Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
6,7-DIHYDRO-4H-PYRAZOLO[1,5-A]PYRAZINE INDOLE-2-CARBOXAMIDES ACTIVE AGAINST THE HEPATITIS B VIRUS (HBV)
Document Type and Number:
WIPO Patent Application WO/2020/089455
Kind Code:
A1
Abstract:
The present invention relates generally to antiviral agents. Specifically, the present invention relates to compounds of formula I which can inhibit the protein(s) encoded by hepatitis B virus (HBV) or interfere with the function of the HBV replication cycle, compositions comprising such compounds, methods for inhibiting HBV viral replication, methods for treating or preventing HBV infection, and processes and intermediates for making the compounds.

Inventors:
DONALD ALASTAIR (DE)
URBAN ANDREAS (DE)
BONSMANN SUSANNE (DE)
SPRINGER JASPER (NL)
Application Number:
PCT/EP2019/079969
Publication Date:
May 07, 2020
Filing Date:
November 01, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AICURIS GMBH & CO KG (DE)
International Classes:
A61K31/4985; C07D487/04; A61P31/12; C07D519/00
Domestic Patent References:
WO2018011160A12018-01-18
WO2018011162A12018-01-18
WO2017198744A12017-11-23
WO2016113273A12016-07-21
WO2013156608A12013-10-24
WO2012170098A22012-12-13
WO2011130163A12011-10-20
WO2006033995A22006-03-30
WO2016113273A12016-07-21
WO2017198744A12017-11-23
WO2018011162A12018-01-18
WO2018011160A12018-01-18
WO2018011163A12018-01-18
WO1999054326A11999-10-28
WO2000058302A12000-10-05
WO2001045712A12001-06-28
WO2001006840A12001-02-01
WO2014184328A12014-11-20
WO2015132276A12015-09-11
WO2016146598A12016-09-22
WO2015144093A12015-10-01
WO2013102655A12013-07-11
WO2017011552A12017-01-19
WO2017076286A12017-05-11
WO2016023877A12016-02-18
WO2016177655A12016-11-10
WO2017013046A12017-01-26
WO2013006394A12013-01-10
WO2013096744A12013-06-27
WO2014165128A22014-10-09
WO2014184365A12014-11-20
WO2015109130A12015-07-23
WO2016089990A12016-06-09
WO2016109663A22016-07-07
WO2016109684A22016-07-07
WO2016109689A22016-07-07
WO2017059059A12017-04-06
WO2015073774A12015-05-21
WO2017015451A12017-01-26
WO2014033167A12014-03-06
WO2014033170A12014-03-06
WO2017001655A12017-01-05
WO2015011281A12015-01-29
WO2018039531A12018-03-01
WO2016161268A12016-10-06
WO2016183266A12016-11-17
WO2017136403A12017-08-10
WO2015057945A12015-04-23
WO2015172128A12015-11-12
WO2015138895A12015-09-17
WO2017048950A12017-03-23
WO2018160878A12018-09-07
WO2018052967A12018-03-22
WO2018172852A12018-09-27
WO2009147188A12009-12-10
WO2014152725A12014-09-25
WO2016046530A12016-03-31
WO2017040757A12017-03-09
WO2012170752A12012-12-13
Foreign References:
US20170253609A12017-09-07
Other References:
FELD J. ET AL., ANTIVIRAL RES., vol. 76, 2007, pages 168
WEBER ET AL., ANTIVIRAL RES., vol. 54, 2002, pages 69
QIU ET AL., J. MED. CHEM., vol. 59, no. 16, 2016, pages 7651 - 7666
BIOORG. MED. CHEM., vol. 25, no. 3, 2017, pages 1042 - 1056
J. MED. CHEM, vol. 61, no. 14, 2018, pages 6247 - 6260
ZLOTNICK A ET AL., J. VIROL., vol. 4848, 2002
"Remington's Pharmaceutical Sciences", vol. 42, 1985, MACK PUBLISHING COMPANY, pages: 1418 - 396
"Journal of Pharmaceutical Science", vol. 66, 1977, pages: 2
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, 1991, pages 6690 - 6692
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285
N. KAKEYA ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
CAMPAGNA ET AL., J. VIROL., vol. 87, 2013, pages 6931 - 6942
MEHELLOU ET AL., CHEMMEDCHEM, vol. 4, 2009, pages 1779 - 1791
ORGANIC LETTERS, vol. 13, no. 8, 2011, pages 2012 - 2014
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, 2007, pages 7500 - 7501
MONATSHEFTE FUR CHEMIE, vol. 103, no. 1, pages 194 - 204
BERICHTE DER DEUTSCHEN CHEMISCHEN GESELLSCHAFT, vol. 17, no. 1, pages 559 - 568
A. ISIDRO-LLOBET ET AL., CHEM. REV., vol. 109, 2009, pages 2455 - 2504
A. EL-FAHAMF. ALBERICIO, CHEM. REV., vol. 111, 2011, pages 6557 - 6602
P.N. COLLIER ET AL., J. MED. CHEM., vol. 58, 2015, pages 5684 - 5688
JIMENEZ-SOMARRIBAS ET AL., J. MED. CHEM., vol. 60, 2017, pages 2305 - 2325
YANGBUCHWALD, JOURNAL OF ORGANOMETALLIC CHEMISTRY, 1999, pages 125 - 146
DANDRI ET AL., BEST PRACT RES CLIN GASTROENTEROL, vol. 31, 2017, pages 273 - 279
GUIDOTTI ET AL., J. VIROL., vol. 69, 1995, pages 6158 - 6169
WEBER ET AL., ANTIVIRAL RESEARCH, vol. 54, 2002, pages 69 - 78
JULANDER ET AL., ANTIVIR. RES., vol. 59, 2003, pages 155 - 161
PAULSEN ET AL., PLOSONE, vol. 10, 2015, pages e0144383
LI ET AL., HEPAT. MON., vol. 16, 2016, pages e34420
LUTGEHETMANN ET AL., GASTROENTEROLOGY, vol. 140, 2011, pages 2074 - 2083
HUANG ET AL., GASTROENTEROLOGY, vol. 142, 2012, pages 1447 - 1450
DION ET AL., J VIROL., vol. 87, 2013, pages 5554 - 5563
Attorney, Agent or Firm:
BOEHMERT & BOEHMERT (DE)
Download PDF:
Claims:
Claims

1. Compound ofFormula I

I

in which

- Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF2H, CF3, CF2CH3, F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, CH2OH, CH(CH3)OH, GH2F, CH(F)CH3, 1, C=C, CºC, CºN, C(CH3)2OH5 SCH3J OH, and OCH3

- R5 is H or methyl

- R6 is selected from the group comprising H, D, S02-Cl-C6-alkyl, S02-C3-C7- cycloalkyl, S02-C3-C7-heterocycloalkyl, S02-C2-C6-hydroxyalkyl, S02-C2-C6-alkyl-0-

Cl-C6-alkyl, SO2-C 1 -C4-carboxyalkyl, S02-aryl, S02-heteroaryl, S02-N(Rl2)(Rl3), C(=0)R8, C(=0)N(Rl2)(Rl3), C(=0)C(=0)N(Rl 2)(R13), Cl-C6-alkyl, C3-C6- cycloalkyl, C 1 -C4-carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, C1-C4- carboxamidoalkyl, C3-C7-heterocycloalkyl, C2-C6-aminoalkyl, Cl-C6-alkyl-0-Cl -06- alkyl, C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S02CH3, SO3H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C7- cycloalkyl, C3 -C7 -heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl-C6-alkyl-0- Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy, wherein C3-C7- heterocycloalkyl is optionally substituted with 1, 2, or 3 groups each independently selected from Cl-C6-alkyl or Cl-C6-alkoxy

- R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH2, acyl, S02CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6- alkenyloxy

- R12 and R13 are independently selected from the group comprising H, Cl-C6-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH2, acyl, S02CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7 -heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula I or the pharmaceutically acceptable salt thereof or a prodrug of a compound of Formula I or a pharmaceutically acceptable salt or a solvate or a hydrate thereof.

2. A compound of Formula I according to claim 1

- Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF2H, CF3, CF2CH3, F, Cl, Br, CH3, Et, i-Pr, c-Pr, D, CH2OH, CH(CH3)OH, CH2F, CH(F)CH3, 1, C=C, CºC, CºN, C(CH3)2OH, SCH3, OH, and OCH3

- R5 is H or methyl

- R6 is selected from the group comprising H, D, S02-Cl-C6-alkyl, S02-C3-C7- cycloalkyl, S02-C3-C7-heterocycloalkyl, S02-C2-C6-hydroxyalkyl, S02-C2-C6-alkyl-0- Cl-C6-alkyl, S02-Cl -C4-carboxyalkyl, S02-aryl, S02-heteroaryl, S02-N(Rl2)(Rl3), C(=0)R8, C(=0)N(R12)(Rl3), C(=0)C(=0)N(Rl 2)(R13), Cl-C6-alkyl, C3-C6- cycloalkyl, C 1 -C4-carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, C1-C4- carboxamidoalkyl, C3 -C7 -heterocycloalkyl, C2-C6-aminoalkyl, C 1 -C6-alkyl-0-C 1 -C6- alkyl, C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S02CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C7- cycloalkyl, C3 -C7 -heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C1-C6- hydroxyalkyl, and C2-C6 alkenyloxy

- R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3 -C7 -cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH2, acyl, SO2CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6- alkenyloxy

- R12 and R13 are independently selected from the group comprising H, Cl-C6-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH2, acyl, S02CH3, SO3H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7 -heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula I or the pharmaceutically acceptable salt thereof or a prodrug of a compound of Formula I or a pharmaceutically acceptable salt or a solvate or a hydrate thereof.

3. A compound of Formula I according to any of claims 1 or 2, wherein S02-aryl is S02-C6- aryl, and/or S02-heteroaryl is S02-Cl-C9 heteroaryl and/or heteroaryl is C 1 -C9-heteroaryl and wherein heteroaryl, S02-heteroaryl, S 02-heterocycloalkyl and heterocycloalkyl each has in the ring system 1 to 4 heteroatoms each independently selected from N, O and S, or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula I or the pharmaceutically acceptable salt thereof or a prodrug of a compound of Formula I or a pharmaceutically acceptable salt or a solvate or a hydrate thereof.

