 6008289 using ECHO dose response protocol 100 nL normal to proxi_8pt_200 uM.

Assay buffer (Keep on ice): HEPES pH 6.8, MgCI 2 , EGTA, Triton X-100, DTT, HPLC H 2 O

Microtubule Working Solution (3.2):

- 1 ml PM buffer: PIPES pH 7.0, MgCI 2 , HPLC H 2 O, Paclitaxel, mix well, store at rt

- Then add 26.4 pL of 10 mg/ml microtubules to724 pL of the above PM buffer to make 750 pL of microtubule working solution @ 350 pg/mL microtubules (store at rt)

Stock HSET enzyme solution (3.1) - keep on ice

- Add 1.33 pL 10.2 pM HSET Prepl (SEQ-000096_002-01_01) to 1358 pL assay buffer to make a 10 nM stock for a 5 nM final assay concentration (1/1020 dilution).

HSET/Microtubule working solution (3.3)

- Mix solutions 3.1 and 3.2 in the ratio of 2.5:1 (1214.3 pL 3.1 + 485.7 pL 3.2) keep at rt for 15 min

- 3.2 is diluted 3.5-fold, 3.1 is diluted 1.4-fold

BLANK solution is 357 pL 2XAB + 143 pL microtubule working solution 3.2.

(Same proportions as HSET/Microtubule working solution (3.3))

ATP Working solution is 1 pL 10 mM UltraPure ATP (Promega kit) + 999 pL ddH 2 O gives 10 pM ATP for a 3 pM final assay concentration, stored on ice.

Assay procedure in PROXI PLATE 384 PLUS WHITE (Perkin Elmer) plates (Remove 2.4 mL ADP Gio and 4.5 mL Kinase detection reagent from freezer to warm up to rt)

- Add 3.5 pL BLANK solution to assay plate (column 12)

- Add 3.5 pL HSET/Microtubule solution (3.3) (columns 1-11 & 13-24)

- Centrifuge at 1000 rpm for 1 min - (pre-incubate enzyme and compound for 10 min)

Add 1.5 pL of 10 pM ATP to start reaction gives a final [HSET] of 5 nM, [ATP] of 3 pM and [microtubule] of 70 pg/mL, centrifuge at 1000 rpm for 1 min and incubated at rt for 80 min.

After the 80-minute incubation, stop reaction by adding 5 pL ADP-Glo reagent to all wells. Centrifuge for 1 min at 1000 rpm, leave for 40 min at rt.

In the dark/away from direct light, add 10 pL Kinase Detection Reagent (KDR) to all wells, Seal plate with a Topseal (Perkin Elmer Cat# 6050185) and centrifuge as above, leave for 40 min at rt covered in foil.

Read luminescence on New Envision using Protocol: US luminescence 384.

Results:

Example 114: Injection vials

A solution of 100 g of a compound of the present invention and 5 g of disodium hydrogenphosphate in 3 L of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, filtered under sterile conditions, transferred into injection vials, lyophilised under sterile conditions and sealed under sterile conditions. Each injection vial contains 5 mg of a compound of the present invention.

Example 115: Solution

A solution is prepared from 1 g of a compound of the present invention, 9.38 g of NaH2PC>4 . 2 H2O, 28.48 g of Na2HPC>4 . 12 H2O and 0.1 g of benzalkonium chloride in 940 mL of bidistilled water. The pH is adjusted to 6.8, and the solution is made up to 1 L and sterilised by irradiation.

Example 116: Ampoules

A solution of 1 kg of a compound of the present invention in 60 L of bidistilled water is filtered under sterile conditions, transferred into ampoules, lyophilised under sterile conditions and sealed under sterile conditions. Each ampoule contains 10 mg of a compound of the present invention.

References

Basto, R., Brunk, K., Vinadogrova, T., Peel, N., Franz, A., Khodjakov, A., and Raff, J.W. (2008). Centrosome amplification can initiate tumorigenesis in flies. Cell 133, 1032-1042.