4. A compound of Formula I according to any one of claims 1 to 3,

or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula I or the pharmaceutically acceptable salt thereof or a prodrug of a compound of Formula I or a pharmaceutically acceptable salt or a solvate or a hydrate thereof, wherein the prodrag is selected from the group comprising esters, carbonates, acetyloxy derivatives, amino acid derivatives and phosphoramidate derivatives.

5. A compound of Formula I according to any of claims 1 to 4 that is a compound of

Formula II

II

in which

- Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF2H, CF3, CF2CH3, F, Cl, Br, Q¾, Et, i-Pr, c-Pr, D, CH2OH, CH(C¾)OH, C¾F, CH(F)CH3, 1, C=C, CºC, CºN, C(CH3)2OH, SC¾, OH, and OC¾

- R5 is H or methyl

- R7 is selected from the group comprising Cl-C6-alkyl, C2-C6-hydroxyalkyl, C2-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, Cl -C4-carboxyalkyl, C3-C7 -heterocycloalkyl, C6-aryl, and heteroaryl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S02CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula II or the pharmaceutically acceptable salt thereof or a prodrug of a compound of Formula II or a pharmaceutically acceptable salt or a solvate or a hydrate thereof.

6. A compound of Formula I according to any of claims 1 to 4 that is a compound of Formula III

in which

Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF2H, CF3, CF2CH3, F, Cl, Br, CH3, Et, i-Pr, c-Pr, D, CH2OH, CH(CH3)OH, CH2F, CH(F)CH3, 1, C=C, CºC, CºN, C(CH3)2OH, SC¾, OH, and OCH3

— R5 is H or methyl

— R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S02CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula III or the pharmaceutically acceptable salt thereof or a prodrug of a compound of Formula III or a pharmaceutically acceptable salt or a solvate or a hydrate thereof

7. A compound of Formula I according to any of claims 1 to 4 that is a compound of

Formula IV

in which

— Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF2H, CF3, CF2C¾, F, Cl, Br, CH3, Et, i-Pr, c-Pr, D, CH2OH, CH(CH3)0H, C¾F, CH(F)CH3, 1, C=C, CºC, CºN, C(CH3)2OH, SC¾, OH, and OC¾

— R5 is H or methyl

— R9, R10 and Rl 1 are independently selected from the group comprising H, C1-C5 -alkyl,

C 1 -C5-hydroxyalkyl, C 1 -C5-alkyl-0-C 1 -C6-alkyl, C3-C7-cycloalkyl, C1-C3- carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl, wherein Cl-C5-alkyl, Cl -C5-hydroxyalkyl, C 1 -C5-alkyl-0-C 1 -C6-alkyl and C 1 -C3 -carboxyalkyl are optionally substituted with 1 , 2, or 3 groups each independently selected from OH, halo, NH2, acyl, S02CH3, S03H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7 -heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, Cl -C6-hydroxyalkyl, and C2-C6 alkenyloxy

— R9 and RIO are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of a compound of Formula IV or the pharmaceutically acceptable salt thereof or a prodrag of a compound of Formula IV or a pharmaceutically acceptable salt or a solvate or a hydrate thereof.

8. A compound according to any of claims 1 to 7 or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of said compound or the pharmaceutically acceptable salt thereof or a prodrug of said compound or a pharmaceutically acceptable salt or a solvate or a hydrate thereof for use in the prevention or treatment of an HBV infection in subject.

9. A pharmaceutical composition comprising a compound according to any of claims 1 to 7 or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of said compound or the pharmaceutically acceptable salt thereof or a prodrag of said compound or a pharmaceutically acceptable salt or a solvate or a hydrate thereof, together with a pharmaceutically acceptable carrier.

10. A method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound according to any of claims 1 to 7 or a pharmaceutically acceptable salt thereof or a solvate or a hydrate of said compound or the pharmaceutically acceptable salt thereof or a prodrug of said compound or a pharmaceutically acceptable salt or a solvate or a hydrate thereof.

11. Method for the preparation of a compound of Formula I according to any of claims 1 to 4 by reacting a compound of Formula V

V

in which Rl, R2, R3 and R4 are as defined in claim 1, with a compound of Formula VI

VI

in which R5 and R6 are as defined in any of claims 1 to 4.

Description:
NOVEL 6,7-DIHYDRO-4H-PYRAZOLO [1 ,5- A] PYRAZINE INDOLE-2-

CARBOXAMIDES ACTIVE AGAINST THE HEPATITIS B VIRUS (HBV)

Technical Field

The present invention relates generally to novel antiviral agents. Specifically, the present invention relates to compounds which can inhibit the protein(s) encoded by hepatitis B virus (HBV) or interfere with the function of the HBV replication cycle, compositions comprising such compounds, methods for inhibiting HBV viral replication, methods for treating or preventing HBV infection, and processes for making the compounds.

Background of the Invention

Chronic HBV infection is a significant global health problem, affecting over 5% of the world population (over 350 million people worldwide and 1.25 million individuals in the US). Despite the availability of a prophylactic HBV vaccine, the burden of chronic HBV infection continues to be a significant unmet worldwide medical problem, due to suboptimal treatment options and sustained rates of new infections in most parts of the developing world. Current treatments do not provide a cure and are limited to only two classes of agents (interferon alpha and nucleoside analogues/inhibitors of the viral polymerase); drag resistance, low efficacy, and tolerability issues limit their impact.

The low cure rates of HBV are attributed at least in part to the fact that complete suppression of vims production is difficult to achieve with a single antiviral agent, and to the presence and persistence of covalently closed circular DNA (cccDNA) in the nucleus of infected hepatocytes. However, persistent suppression of HBV DNA slows liver disease progression and helps to prevent hepatocellular carcinoma (HCC).

Current therapy goals for HBV-infected patients are directed to reducing serum HBV DNA to low or undetectable levels, and to ultimately reducing or preventing the development of cirrhosis and HCC.

The HBV is an enveloped, partially double-stranded DNA (dsDNA) virus of the hepadnavirus family (Hepadnaviridae). HBV capsid protein (HBV-CP) plays essential roles in HBV replication. The predominant biological function of HBV-CP is to act as a structural protein to encapsidate pre-genomic RNA and form immature capsid particles, which spontaneously self- assemble from many copies of capsid protein dimers in the cytoplasm.

HBV-CP also regulates viral DNA synthesis through differential phosphorylation states of its C-terminal phosphorylation sites. Also, HBV-CP might facilitate the nuclear translocation of viral relaxed circular genome by means of the nuclear localization signals located in the arginine-rich domain of the C-terminal region of HBV-CP.

In the nucleus, as a component of the viral cccDNA mini-chromosome, HBV-CP could play a structural and regulatory role in the functionality of cccDNA mini-chromosomes. HBV-CP also interacts with viral large envelope protein in the endoplasmic reticulum (ER), and triggers the release of intact viral particles from hepatocytes.

HBV-CP related anti-HBV compounds have been reported. For example, phenylpropenamide derivatives, including compounds named AT-61 and AT-130 (Feld J. et al. Antiviral Res. 2007, 76, 168), and a class of thiazolidin-4-ones from Valeant (W02006/033995), have been shown to inhibit pre-genomic RNA (pgRNA) packaging.

F. Hoffinann-La Roche AG have disclosed a series of 3 -substituted tetrahydro-pyrazolo [1,5- ajpyrazines for the therapy of HBV (WO2016/113273, WO2017/198744, W02018/011162, W02018/011160, W02018/011163).

Heteroaryldihydropyrimidines (HAPs) were discovered in a tissue culture-based screening (Weber et al., Antiviral Res. 2002, 54, 69). These HAP analogs act as synthetic allosteric activators and are able to induce aberrant capsid formation that leads to degradation of HBV-CP (WO 99/54326, WO 00/58302, WO 01/45712, WO 01/6840). Further HAP analogs have also been described (J. Med. Chem. 2016, 59 (16), 7651-7666).

A subclass of HAPs from F. Hoffrnan-La Roche also shows activity against HBV (WO2014/184328, WO2015/132276, and WO2016/146598). A similar subclass from Sunshine Lake Pharma also shows activity against HBV ( W 02015/144093). Further HAPs have also been shown to possess activity against HBV (WO2013/102655, Bioorg. Med. Chem. 2017, 25(3) pp. 1042-1056, and a similar subclass from Enanta Therapeutics shows similar activity (WO2017/011552). A further subclass from Medshine Discovery shows similar activity (WO2017/076286). A further subclass (Janssen Pharma) shows similar activity (WO2013/102655).

A subclass of pyridazones and triazinones (F. Hoffman-La Roche) also show activity against

HBV (WO2016/023877), as do a subclass of tetrahydropyridopyridines (WO2016/177655). A subclass of tricyclic 4-pyridone-3 -carboxylic acid derivatives from Roche also show similar anti-HBV activity (W02017/013046).

A subclass of sulfamoyl-arylamides from Novira Therapeutics (now part of Johnson & Johnson Inc.) also shows activity against HBV (W02013/006394, W02013/096744, WO2014/165128, W02014/184365, WO2015/109130, WO2016/089990, WO2016/ 109663, WO2016/109684, WO2016/109689, W02017/059059). A similar subclass of thioether-arylamides (also from Novira Therapeutics) shows activity against HBV (W02016/089990). Additionally, a subclass of aryl-azepanes (also from Novira Therapeutics) shows activity against HBV (WO2015/073774). A similar subclass of arylamides from Enanta Therapeutics show activity against HBV (W02017/015451).

Sulfamoyl derivatives from Janssen Pharma have also been shown to possess activity against HBV (WO2014/033167, W02014/033170, WO2017/001655, J. Med. Chem, 2018, 61(14) 6247-6260).

A subclass of glyoxamide substituted pyrrolamide derivatives also from Janssen Pharma have also been shown to possess activity against HBV (W02015/011281). A similar class of glyoxamide substituted pyrrolamides (Gilead Sciences) has also been described (WO2018/039531).

A subclass of sulfamoyl- and oxalyl-heterobiaryls from Enanta Therapeutics also show activity against HBV (WO2016/161268, WO2016/183266, WO2017/015451, WO2017/136403 & US20170253609).

A subclass of aniline-pyrimidines from Assembly Biosciences also show activity against HBV (WO2015/057945, WO2015/172128). A subclass of fused tri-cycles from Assembly Biosciences (dibenzo-thiazepinones, dibenzo-diazepinones, dibenzo-oxazepinones) show activity against HBV (WO2015/138895, W02017/048950). A series of cyclic sulfamides has been described as modulators of HBV-CP function by Assembly Biosciences (WO2018/160878). Arbutus Biopharma have disclosed a series of benzamides for the therapy of HBV (WO2018/052967, WO2018/172852).

It was also shown that the small molecule bis-ANS acts as a molecular 'wedge' and interferes with normal capsid-protein geometry and capsid formation (Zlotnick A et al. J. Virol. 2002,

4848).

Problems that HBV direct acting antivirals may encounter are toxicity, mutagenicity, lack of selectivity, poor efficacy, poor bioavailability, low solubility and difficulty of synthesis. There is a thus a need for additional inhibitors for the treatment, amelioration or prevention of HBV that may overcome at least one of these disadvantages or that have additional advantages such as increased potency or an increased safety window.

Administration of such therapeutic agents to an HBV infected patient, either as monotherapy or in combination with other HBV treatments or ancillary treatments, will lead to significantly reduced vims burden, improved prognosis, diminished progression of the disease and/or enhanced seroconversion rates.

Summary of the invention

Provided herein are compounds useful for the treatment or prevention of HBV infection in a subject in need thereof, and intermediates useful in their preparation. The subject matter of the invention is a compound of Formula I:

— Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, C¾OH, CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OC¾

— R5 is H or methyl

— R6 is selected from the group comprising H, D, S0 2 -C 1 -C6-alkyl, S0 2 -C3-C7- cycloalkyl, S0 2 -C3-C7-heterocycloalkyl, S0 2 -C2-C6-hydroxyalkyl, S0 2 -C2-C6-alkyl-0- Cl-C6-alkyl, S0 2 -C 1 -C4-carboxyalkyl, S0 2 -aryl, S0 2 -heteroaryl, S0 2 -N(R12)(R13), C(=0)R8, C(=0)N(R12)(R13), C(=0)C(=0)N(R12)(R13), Cl-C6-alkyl, C3-C6- cycloalkyl, C 1 -C4-carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, C1-C4- carboxamidoalkyl, C3 -C7 -heterocycloalkyl, C2-C6-aminoalkyl, C 1 -C6-alkyl-0-C 1 -C6- alkyl C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 C¾, S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C7- cycloalkyl, C3-C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl-C6-alkyl-0- Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy, wherein C3-C7- heterocycloalkyl is optionally substituted with 1, 2, or 3 groups each independently selected from Cl-C6-alkyl or Cl-C6-a!koxy,

— R8 is selected from the group comprising Cl-C6-alkyl, Cl -C6-hydroxyalkyl, C1-C6- alkyl-0-Cl-C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocyclo alkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6- a!kenyloxy

- R12 and R13 are independently selected from the group comprising H, Cl-Cd-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7 -heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms

In one embodiment of the invention subject matter of the invention is a compound of Formula I in which

- Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, CH 3 , Et, i-Pr, c-Pr, D, CH 2 OH, CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SC¾, OH, and OCH 3

- R5 is H or methyl

- R6 is selected from the group comprising H, D, S0 2 -Cl-C6-alkyl, S0 2 -C3-C7- cycloalkyl, S0 2 -C3-C7 -heterocycloalkyl, S0 2 -C2-C6-hydroxyalkyl, S0 2 -C2-C6-alkyl-0- Cl-C6-alkyl, S0 2 -Cl -C4-carboxyalkyl, S0 2 -aryl, S0 2 -heteroaryl, S0 2 -N(Rl2)(Rl3), C(=0)R8, C(=0)N(Rl 2)(Rl 3), C(=0)C(=0)N(Rl2)(Rl3), Cl-C6-alkyl, C3-C6- cycloalkyl, C 1 -C4-carboxyalkyl, Cl -C4-acylsulfonamido-alkyl, C1-C4- carboxamidoalkyl, C3-C7-heterocycloalkyl, C2-C6-aminoalkyl, Cl-C6-alkyl-0-Cl-C6- alkyl C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C7- cycloalkyl, C3-C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl-C6-alkyl-0- Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy, wherein C3-C7- heterocycloalkyl is optionally substituted with 1, 2, or 3 groups each independently selected from Cl-C6-alkyl or Cl-Cb-alkoxy,

- R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cydoalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S0 2 C¾, S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6- alkenyloxy

- R12 and R13 are independently selected from the group comprising H, Cl-C6-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7-heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms In one embodiment subject matter of the present invention is a compound according to Formula I in which Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 C¾, F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, C¾OH, CH(C¾)OH, C¾F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OC¾, preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, CH 3 , Et, and i-Pr, and most preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, CH 3 , and Et.

In one embodiment subject matter of the present invention is a compound according to Formula I in which R5 is H. In one embodiment subject matter of the present invention is a compound according to Formula I in which R5 is methyl.

In one embodiment subject mater of the present invention is a compound according to Formula I in which R6 is selected from the group comprising H, D, S0 2 -C 1 -C6-alkyl, S0 2 -C3-C7- cycloalkyl, S0 2 -C3-C7-heterocycloalkyl, S0 2 -C2-C6-hydroxyalkyl, S0 2 -C2-C6-alkyl-0-C 1 -C6- alkyl, S0 2 -C1 -C4-carboxyalkyl, S0 2 -aryl, S0 2 -heteroaryl, S0 2 -N(Rl2)(R13), C(=0)R8, C(=0)N (R 12)(R 13 ), C(=0)C(=0)N(Rl 2)(Rl 3), Cl-C6-alkyl, C3-C6-cycloalkyl, C1-C4- carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, C 1 -C4-carboxamidoalkyl, C3 -C7 -heterocycloalkyl, C2-C6-aminoalkyl, C 1 -C6-alkyl-0-C 1 -C6-alkyl, C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl -06-alkyl, C3-C7-cycloaikyl, C3-C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl- C6-alkyl-0-C 1 -C6-alkyl, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy, wherein C3-C7- heterocyclo alkyl is optionally substituted with 1, 2, or 3 groups each independently selected from

Cl-C6-alkyl or Cl-C6-alkoxy, preferably H, Cl-C6-alkyl, C3-C6-cycloalkyl, C4-C7- heterocycloalkyl and C2-C6-hydroxyalkyl optionally substituted with OH, Cl-C6-alkoxy, Cl- C3-C7-cycloalkyl, C6-hydroxyalkyl and C3 -C7 -heterocycloalkyl.

In one embodiment subject matter of the present invention is a compound according to Formula I in which R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3 -C7 -cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7 -heterocycloalkyl, C6-aryl, heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6- aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7-heterocycloalkyl, Cl -C6-haloalkyl, Cl- C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6-alkenyloxy.

In one embodiment subject matter of the invention is a compound according to Formula I in which R12 and R13 are independently selected from the group comprising H, Cl-C6-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH ¾ acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyI , C3 -C7 -heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl- C6-hydroxyalkyl, and C2-C6 alkenyloxy.

In one embodiment subject matter of the invention is a compound according to Formula I in which R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms.

One embodiment of the invention is a compound of Formula I or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject.

One embodiment of the invention is a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof according to the present invention, together with a pharmaceutically acceptable carrier. One embodiment of the invention is a method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof according to the present invention.

A further embodiment of the invention is a compound of Formula I or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject in need thereof.

— Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, C¾OH, CH(CH 3 )OH, C¾F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OC¾

— R5 is H or methyl

— R6 is selected from the group comprising H, D, S0 2 -C 1 -C6-alkyl, S0 2 -C3-C7- cycloalkyl, S0 2 -C3-C7-heterocycloalkyl, S0 2 -C2-C6-hydroxyalkyl, S0 2 -C2-C6-alkyl-0- Cl-Cb-alkyl, S 0 2 -C 1 -C4-carboxyalkyl, S0 2 -aryl, S0 2 -heteroaryl, S0 2 -N(R12)(R13), C(=0)R8, C(=0)N (R 12)(R 13 ), C(=0)C(=0)N(R12)(Rl3), Cl-C6-alkyl, C3-C6- cycloalkyl, C 1 -C4-carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, C 1 -C4- carboxamidoalkyl, C3-C7-heterocycloalkyl, C2-C6-aminoalkyl, C 1 -C6-alkyl-0-C 1 -C6- alkyl C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C7- cycloalkyl, C3 -C7 -heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C1-C6- hydroxyalkyl, and C2-C6 alkenyloxy

— R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7- heterocydoalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6- alkenyloxy

- R12 and R13 are independently selected from the group comprising H, Cl-C6-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl , C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3 -C7 -heterocycloalkyl , C1-C6- haloalkyl, Cl-C6-alkoxy, Cl -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms

In one embodiment of the invention subject matter of the invention is a compound of Formula I in which

— Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, C¾OH, CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OCH 3

- R5 is H or methyl

— R6 is selected from the group comprising H, D, S0 -Cl-C6-alkyl, S0 2 -C3-C7- cycloalkyl, S0 2 -C3-C7-heterocycloalkyl, S0 2 -C2-C6-hydroxyalkyl, S0 C2-C6-alkyl-0- Cl-C6-alkyl, S0 2 -C 1 -C4-carboxyalkyl, S0 2 -aryl, S0 2 -heteroaryl, S0 2 -N(R12)(Rl3), C(=0)R8, C(=0)N (R 12)(R 13 ), C(=0)C(=0)N(Rl2)(Rl3), Cl-C6-alkyl, C3-C6- cycloalkyl, C 1 -C4-carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, C 1 -C4- carboxamidoalkyl, C3-C7-heterocycloalkyl, C2-C6-aminoalkyl, C 1 -C6-alkyl-0-C 1 -C6- alkyl C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C7- cycloalkyl, C3 -C7 -heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C1-C6- hydroxyalkyl, and C2-C6 alkenyloxy

- R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6- alkenyloxy

- R12 and R13 are independently selected from the group comprising H, Cl-C6-alkyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , SO 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7-heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms

In one embodiment subject matter of the present invention is a compound according to Formula I in which Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, CH 2 OH, CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OC¾, preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, CH 3 , Et, and i-Pr, and most preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, C¾, and Et.