Bender, A., Young, D.W., Jenkins, J.L., Serrano, M., Mikhailov, D., Clemons, P.A., and Davies, J.W. (2007). Chemogenomic data analysis: prediction of small-molecule targets and the advent of biological fingerprint. Comb. Chem. High Throughput Screen. 10, 719-731.

Bickerton, G.R., Paolini, G.V., Besnard, J., Muresan, S., and Hopkins, A.L. (2012). Quantifying the chemical beauty of drugs. Nat. Chem. 4, 90-98. Boveri, T. (2008). Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. J. Cell Sci. 121 (Suppl 1 ), 1-84.

Cai, S., Weaver, L.N., Ems-McClung, S.C., and Walczak, C.E. (2010). Proper organization of microtubule minus ends is needed for midzone stability and cytokinesis. Curr. Biol. 20, 880-885.

Cai, S., Weaver, L.N., Ems-McClung, S.C., Walczak, C.E. (2009) Kinesin-14 family proteins HSET/XCTK2 control spindle length by cross-linking and sliding microtubules. Molecular Biology of the Cell 20(5), pp. 1348-1359

Chan, J.Y. (2011). A clinical overview of centrosome amplification in human cancers. Int. J. Biol. Sci. 7, 1122-1144.

De, S., Cipriano, R., Jackson, M.W., Stark, G.R.(2009) Overexpression of kinesins mediates docetaxel resistance in breast cancer cells. Cancer Research 69(20), pp. 8035-8042

Drosopoulos K., Tang C., Chao W. C., Linardopoulos S. (2014) APC/C is an essential regulator of centrosome clustering. Nat Commun. 2014, 5: 3686

Ganem, N.J., and Compton, D.A. (2004). The Kinl kinesin Kif2a is required for bipolar spindle assembly through a functional relationship with MCAK. J. Cell Biol. 166, 473-478.

Ganem, N.J., Godinho, S.A., and Pellman, D. (2009). A mechanism linking extra centrosomes to chromosomal instability. Nature 460, 278-282.

Gatlin, J.C., and Bloom, K. (2010). Microtubule motors in eukaryotic spindle assembly and maintenance. Semin. Cell Dev. Biol. 21 , 248-254.

Gaulton, A., Bellis, L.J., Bento, A.P., Chambers, J., Davies, M., Hersey, A., Light, Y., McGIinchey, S., Michalovich, D., Al-Lazikani, B., and Overington, J.P. (2012). ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 40(Database issue), D1100-D1107.

Gordon, M.B., Howard, L., and Compton, D.A. (2001). Chromosome movement in mitosis requires microtubule anchorage at spindle poles. J. Cell Biol. 152, 425-434.

Goshima, G., Nedelec, F., and Vale, R.D. (2005). Mechanisms for focusing mitotic spindle poles by minus end-directed motor proteins. J. Cell Biol. 171 , 229-240.

Grinberg-Rashi, H., Ofek, E., Perelman, M., Skarda, J., Yaron, P., Hajduch, M., Jacob-Hirsch, J., Amariglio, N., Krupsky, M., Simansky, D.A., et al. (2009). The expression of three genes in primary non-small cell lung cancer is associated with metastatic spread to the brain. Clin. Cancer Res. 15, 1755-1761.

Hall, I.M., Noma, K., and Grewal, S.l. (2003). RNA interference machinery regulates chromosome dynamics during mitosis and meiosis in fission yeast. Proc. Natl. Acad. Sci. USA 100, 193-198.

Holdgate, G.A., and Ward, W.H.J. (2005). Measurements of binding thermodynamics in drug discovery. Drug Discov. Today 10, 1543-1550.

Huszar, D., Theoclitou, M.E., Skolnik, J., and Herbst, R. (2009). Kinesin motor proteins as targets for cancer therapy. Cancer Metastasis Rev. 28, 197-208.