In one embodiment subject matter of the present invention is a compound according to Formula I in which R5 is selected from the group comprising H, and methyl.

In one embodiment subject matter of the present invention is a compound according to Formula I in which R6 is selected from the group comprising H, D, S0 2 -Cl-C6-alkyl, S0 2 -C3-C7- cycloa!kyl, S 0 2 -C3 -C7 -heterocycloalkyl, S0 2 -C2-C6-hydroxyalkyl, S0 2 -C2-C6-alkyl-0-Cl -Chalky!, S0 2 -Cl-C4-carboxyalkyl, S0 2 -aryl, S0 2 -heteroaryl, S0 2 -N(R12)(R13), C(=0)R8, C(=0)N(R12)(R13), C(=0)C(=0)N(Rl2)(R13), Cl-C6-alkyl, C3-C6-cycloalkyl, C1-C4- carboxyalkyl, C 1 -C4-acylsulfonamido-alkyl, Cl -C4-carboxamidoalkyl, C3-C7-heterocycloalkyl, C2-C6-aminoalkyl, C 1 -C6-alkyl-0-C 1 -C6-alkyl, C2-C6-hydroxyalkyl, and acyl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl,

Cl-C6-alkyl, C3-C7-cycloalkyl, C3-C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl - C6-hydroxyalkyl, and C2-C6 alkenyloxy, preferably H, Cl-C6-alkyl, C3-C6-cycloalkyl, C4-C7- heterocycloalkyl and C2-C6-hydroxyalkyl optionally substituted with OH, Cl-C6-alkoxy, Cl- C3-C7-cycloalkyl, C6-hydroxyalkyl and C3-C7-heterocyeloalkyl.

In one embodiment subject matter of the present invention is a compound according to Formula I in which R8 is selected from the group comprising Cl-Cb-alky!, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3 -C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6- aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl- C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6-alkenyloxy.

In one embodiment subject matter of the invention is a compound according to Formula I in which R12 and R13 are independently selected from the group comprising H, Cl-C6-aikyl, C3- C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , SO 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7 -heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl- C6-hydroxyalkyl, and C2-C6 alkenyloxy.

In one embodiment subject matter of the invention is a compound according to Formula I in which R12 and R13 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms.

One embodiment of the invention is a compound of Formula I or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject. One embodiment of the invention is a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof according to the present invention, together with a pharmaceutically acceptable carrier.

One embodiment of the invention is a method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof according to the present invention.

A further embodiment of the invention is a compound of Formula II or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject in need thereof.

in which

- Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, CH 3 , Et, i-Pr, c-Pr, D, CH 2 OH, CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OCH 3

- R5 is H or methyl

- R7 is selected from the group comprising Cl-C6-alkyl, C2-C6-hydroxyalkyl, C2-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, and heteroaryl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, SO2CH3, S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, Cl -C6-hydroxyalkyl, and C2-C6 alkenyloxy. In one embodiment subject matter of the present invention is a compound according to Formula II in which Rl , R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, CH 3 , Et, i-Pr, c-Pr, D, CH 2 OH, CH(C¾)OH, CH 2 F, CH(F)C¾, 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH3, OH, and OC¾, preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, CH 3 , and Et.

In one embodiment subject matter of the present invention is a compound according to Formula II in which R5 is selected from the group comprising H and methyl.

In one embodiment subject matter of the present invention is a compound according to Formula II in which R7 is selected from the group comprising Cl-C6-alkyl, C2-C6-hydroxyalkyl, C2-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, Cb-aryl, and heteroaryl, optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl-06-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy.

One embodiment of the invention is a compound of Formula II or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject.

One embodiment of the invention is a pharmaceutical composition comprising a compound of Formula II or a pharmaceutically acceptable salt thereof according to the present invention, together with a pharmaceutically acceptable carrier.

One embodiment of the invention is a method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of Formula II or a pharmaceutically acceptable salt thereof according to the present invention.

A further embodiment of the invention is a compound of Formula III or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject in need thereof.

III

in which

Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, CH 3 , Et, i-Pr, c-Pr, D, CH 2 OH,

CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OC¾

— R5 is H or methyl

— R8 is selected from the group comprising Cl-C6-alkyl, C 1 -C6-hydroxyalkyl, C1-C6- alkyl-O-C 1 -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 C¾, S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7- heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy.

In one embodiment subject matter of the present invention is a compound according to Formula III in which Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, CH 2 OH, CH(CH 3 )OH,

C¾F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OCH 3 , preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, CH 3 , and Et.

In one embodiment subject matter of the present invention is a compound according to Formula III in which R5 is selected from the group comprising H and methyl.

In one embodiment subject matter of the present invention is a compound according to Formula III in which R8 is selected from the group comprising Cl -C6-alkyl, C 1 -C6-hydroxyalkyl, Cl- C6-alkyl-0-Cl -C6-alkyl, C3-C7-cycloalkyl, C 1 -C4-carboxyalkyl, C3-C7-heterocycloalkyl, C6- aryl, and heteroaryl optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, N¾, acyl, S0 2 C¾, S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3 -C7 -heterocycloalkyl, Cl- C6-haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy.

One embodiment of the invention is a compound of Formula III or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject.

One embodiment of the invention is a pharmaceutical composition comprising a compound of Formula III or a pharmaceutically acceptable salt thereof according to the present invention, together with a pharmaceutically acceptable carrier.

One embodiment of the invention is a method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of Formula III or a pharmaceutically acceptable salt thereof according to the present invention.

A further embodiment of the invention is a compound of Formula IV or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject in need thereof.

in which

- Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, CH 2 OH, CH(CH 3 )OH, CH 2 F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OQ¾

- R5 is H or methyl - R9, RIO and Rl 1 are independently selected from the group comprising H, Cl -C5 -alkyl,

C 1 -C5-hydroxyalkyl, Cl -C5-alkyl-0-Cl -C6-alkyl, C3-C7-cycloalkyl, C1-C3- carboxyalkyl, C3-C7-heterocycloalkyl, C6-aryl, and heteroaryl, wherein Cl-C5-alkyl, C 1 -C5-hydroxyalkyl, C 1 -C5-alkyl-0-Cl -C6-alkyl and C 1 -C3 -carboxyalkyl are optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3-C6-cycloalkyl, C3-C7-heterocycloalkyl, C1-C6- haloalkyl, Cl-C6-alkoxy, C 1 -C6-hydroxyalkyl, and C2-C6 alkenyloxy

- R9 and R10 are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocycloalkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms.

In one embodiment subject matter of the present invention is a compound according to Formula IV in which Rl, R2, R3 and R4 are for each position independently selected from the group comprising H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, Br, C¾, Et, i-Pr, c-Pr, D, CH 2 OH, CH(C¾)OH, C¾F, CH(F)CH 3 , 1, C=C, CºC, CºN, C(CH 3 ) 2 OH, SCH 3 , OH, and OCH 3 , preferably H, CF 2 H, CF 3 , CF 2 CH 3 , F, Cl, C¾, and Et.

In one embodiment subject matter of the present invention is a compound according to Formula IV in which R5 is selected from the group comprising H and methyl.

In one embodiment subject matter of the present invention is a compound according to Formula IV in which R9, RIO and R11 are independently selected from the group comprising H, C1-C5- alkyl, Cl-C5-hydroxyalkyl, Cl-C5-alkyl-0-Cl -C6-alkyl, C3-C7-cycloalkyl, C1-C3- carboxyalkyl, C3 -C7 -heterocycloalkyl, C6-aryl, and heteroaryl, wherein Cl-C5-a!kyI, C1-C5- hydroxyalkyl, C 1 -C5-alkyl-0-C 1 -C6-alkyl and C 1 -C3-carboxyalkyl are optionally substituted with 1, 2, or 3 groups each independently selected from OH, halo, NH 2 , acyl, S0 2 CH 3 , S0 3 H, carboxy, carboxyl ester, carbamoyl, substituted carbamoyl, C6-aryl, heteroaryl, Cl-C6-alkyl, C3 -C6-cycloalkyl, C3-C7-heterocycloalkyl, Cl-C6-haloalkyl, Cl-C6-alkoxy, C1-C6- hydroxyalkyl, and C2-C6 alkenyloxy.

In one embodiment subject matter of the invention is a compound according to Formula IV in which R9 and RIO are optionally connected to form a C3-C7 cycloalkyl ring, or a C4-C7- heterocyclo alkyl ring containing 1 or 2 nitrogen, sulfur or oxygen atoms. One embodiment of the invention is a compound of Formula IV or a pharmaceutically acceptable salt thereof according to the invention, for use in the prevention or treatment of an HBV infection in subject.

One embodiment of the invention is a pharmaceutical composition comprising a compound of Formula IV or a pharmaceutically acceptable salt thereof according to the present invention, together with a pharmaceutically acceptable carrier.

One embodiment of the invention is a method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of Formula IV or a pharmaceutically acceptable salt thereof according to the present invention.

In some embodiments, the dose of a compound of the invention is from about 1 mg to about 2,500 mg. In some embodiments, a dose of a compound of the invention used in compositions described herein is less than about 10,000 mg, or less than about 8,000 mg, or less than about 6,000 mg, or less than about 5,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1,000 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg. Similarly, in some embodiments, a dose of a second compound (i.e., another drag for HBV treatment) as described herein is less than about 1 ,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof. All before mentioned doses refer to daily doses per patient.

In general it is contemplated that an antiviral effective daily amount would be from about 0.01 to about 50 mg/kg, or about 0.01 to about 30 g/kg body weight. It maybe appropriate to administer the required dose as two, three, four or more sub-doses at appropriate intervals throughout the day. Said sub-doses may be formulated as unit dosage forms, for example containing about 1 to about 500 mg, or about 1 to about 300 mg or about 1 to about 100 mg, or about 2 to about 50 mg of active ingredient per unit dosage form. The compounds of the invention may, depending on their structure, exist as salts, solvates or hydrates. The invention therefore also encompasses the salts, solvates or hydrates and respective mixtures thereof.

The compounds of the invention may, depending on their structure, exist in tautomeric or stereoisomeric forms (enantiomers, diastereomers). The invention therefore also encompasses the tautomers, enantiomers or diastereomers and respective mixtures thereof. The stereoisomerically uniform constituents can be isolated in a known manner from such mixtures of enantiomers and/or diastereomers.

i r

Definitions

Listed below are definitions of various terns used to describe this invention. These definitions apply to the terms as they are used throughout this specification and claims unless otherwise limited in specific instances either individually or as part of a larger group.

Unless defined otherwise all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Generally the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry and peptide chemistry are those well known and commonly employed in the art.

As used herein the articles "a” and "an” refer to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example, "an element” means one element or more than one element. Furthermore, use of the term "including” as well as other forms such as "include 44 ,“includes” and "included", is not limiting.

As used herein the term "capsid assembly modulator” refers to a compound that disrupts or accelerates or inhibits or hinders or delays or reduces or modifies normal capsid assembly (e.g. during maturation) or normal capsid disassembly (e.g. during infectivity) or perturbs capsid stability, thereby inducing aberrant capsid morphology or aberrant capsid function. In one embodiment, a capsid assembly modulator accelerates capsid assembly or disassembly thereby inducing aberrant capsid morphology. In another embodiment a capsid assembly modulator interacts (e.g. binds at an active site, binds at an allosteric site or modifies and/or hinders folding and the like), with the major capsid assembly protein (HBV-CP), thereby disrupting capsid assembly or disassembly. In yet another embodiment a capsid assembly modulator causes a perturbation in the structure or function of HBV-CP (e.g. the ability of HBV-CP to assemble, disassemble, bind to a substrate, fold into a suitable conformation or the like which attenuates viral infectivity and/or is lethal to the virus).

As used herein the term "treatment” or "treating” is defined as the application or administration of a therapeutic agent i.e., a compound of the invention (alone or in combination with another pharmaceutical agent) to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g for diagnosis or ex vivo applications) who has an HBV infection, a symptom of HBV infection, or the potential to develop an HBV infection with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the HBV infection, the symptoms of HBV infection or the potential to develop an HBV infection. Such treatments may be specifically tailored or modified based on knowledge obtained from the field of pharmacogenomics.

As used herein the term "prevent” or“prevention” means no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease.

As used herein the term "patient",“individual” or "subject” refers to a human or a non-human mammal. Non-human mammals include for example livestock and pets such as ovine, bovine, porcine, feline, and murine mammals. Preferably the patient, subject, or individual is human.

As used herein the terms "effective amount”, "pharmaceutically effective amount”, and "therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.

As used herein the term“pharmaceutically acceptable” refers to a material such as a carrier or diluent which does not abrogate the biological activity or properties of the compound and is relatively non-toxic i.e. the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.

As used herein the term "pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent or in a mixture of the two; generally nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences l7 th ed. Mack Publishing Company, Easton, Pa., 1985 r.1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.

As used herein the term "composition” or“pharmaceutical composition” refers to a mixture of at least one compound useful within the invention with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including but not limited to intravenous, oral, aerosol, rectal, parenteral, ophthalmic, pulmonary and topical administration.

As used herein the term "pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function. Typically such constructs are carried or transported from one organ, or portion of the body, to another organ or portion of the body. Each carrier must be“acceptable" in the sense of being compatible with the other ingredients of the formulation including the compound use within the invention and not injurious to the patient. Some examples of materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches such as com starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt, gelatin, talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; glycols such as propylene glycol; polyols such as glycerin, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminium hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol; phosphate buffer solutions and other non-toxic compatible substances employed in pharmaceutical formulations.

As used herein "pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents and absorption delaying agents and the like that are compatible with the activity of the compound useful within the invention and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions. The "pharmaceutically acceptable carrier” may farther include a pharmaceutically acceptable salt of the compound useful within the invention. Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are known in the art and described for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Company, Easton, Pa., 1985) which is incorporated herein by reference.

As used herein, the term "substituted” means that an atom or group of atoms has replaced hydrogen as the substituent attached to another group.

As used herein, the term "comprising” also encompasses the option“consisting of’.

As used herein, the term "alkyl" by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e. Cl-C6-alkyl means one to six carbon atoms) and includes straight and branched r chains. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, and hexyl. In addition, the term“alkyl” by itself or as part of another substituent can also mean a C1-C3 straight chain hydrocarbon substituted with a C3-C5-carbocylic ring. Examples include (cyclopropyl)methyl, (cyclobutyl)methyl and (cyclopentyl)methyl . For the avoidance of doubt, where two alkyl moieties are present in a group, the alkyl moieties may be the same or different.

As used herein the term "alkenyl" denotes a monovalent group derived from a hydrocarbon moiety containing at least two carbon atoms and at least one carbon-carbon double bond of either E or Z stereochemistry. The double bond may or may not be the point of attachment to another group. Alkenyl groups (e.g. C2-C8-alkenyl) include, but are not limited to for example ethenyl, propenyl, prop-l-en-2-yl, butenyl, methyl-2 -buten- 1 -yl, heptenyl and octenyl. For the avoidance of doubt, where two alkenyl moieties are present in a group, the alkyl moieties may be the same or different.

As used herein, a C2-C6-alkynyl group or moiety is a linear or branched alkynyl group or moiety containing from 2 to 6 carbon atoms, for example a C2-C4 alkynyl group or moiety containing from 2 to 4 carbon atoms. Exemplary alkynyl groups include -CºCH or -CH 2 -CºC, as well as 1- and 2-butynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 2-hexynyl, 3-hexynyl, 4- hexynyl and 5-hexynyl. For the avoidance of doubt, where two alkynyl moieties are present in a group, they may be the same or different.

As used herein, the term "halo” or "halogen” alone or as part of another substituent means unless otherwise stated a fluorine, chlorine, bromine, or iodine atom, preferably fluorine, chlorine, or bromine, more preferably fluorine or chlorine. For the avoidance of doubt, where two halo moieties are present in a group, they may be the same or different.

As used herein, a Cl-C6-alkoxy group or C2-C6-alkenyloxy group is typically a said C1-C6- alkyl (e.g. a C1-C4 alkyl) group or a said C2-C6-alkenyl (e.g. a C2-C4 alkenyl) group respectively which is attached to an oxygen atom.

As used herein the term "aryl" employed alone or in combination with other terms, means unless otherwise stated a carbocyclic aromatic system containing one or more rings (typically one, two or three rings) wherein such rings may be attached together in a pendant manner such as a biphenyl, or may be fused, such as naphthalene. Examples of aryl groups include phenyl, anthracyl, and naphthyl. Preferred examples are phenyl (e.g. C6-aryl) and biphenyl (e.g. 02- aryl). In some embodiments aryl groups have from six to sixteen carbon atoms. In some embodiments aryl groups have from six to twelve carbon atoms (e.g. C6-C12-aryl). In some embodiments, aryl groups have six carbon atoms (e.g. C6-aryl).

As used herein the terms "heteroaryl" and "heteroaromatic” refer to a heterocycle having aromatic character containing one or more rings (typically one, two or three rings). Heteroaryl substituents may be defined by the number of carbon atoms e.g. Cl-C9-heteroaryl indicates the number of carbon atoms contained in the heteroaryl group without including the number of heteroatoms. For example a C 1 -C9-heteroaryl will include an additional one to four heteroatoms. A polycyclic heteroaryl may include one or more rings that are partially saturated. Non-limiting examples of heteroaryls include:

Additional non-limiting examples of heteroaryl groups include pyridyl, pyrazinyl, pyrimidinyl (including e.g. 2-and 4-pyrimidinyl), pyridazinyU thienyl, furyl, pyrrolyl (including e.g., 2-pyrrolyl), imidazolyl, thiazolyl, oxazolyl, pyrazolyl (including e.g. 3- and 5-pyrazolyl), isothiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3- oxadiazolyl, 1 ,3 ,4-thiadiazolyland 1 ,3,4-oxadiazolyl. Non-limiting examples of polycyclic heterocycles and heteroaryls include indolyl (including 3-, 4-, 5-, 6-and 7-indolyl), indolinyl, quinolyl, tetrahydroquinolyl , isoquinolyl (including, e.g. 1-and 5-isoquinolyl), 1 ,2,3,4- tetrahydroisoquinolyl, cinnolinyl, quinoxalinyl (including, e .g 2-and 5-quinoxalinyl), quinazolinyl, phthalazinyl, 1 ,8-naphthyridinyl, 1 ,4-benzodioxanyl, coumarin,

dihydrocoumarin, 1 ,5-naphthyridinyl, benzofuryl (including, e .g. 3-, 4-, 5-, 6-, and 7- benzofuryl), 2,3 -dihydrobenzo furyl, 1 ,2-benzisoxazolyl , benzothienyl (including e.g. 3-, 4-, 5-, 6-, and 7-benzothienyl), benzoxazolyl, benzothiazolyl (including e.g. 2-benzothiazolyl and 5- benzothiazolyl), purinyl, benzimidazolyl (including e.g. r 2-benzimidazolyl), benzotriazolyl, thioxanthinyl, carbazoly!, carbolinyl, acridinyl, pyrrolizidinyl and quinolizidinyl.

As used herein the term "haloalkyl” is typically a said alkyl, alkenyl, alkoxy or alkenoxy group respectively wherein any one or more of the carbon atoms is substituted with one or more said halo atoms as defined above. Haloalkyl embraces monohaloalkyl, dihaloalkyl, and polyhaloalkyl radicals. The term "haloalkyP’includes but is not limited to fluoromethyl, 1- fluoroethyl, difluoromethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, difluoromethoxy, and trifluoromethoxy.

As used herein, a C 1 -C6-hydroxyalkyl group is a said Cl -C6 alkyl group substituted by one or more hydroxy groups. Typically, it is substituted by one, two or three hydroxyl groups. Preferably, it is substituted by a single hydroxy group.

As used herein, a C 1 -C6-aminoalkyl group is a said C1-C6 alkyl group substituted by one or more amino groups. Typically, it is substituted by one, two or three amino groups. Preferably, it is substituted by a single amino group.

As used herein, a C 1 -C4-carboxyalkyl group is a said C1-C4 alkyl group substituted by carboxyl group.

As used herein, a C 1 -C4-carboxamidoalkyl group is a said C1-C4 alkyl group substituted by a substituted or unsubstituted carboxamide group.

As used herein, a Cl -C4-acylsulfonamido-alkyl group is a said C1-C4 alkyl group substituted by an acylsulfonamide group of general formula C(=0)NHS0 2 CH 3 or C(=0)NH S 0 2 -c-Pr.