Jordan, M.A., and Wilson, L. (2004). Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 4, 253-265.

Klabunde, T. (2007). Chemogenomic approaches to drug discovery: similar receptors bind similar ligands. Br. J. Pharmacol. 152, 5-7.

Kleylein-Sohn, J., Pollinger, B., Ohmer, M., Hofmann, F., Nigg, E.A., Hemmings, B.A., and Wartmann, M. (2012). Acentrosomal spindle organization renders cancer cells dependent on the kinesin HSET. J. Cell Sci. 125, 5391-5402. Kraljevic Pavelic, S., Sedic, M., Bosnjak, H., Spaventi, S., and Pavelic, K. (2011) Metastasis: new perspectives on an old problem. Mol. Cancer 10, 22.

Kramer, A., Maier, B., and Bartek, J. (2011). Centrosome clustering and chromosomal (in)stability: a matter of life and death. Mol. Oncol. 5, 324-335.

Kwon, M., Godinho, S.A., Chandhok, N.S., Ganem, N.J., Azioune, A., Thery, M., and Pellman, D. (2008). Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 22, 2189-2203.

Li, Y., Lu, W., Chen, D., Boohaker, R. J., Zhai, L., Padmalayam, I., Wennerberg, K., Xu, B., Zhang, W. 2015) KIFC1 is a novel potential therapeutic target for breast cancer. Cancer Biology and Therapy 16(9), pp. 1316-1322

Lipinski, C.A., Lombardo, F., Dominy, B.W., and Feeney, P.J. (2001). Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 46, 3-26.

Luo, L., Carson, J.D., Dhanak, D., Jackson, J.R., Huang, P.S., Lee, Y., Sakowicz, R., and Copeland, R.A. (2004). Mechanism of inhibition of human KSP by monastrol: insights from kinetic analysis and the effect of ionic strength on KSP inhibition. Biochemistry 43, 15258-15266.

Mahoney, N.M., Goshima, G., Douglass, A.D., and Vale, R.D. (2006). Making microtubules and mitotic spindles in cells without functional centrosomes. Curr. Biol. 16, 564-569.

Marx, A., Hoenger, A., and Mandelkow, E. (2009). Structures of kinesin motor proteins. Cell Motil. Cytoskeleton 66, 958-966.

Mayer, T.U., Kapoor, T.M., Haggarty, S.J., King, R.W., Schreiber, S.L., and Mitchison, T.J. (1999). Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science 286, 971-974. Meunier, S., and Vernos, I. (2012). Microtubule assembly during mitosis - from distinct origins to distinct functions? J. Cell Sci. 125, 2805-2814.

Mountain, V., Simerly, C., Howard, L., Ando, A., Schatten, G., and Compton, D.A. (1999). The kinesin-related protein, HSET, opposes the activity of Eg5 and crosslinks microtubules in the mammalian mitotic spindle. J. Cell Biol. 147, 351-366.

Myers and Collins, 2016: Myers S., Collins I. (2016). Recent findings and future directions for interpolar mitotic kinesin inhibitors in cancer therapy. Future Med Chem. 8: 463-89

Neres, J., Wilson, D.J., Celia, L., Beck, B.J., and Aldrich, C.C. (2008). Aryl acid adenylating enzymes involved in siderophore biosynthesis: fluorescence polarization assay, ligand specificity, and discovery of non-nucleoside inhibitors via high- throughput screening. Biochemistry 47, 11735-11749.

Niesen, F.H., Berglund, H., and Vedadi, M. (2007). The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability. Nat. Protoc. 2, 2212-2221.

Nigg, E.A. (2002). Centrosome aberrations: cause or consequence of cancer progression? Nat. Rev. Cancer 2, 815-825.