As used herein the term "cycloalkyl" refers to a monocyclic or polycyclic nonaromatic group wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. In one embodiment, the cycloalkyl group is saturated or partially unsaturated. In another embodiment, the cycloalkyl group is fused with an aromatic ring. Cycloalkyl groups include groups having 3 to 10 ring atoms (C3-C 10-cycloalkyl) , groups having 3 to 8 ring atoms (C3-C8-cycloalkyl), groups having 3 to 7 ring atoms (C3-C7-cycloalkyl) and groups having 3 to 6 ring atoms (C3- C6-cyeloalkyl). Illustrative examples of cycloalkyl groups include, but are not limited to the following moieties:

Monocyclic cycloalkyls include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Dicyclic cycloalkyls include but are not limited to tetrahydronaphthyl, indanyl, and tetrahydropentalene. Polycyclic cycloalkyls include adamantine and norbomane. The term cycloalkyl includes "unsaturated nonaromatic carbocyclyl” or "nonaromatic unsaturated carbocyclyl" groups both of which refer to a nonaromatic carbocycle as defined herein which contains at least one carbon-carbon double bond or one carbon-carbon triple bond.

As used herein the terms "heterocycloalky!" and "heterocyc!y!” refer to a heteroalicyclic group containing one or more rings (typically one, two or three rings), that contains one to four ring heteroatoms each selected from oxygen, sulfur and nitrogen. In one embodiment each heterocyclyl group has from 3 to 10 atoms in its ring system with the proviso that the ring of said group does not contain two adjacent oxygen or sulfur atoms. In one embodiment each heterocyclyl group has a fused bicyclic ring system with 3 to 10 atoms in the ring system, again with the proviso that the ring of said group does not contain two adjacent oxygen or sulfur atoms. In one embodiment each heterocyclyl group has a bridged bicyclic ring system with 3 to 10 atoms in the ring system, again with the proviso that the ring of said group does not contain two adjacent oxygen or sulfur atoms. In one embodiment each heterocyclyl group has a spiro- bicyclic ring system with 3 to 10 atoms in the ring system, again with the proviso that the ring of said group does not contain two adjacent oxygen or sulfur atoms. Heterocyclyl substituents may be alternatively defined by the number of carbon atoms e.g. C2-C8-heterocyclyl indicates the number of carbon atoms contained in the heterocyclic group without including the number of heteroatoms. For example a C2-C8-heterocyclyl will include an additional one to four heteroatoms. In another embodiment the heterocycloalkyl group is fused with an aromatic ring. . In another embodiment the heterocycloalkyl group is fused with a heteroaryl ring. In one embodiment the nitrogen and sulfur heteroatoms may be optionally oxidized and the nitrogen atom may be optionally quatemized. The heterocyclic system may be attached, unless otherwise stated, at any heteroatom or carbon atom that affords a stable structure. An example of a 3-membered heterocyclyl group includes and is not limited to aziridine. Examples of 4-membered heterocycloalkyl groups include, and are not limited to azetidine and a beta-lactam. Examples of 5-membered heterocyclyl groups include, and are not limited to pyrrolidine, oxazolidine and thiazolidinedione. Examples of 6-membered heterocycloalkyl groups include, and are not limited to, piperidine, morpholine, piperazine, N-acetylpiperazine and N-acetylmorpholine. Other non-limiting examples of heterocyclyl groups are

Examples of heterocycles include monocyclic groups such as aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, pyrroline, pyrazolidine, imidazoline, dioxolane, sulfolane, 2,3-dihydrofuran, 2,5-dihydrofuran, tetrahydrofuran, thiophane, piperidine, 1 ,2,3,6- tetrahydropyridine , 1 ,4-dihydropyridine, piperazine, morpholine, thiomorpholine, pyran, 2,3- dihydropyran, tetrahydropyran, 1 ,4-dioxane, l,3-dioxane, 1,3 -dioxolane, homopiperazine, homopiperidine, l,3-dioxepane, 47-dihydro-l,3-dioxepin, and hexamethyleneoxide. The terms “C3-C7-heterocycloalkyl” includes but is not limited to tetrahydrofuran-2-yl, tetrahydrofuran-3- yl, 3-oxabicyclo[3.1.0]hexan-6-yl, 3 -azabicyclo[3.1.0]hexan-6-yl, tetrahydropyran-4-yl, tetrahydropyran-3 -yl, tetrahydropyran-2-yl, and azeti din-3 -yl. As used herein, the term "aromatic” refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character i.e. having (4n + 2) delocalized p(rΐ) electrons where n is an integer.

As used herein, the term“acy!”, employed alone or in combination with other terms, means, unless otherwise stated, to mean to an alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl group linked via a carbonyl group.

As used herein, the terms“carbamoyl” and“substituted carbamoyl”, employed alone or in combination with other terms, means, unless otherwise stated, to mean a carbonyl group linked to an amino group optionally mono or di-substituted by hydrogen, alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl. In some embodiments, the nitrogen substituents will be connected to form a heterocyclyl ring as defined above.

As used herein, the term "carboxy" and by itself or as part of another substituent means, unless otherwise stated, a group of formula C(=0)0H.

As used herein, the term“carboxyl ester” by itself or as part of another substituent means, unless otherwise stated, a group of formula C(=0)0X, wherein X is selected from the group consisting of Cl-C6-alkyl, C3 -C7 -cycloalkyl, and aryl.

As used herein the term“prodrug” represents a derivative of a compound of Formula I or Formula II or Formula III or Formula IV which is administered in a form which, once administered, is metabolised in vivo into an active metabolite also of Formula I or Formula II or Formula III or Formula IV.

Various forms of prodrug are known in the art. For examples of such prodrugs see: Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985) and Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al. (Academic Press, 1985); A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H. Bundgaard, Chapter 5“Design and Application of Prodrugs” by H. Bundgaard p. 113-191 (1991); H. Bundgaard, Advanced Drug Delivery Reviews 8, 1-38 (1992); H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77, 285 (1988); and N. Kakeya, et al., Chem. Pharm. Bull., 32, 692 (1984). Examples of prodrugs include cleavable esters of compounds of Formula I, II, III and IV. An in vivo cleavable ester of a compound of the invention containing a carboxy group is, for example, a pharmaceutically acceptable ester which is cleaved in the human or animal body to produce the parent acid. Suitable pharmaceutically acceptable esters for carboxy include C1-C6 alkyl ester, for example methyl or ethyl esters; C1-C6 alkoxymethyl esters, for example methoxymethyl ester; C1-C6 acyloxymethyl esters; phthalidyl esters; C3-C8 cycloalkoxycarbonyloxyC 1 -C6 alkyl esters, for example 1 -cyclohexylcarbonyloxyethyl; 1 -3-dioxolan-2-ylmethylesters, for example 5-methyl- 1 ,3-dioxolan-2-ylmethyl; C1-C6 alkoxycarbonyloxyethyl esters, for example 1 -methoxycarbonyloxyethyl; aminocarbonylmethyl esters and mono-or di-N-(Cl-C6 alkyl) versions thereof, for example N, N-dimethylaminocarbonylmethyl esters and N- ethylaminocarbonylmethyl esters; and may be formed at any carboxy group in the compounds of the invention.

An in vivo cleavable ester of a compound of the invention containing a hydroxy group is, for example, a pharmaceutically-acceptable ester which is cleaved in the human or animal body to produce the parent hydroxy group. Suitable pharmaceutically acceptable esters for hydroxy include Cl-C6-acyl esters, for example acetyl esters; and benzoyl esters wherein the phenyl group may be substituted with aminomethyl or N-substituted mono-or di-Cl-C6 alkyl aminomethyl, for example 4-aminomethylbenzoyl esters and 4-N,N- dimethylaminomethylbenzoyl esters.

Preferred prodrugs of the invention include acetyloxy and carbonate derivatives. For example, a hydroxy group of compounds of Formula I, II, III and IV can be present in a prodrug as -O-COR 1 or -0-C(0)0R 1 where R 1 is unsubstituted or substituted C1-C4 alkyl. Substituents on the alkyl groups are as defined earlier. Preferably the alkyl groups in R 1 is unsubstituted, preferable methyl, ethyl, isopropyl or cyclopropyl.

Other preferred prodrugs of the invention include amino acid derivatives. Suitable amino acids include a-amino acids linked to compounds of Formula I, II, III and IV via their C(0)0H group. Such prodrugs cleave in vivo to produce compounds of Formula I bearing a hydroxy group. Accordingly, such amino acid groups are preferably employed positions of Formula I, II, III and IV where a hydroxy group is eventually required. Exemplary prodrugs of this embodiment of the invention are therefore compounds of Formula I bearing a group of Formula -OC(O)- CH(NH2)R“ where R" is an amino acid side chain. Preferred amino acids include glycine, alanine, valine and serine. The amino acid can also be functionalised, for example the amino group can be alkylated. A suitable functionalised amino acid is N,N -dimethylglycine. Preferably the amino acid is valine.

Other preferred prodrugs of the invention include phosphoramidate derivatives. Various forms of phosphoramidate prodrags are known in the art. For example of such prodmgs see Serpi et al., Curr. Protoc. Nucleic Acid Chem. 2013, Chapter 15, Unit 15.5 and Mehellou et al., ChemMedChem, 2009, 4 pp. 1779-1791. Suitable phosphoramidates include (phenoxy)-a-amino acids linked to compounds of Formula I via their -OH group. Such prodrugs cleave in vivo to produce compounds of Formula I bearing a hydroxy group. Accordingly, such phosphoramidate groups are preferably employed positions of Formula I, II, III and IV where a hydroxy group is eventually required. Exemplary prodrugs of this embodiment of the invention are therefore compounds of Formula I, II, III and IV bearing a group of Formula -OP(0)(OR m )R lv where R m is alkyl, cycloalkyl, aryl or heteroaryl, and R 1V is a group of Formula -NH-CH(R v )C(0)OR vl . wherein R v is an amino acid side chain and R V1 is alkyl, cycloalkyl, aryl or heterocyclyl. Preferred amino acids include glycine, alanine, valine and serine. Preferably the amino acid is alanine. R v is preferably alkyl, most preferably isopropyl.

Subject matter of the present invention is also a method of preparing the compounds of the present invention. Subject matter of the invention is, thus, a method for the preparation of a compound of Formula I according to the present invention by reacting a compound of Formula V

V

in which Rl, R2, R3 and R4 are as above-defined, with a compound of Formula VI in which R5 and R6 are as above-defined.

Examples

The invention is now described with reference to the following Examples. These Examples are provided for the purpose of illustration only, and the invention is not limited to these Examples, but rather encompasses all variations that are evident as a result of the teachings provided herein.

The required substituted indole-2-carboxylic acids may be prepared in a number of ways; the main routes employed being outlined in Schemes 1-4. To the chemist skilled in the art it will be apparent that there are other methodologies that will also achieve the preparation of these intermediates.

Substituted indole-2-carboxylic acids can be prepared via the Hemetsberger-Knittel reaction (Organic Letters, 2011, 13(8) pp. 2012-2014, Journal of the American Chemical Society, 2007, pp. 7500-7501, and Monatshefte fur Chemie, 103(1), pp. 194-204) (Scheme 1).

Scheme 1: Indoles from vinyl azides

Substituted indoles may also be prepared using the Fischer method (Berichte der Deutschen Chemischen Gesellschaft. 17 (1): 559-568) (Scheme 2).

Scheme 2: The Fischer indole synthesis

A further method for the preparation of substituted indoles is the palladium catalysed alkyne annulation reaction (Journal of the American Chemical Society, 1991, pp. 6690-6692) (Scheme 3).

Scheme 3: Preparation of indoles via alkyne annulation

Additionally, indoles may be prepared from other suitably functionalized (halogenated) indoles (for example via palladium catalysed cross coupling or nucleophilic substitution reactions) as illustrated in Scheme 4.

Scheme 4: Palladium catalysed functionalization of halogenated indoles

Chemists skilled in the art will appreciate that other methods are available for the synthesis of suitably functionalized indole-2-carboxylic acids and activated esters thereof.

The HBV core protein modulators can be prepared in a number of ways. Schemes 5-11 illustrate the main routes employed for their preparation for the purpose of this application. To the chemist skilled in the art it will be apparent that there are other methodologies that will also achieve the preparation of these intermediates and Examples.

In a preferred embodiment compounds of Formula I can be prepared as shown in Scheme 5 below.

1 2

Step deprotection

3

4

Scheme 5: Synthesis of compounds of Formula I Compound 1 described in Scheme 5 is in step 1 reductively aminated (WO2009147188, WO2014152725) to obtain compounds with the general structure 2. Deprotection of the nitrogen protective group (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504), drawn as but not limited to Boc, e.g. with HC1 gives amine 3. An amide coupling in step 3 with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU results in compounds of Formula I.

In a preferred embodiment compounds of Formula III can be prepared as shown in Scheme 6 below.

Scheme 6: Synthesis of compounds Formula III

Compound 1 described in Scheme 6 is in step 1 acylated (P.N. Collier et al., J. Med. Chem., 2015, 58, 5684-5688, WO2016046530) to obtain compounds with the general structure 6. In step 2 a reduction e.g. with LiAlH 4 (W 02017040757), gives compounds of general structure 7. Deprotection of the nitrogen protective group (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504), drawn as but not limited to Boc, e.g. with HC1 gives amine 7. An amide coupling in step 3 with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU results in compounds of Formula III.

In a preferred embodiment compounds of Formula IV can be prepared as shown in Scheme 7 below.

Scheme 7: Synthesis of compounds of Formula IV

Compound 1 described in Scheme 7 is in step 1 reductively animated (WO2009147188, WO2014152725) to obtain compounds with the general structure 10. Deprotection of the nitrogen protective group (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504), drawn as but not limited to Boc, e.g. with HC1 gives amine 11. An amide coupling in step 3 with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU results in compounds of Formula IV.

In a preferred embodiment compounds of Formula II can be prepared as shown in Scheme 8 below.

Scheme 8: Synthesis of compounds of Formula II Compound 1 described in Scheme 8 is in step 1 sulfonylated (Jimenez-Somarribas et al., I. Med. Chem., 2017, 60, 2305-2325) to obtain compounds with the general structure 13. Deprotection of the nitrogen protective group (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504), drawn as but not limited to Boc, e.g. with HC1 gives amine 14 An amide coupling in step 3 with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU results in compounds of Formula II.

In a preferred embodiment compounds of Formula II can be prepared as shown in Scheme 9 below.

Scheme 9: Synthesis of compounds of Formula II

Compound 16 described in Scheme 9 is in step 1 deprotected (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504), drawn as but not limited to Boc, e.g. with HC1 gives amine 17. 17 is then amidated with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU to give compound 18. Deprotection of the nitrogen protective group (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504) drawn as, but not limited to Cbz, e.g. with ¾ and palladium on carbon gives amine 19, which can then be sulfonylated (Jimenez-Somarribas et al., J. Med. Chem., 2017, 60, 2305-2325) to obtain compounds of Formula II.

In a preferred embodiment compounds of Formula III can be prepared as shown in Scheme 10 below.

Scheme 10: Synthesis of compounds of Formula III

Compound 16 described in Scheme 10 is in step 1 deprotected (A. Isidro-Llobet et al., Chem Rev., 2009, 109, 2455-2504), drawn as but not limited to Boc, e.g. with HC1 gives amine 17. 17 is then amidated with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU to give compound 18. Deprotection of the nitrogen protective group (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504) drawn as, but not limited to Cbz, e.g. with H 2 and palladium on carbon gives amine 19, which can then be acylated (A. El- Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602) to obtain compounds of Formula III.

In a preferred embodiment compounds of Formula I can be prepared as shown in Scheme 11 below.

25

Scheme 11: Synthesis of compounds of Formula 1

Compound 22 (WO2012/170752) described in Scheme 11 is in step 1 aminated (Yang and Buchwald, Journal of Organometallic Chemistry, 1999, pp. 125-146) to give amine 23. 23 is then deprotected with methods known in literature (A. Isidro-Llobet et al., Chem. Rev., 2009, 109, 2455-2504) drawn as, but not limited to Boc, e.g. with HC1 to give amine 24, which can then be acylated with methods known in literature (A. El-Faham, F. Albericio, Chem. Rev. 2011, 111, 6557-6602), e.g. with HATU to give compounds of Formula I.

The following examples illustrate the preparation and properties of some specific compounds of the invention.

The following abbreviations are used:

A - DNA nucleobase adenine

ACN - acetonitrile

Ar - argon

BODIPY-FL - 4,4-difluoro-5,7 -dimethyl -4-bora-3 a, 4a-diaza-s-indacene-3 -propionic acid

(fluorescent dye)

Boc - tert-butoxycarbonyl

BnOH - benzyl alcohol

n-BuLi - n-butyl lithium ί-BuLi - t-butyl lithium

C - DNA nucleobase cytosine

CC 5 o - half-maximal cytotoxic concentration

C0 2 - carbon dioxide

CuCN - copper (I) cyanide

DCE - dichloroethane

DCM - dichloromethane

Dess-Martin periodinane - 1,1,1 -triacetoxy- 1 , 1 -dihydro- 1 ,2-benziodoxol-3 ( 1 H)-one

DIPEA - diisopropylethylamine

DIPE - di-isopropyl ether

DMAP - 4-dimethylaminopyridine

DMF - N,N -dimethylformamide

DMP - Dess-Martin periodinane

DMSO - dimethyl sulfoxide

DNA - deoxyribonucleic acid

DPP A - diphenylphosphoryl azide

DTT - dithiothreitol

EC 50 - half-maximal effective concentration

EDCI - N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride

Et 2 0 - diethyl ether

EtOAc - ethyl acetate

EtOH - ethanol

FL- - five prime end labled with fluorescein

NEt 3 - triethylamine

ELS - Evaporative Light Scattering

g - gram(s)

G - DNA nucleobase guanine

HBV - hepatitis B virus

HATU - 2-(l H-7 -azabenzotriazol- 1 -yl)-l ,1 ,3,3 -tetramethyl uronium hexafluorophosphate HC1 - hydrochloric acid

HEPES - 4-(2-hydroxyethyl)- 1 -piperazineethanesulfonic acid

HOAt - 1 -hydroxy-7-azabenzotriazole

HOBt - 1 -hydroxybenzotriazole

HPLC - high performance liquid chromatography ICso - half-maximal inhibitory concentration

LC640- - 3 prime end modification with fluorescent dye LightCycler® Red 640 LC/MS - liquid chromatography/mass spectrometry

LiAlH 4 - lithium aluminium hydride

LiOH - lithium hydroxide

MeOH - methanol

MeCN - acetonitrile

MgS0 4 - magnesium sulfate

mg - milligram(s)

min - minutes

mol - moles

mmol - millimole(s)

mL - millilitre(s)

MTBE - methyl tert-butyl ether

N 2 - nitrogen

Na 2 C0 3 - sodium carbonate

NaHC0 3 - sodium hydrogen carbonate

Na 2 S0 4 - sodium sulfate

Ndel - restriction enzyme recognizes CA A TATG sites

NEt 3 - triethylamine

NaH - sodium hydride

NaOH - sodium hydroxide

NH 3 - ammonia

NH 4 CI - ammonium chloride

NMR - nuclear magnetic resonance

PAGE - polyacrylamide gel electrophoresis

PCR - polymerase chain reaction

qPCR - quantitative PCR

Pd/C - palladium on carbon

-PH - 3 prime end phosphate modification

pTSA - 4-toluene-sulfonic acid

Rt - retention time

r.t. - room temperature

sat. - saturated aqueous solution SDS - sodium dodecyl sulfate

SI - selectivity index (= CC50/ EC50)

STAB - sodium triacetoxyborohydride

T - DNA nucleobase thymine

TBAF - tetrabutylammonium fluoride

TFA - trifluoroacetic acid

THF - tetrahydrofuran

TLC - thin layer chromatography

Tris - tris(hydroxymethyl)-aminomethane

Xhol - restriction enzyme recognizes C A TCGAG sites

Compound identification - NMR

For a number of compounds, NMR spectra were recorded using a Broker DPX400 spectrometer equipped with a 5 mm reverse triple-resonance probe head operating at 400 MHz for the proton and 100 MHz for carbon. Deuterated solvents were chloroform-d (deuterated chloroform, CDCI 3 ) or d6-DMSO (deuterated DMSO, d6-dimethylsulfoxide) . Chemical shifts are reported in parts per million (ppm) relative to tetramethylsilane (TMS) which was used as internal standard.