Ogden, A., Rida, P.C., and Aneja, R. (2012). Let’s huddle to prevent a muddle: centrosome declustering as an attractive anticancer strategy. Cell Death Differ. 19, 1255-1267.

Ogden, A., Rida, P.C., and Aneja, R. (2013). Heading off with the herd: how cancer cells might maneuver supernumerary centrosomes for directional migration. Cancer Metastasis Rev. 32, 269-287.

Rath, O., Kozielski, F. (2012) Kinesins and cancer. Nature Reviews Cancer 12(8), pp. 527-539 Pannu, V., Rida, P.C.G., Ogden, A., Turaga, R. C., Donthamsetty, S, Bowen N. J., Rudd K, Gupta M. V., Reid M. D., Cantuaria G., Walczak, C.E., Aneja, R. (2015) HSET overexpression fuels tumor progression via centrosome clusteringindependent mechanisms in breast cancer patients. Oncotarget. 6: 6076-6091

Patel N. E., Weekes D., Drosopoulos K., Gazinska P., Noel E., Rashid M., Mirza H., Quist J., Braso-Maristany F., Mathew S., Ferro R., Mendes Pereira A., Prince C., Noor F., Francesch-Domenech E., Marlow R., de Rinaldis E., Grigoriadis A., Linardopoulos S., Marra P., Tutt A. N. J. (2018). Integrated genomics and functional validation identifys malignant cell specific dependencies in triple negative breast cancer. Nat. Commun. 9: 1044

Pawar, S., Donthamsetty, S., Pannu, V., Rida, P., Ogden, A., Bowen, N., Osan, R., Cantuaria, G., Aneja, R. (2014) KIFCI, a novel putative prognostic biomarker for ovarian adenocarcinomas: Delineating protein interaction networks and signaling circuitries. Journal of Ovarian Research 7(1), 53

Sharp, D.J., Rogers, G.C., and Scholey, J.M. (2000). Microtubule motors in mitosis. Nature 407, 41-47.

Tillement, V., Remy, M.H., Raynaud-Messina, B., Mazzolini, L., Haren, L., and Merdes, A. (2009). Spindle assembly defects leading to the formation of a monopolar mitotic apparatus. Biol. Cell 101 , 1-11.

Vichai, V., and Kirtikara, K. (2006). Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 1, 1112-1116.

Walczak, C.E., Verma, S., Mitchison, T.J. (1997) XCTK2: A kinesin-related protein that promotes mitotic spindle assembly in Xenopus laevis egg extracts. Journal of Cell Biology 136(4), pp. 859-870

Watts C. A., Richards F. M., Bender A., Bond P. J., Korb O., Kern O., Riddick M., Owen P., Myers R. M., Raff J., Gergely F., Jodrell D. I. (2013) Design, synthesis, and biological evaluation of an allosteric inhibitor of HSET that targets cancer cells with supernumerary centrosomes. Chemistry & Biology 20: 1399-1410 Wu J., Mikule K., Wang W., Su N., Petteruti P., Gharahdaghi F., Code E., Zhu X., Jacques K., Lai Z., Yang B., Lamb M.L. (2013) Discovery and mechanistic study of a small molecule inhibitor for motor protein KIFC1. ACS Chem Biol. 8: 2201-2208 Zhang et al., 2016: Zhang W., Zhai L., Wang Y., Boohaker R. J., Lu W., Gupta V. V., Padmalayam I., Bostwick R. J., White E. L., Ross L. J., Maddry J., Ananthan S. Discovery of a novel inhibitor of kinesin-like protein KIFC1. Biochem J. 473: 1027- 1035

Yang, Z., Loncarek, J., Khodjakov, A., and Rieder, C.L. (2008). Extra centrosomes and/or chromosomes prolong mitosis in human cells. Nat. Cell Biol. 10, 748-751.

Zyss, D., and Gergely, F. (2009). Centrosome function in cancer: guilty or innocent?

Trends Cell Biol. 19, 334-346.