Compound identification - HPLC/MS

For a number of compounds, LC-MS spectra were recorded using the following analytical methods.

Method A

Column - Reverse phase Waters Xselect CSH Cl 8 (50x2. lmm, 3.5 micron)

Flow - 0.8 mL/min, 25 degrees Celsius

Eluent A - 95% acetonitrile + 5% lOmM ammonium carbonate in water (pH 9)

Eluent B - lOmM ammonium carbonate in water (pH 9)

Linear gradient t=0 min 5% A, t=3.5 min 98% A. t=6 min 98% A

Method B

Column - Reverse phase Waters Xselect CSH C18 (50x2. lmm, 3.5 micron)

Flow - 0.8 mL/min, 35 degrees Celsius

Eluent A - 0.1% formic acid in acetonitrile

Eluent B - 0.1% formic acid in water Linear gradient t=0 min 5% A, t=3.5 min 98% A. t=6 min 98% A

Method C

Column - Reverse phase Waters Xselect CSH C18 (50x2. lmm, 3.5 micron)

Flow - 1 mL/min, 35 degrees Celsius

Eluent A - 0.1% formic acid in acetonitrile

Eluent B - 0.1% formic acid in water

Linear gradient t=0 min 5% A, t=1.6 min 98% A. t=3 min 98% A Method D

Column - Phenomenex Gemini NX Cl 8 (50 x 2.0 mm, 3.0 micron)

Flow - 0.8 mL/min, 35 degrees Celsius

Eluent A - 95% acetonitrile + 5% lOmM ammoniumbicarbonate in water

Eluent B - lOmM ammoniumbicarbonate in water pH=9.0

Linear gradient t=0 min 5% A, t=3.5 min 98% A. t=6 min 98% A

Method E

Column - Phenomenex Gemini NX Cl 8 (50 x 2.0mm, 3.0 micron)

Flow - 0.8 mL/min, 25 degrees Celsius

Eluent A - 95% acetonitrile + 5% lOmM ammoniumbicarbonate in water

Eluent B - lOmM ammonium bicarbonate in water (pH 9)

Linear gradient t=0 min 5% A, t=3.5 min 30% A. t=7 min 98% A, t=10 min 98% A

Method F

Column - Waters XSelect HSS C18 (150 x 4.6mm, 3.5 micron)

Flow— 1.0 mL/min, 25 degrees Celsius

Eluent A - 0.1% TFA in acetonitrile

Eluent B - 0.1% TFA in water

Linear gradient t=0 min 2% A, t=l min 2% A, t=15 min 60% A, t=20 min 60% A

Method G

Column - Zorbax SB-C18 1.8 pm 4.6x15mm Rapid Resolution cartridge (PN 821975-932) Flow - 3 mL/min

Eluent A - 0.1% formic acid in acetonitrile Eluent B - 0.1% formic acid in water

Linear gradient t=0 min 0% A, t=l .8 min 100% A

Method H

Column - Waters Xselect CSH Cl 8 (50x2. lmm, 2.5 micron)

Flow - 0.6 mL/min

Eluent A - 0.1% formic acid in acetonitrile

Eluent B - 0.1 % formic acid in water

Linear gradient t=0 min 5% A, t=2.0 min 98% A, t=2.7 min 98% A

Method J

Column - Reverse phase Waters Xselect CSH Cl 8 (50x2. lmm, 2.5 micron)

Flow - 0.6 mL/min

Eluent A - 100% acetonitrile

Eluent B - lGmM ammonium bicarbonate in water (pH 7.9)

Linear gradient t=0 min 5% A, t=2.0 min 98% A, t=2.7 min 98% A

Preparation of 4-chIoro-7-fluoro-lH-indole-2-carboxylic acid

Step A: A mixture of compound 1-HC1 (17.0 g, 86.2 mmol), sodium acetate (7.10 g, 86.6 mmol), and ethyl pyruvate (10.0 g, 86.1 mmol) in ethanol (100 mL) was refluxed for lh, cooled to r.t, and diluted with water (100 mL). The precipitated solid was collected by filtration and dried to obtain 20.0 g (77.3 mmol, 90%) of compound 2 as a mixture of cis- and trans- isomers. Step B: A mixture of compound 2 (20.0 g, 77.3 mmol), obtained in the previous step, and BF 3 -Et 2 0 (50.0 g, 352 mmol) in acetic acid (125 mL) was refluxed for l8h and evaporated under reduced pressure. The residue was mixed with water (100 mL) and extracted with MTBE (2x 50 mL). The combined organic extracts were dried over Na 2 S0 4 and evaporated under reduced pressure. The residue was purified by silica gel column chromatography to give 3.00 g (12.4 mmol, 16%) of compound 3.

Step C: A mixture of compound 3 (3.00 g, 12.4 mmol) and NaOH (0.500 g, 12.5 mmol) in ethanol (30 mL) was refluxed for 30 min and evaporated under reduced pressure. The residue was mixed with water (30 mL) and the insoluble material was filtered off. The filtrate was acidified with concentrated hydrochloric acid (5 mL). The precipitated solid was collected by filtration, washed with water (3 mL), and dried to obtain 2.41 g (11.3 mmol, 91%) of 4-chloro-7- fluoro- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.24 mins, m/z 212 [M-H]

Preparation of 7-fluoro-4-methyl-lH-indole-2-carboxylic acid

Step D: To a solution of sodium methoxide (21.6 g, 400 mmol) in methanol (300 mL) at at - 10°C was added dropwise a solution of compound 4 (26.4 g, 183 mmol) and compound 5 (59.0 g, 457 mmol) in methanol (100 mL). The reaction mass was stirred for 3 h maintaining temperature below 5°C and then quenched with ice water. The resulting mixture was stirred for 10 min, filtered, and washed with water to afford 35.0 g (156 mmol, 72%) of compound 6 as a white solid.

Step E: A solution of compound 6, obtained in the previous step, (35.0 g, 156 mmol) in xylene (250 mL) was refluxed for lh under an argon atmosphere and then evaporated under reduced pressure. The residue was recrystallized form hexane-ethyl acetate mixture (60:40) to give 21.0 g (103 mmol, 60%) of compound 7.

Step F: To a solution of compound 7 (21.0 g, 101 mmol) in ethanol (200 mL) was added 2 N aqueous sodium hydroxide solution (47 mL). The mixture was stirred for 2h at 60°C. The solvent was evaporated and the residue was acidified with aqueous hydrochloric acid to pH 5-6. The resulting precipitate was filtered, washed with water, and dried to obtain 18.0 g (93.2 mmol, 92%) of 7-fluoro-4-methyl- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.12 mins, m/z 192 [M-H]

Preparation of 6,7-difluoro-lH-indole-2-carboxylic acid

Step G: A mixture of compound 8 (5.00 g, 34.7 mmol), acetic acid (1 mL), and ethyl pyruvate (5.00 g, 43.1 mmol) in ethanol (20 mL) was refluxed for lh, cooled to r.t, and diluted with water (20 mL). The precipitated solid was collected by filtration and dried to obtain 5.50 g (22.7 mmol, 66%) of compound 9 as a mixture of cis- and trans- isomers.

Step H: A mixture of compound 9 (5.50 g, 22.7 mmol), obtained in the previous step, and BF 3 -Et 2 0 (10.0 g, 70.5 mmol) in acetic acid (25 mL) was refluxed for l8h and evaporated under reduced pressure. The residue was mixed with water (30 mL) and extracted with MTBE (2x 30 mL). The combined organic extracts were dried over Na 2 S0 4 and evaporated under reduced pressure. The residue was purified by silica gel column chromatography to give 0.460 g (2.04 mmol, 9%) of compound 10. Step I: A mixture of compound 10 (0.450 g, 2.00 mmol) and NaOH (0.100 g, 2.50 mmol) in ethanol (10 mL) was refluxed for 30 min and evaporated under reduced pressure. The residue was mixed with water (10 mL) and the insoluble material was filtered off. The filtrate was acidified with concentrated hydrochloric acid (1 mL). The precipitated solid was collected by filtration, washed with water (3 mL), and dried to obtain 0.38 g (1.93 mmol, 95%) of 6,7- difluoro- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.10 mins, m/z 196 [M-H]

Preparation of 4-cyano-lH-indole-2-carboxylic acid

Step J: To a stirred solution of compound 11 (5.00 g, 19.7 mmol) in DMF (50 mL) was added

CuCN (3.00 g, 33.5 mmol). The mixture was stirred for 4h at 150°C. The mixture was then cooled to r.t, and water (100 mL) added. The resulting mixture was extracted with ethyl acetate (4x 100 mL). The combined organic extracts were washed with water (50 mL) and brine (50 mL), dried over Na 2 S0 4 , and evaporated under reduced pressure to give 2.50 g (12.5 mmol, 63%) of compound 12, pure enough for the next step.

Step K: To a solution of compound 12 (2.50 g, 12.5 mmol) in ethanol (30 mL) was added LiOH-¾0 (0.600 g, 13.0 mmol). The mixture was refluxed for lOh. The solvent was evaporated under reduced pressure and the residue diluted with water (50 mL). The aqueous layer was acidified to pH 6 with 10% aq. hydrochloric acid and the precipitated solid was collected by filtration. The residue was washed with water and dried under vacuum to afford 1.20 g (6.45 mmol, 52%) of 4-cyano- 1 H-indole-2-carboxylic acid as a white solid.

Rt (Method G) 1.00 mins, m/z 197 [M+H] +

Preparation of 4-cyano-7-fluoro-lH-indole-2-carboxyIic acid

Step L: To a stirred solution of compound 13 (5.00 g, 18.4 mmol) in DMF (50 mL) was added CuCN (2.80 g, 31.2 mmol). The mixture was stirred for 4h at l50°C. The mixture was then cooled to r.t., and water (100 mL) added. The resulting mixture was extracted with ethyl acetate (4x 100 mL). The combined organic extracts were washed with water (50 mL) and brine (50 mL), dried over Na 2 S0 4 , and evaporated under reduced pressure to give 1.50 g (6.87 mmol, 37%) of compound 14, pure enough for the next step.

Step M: To a solution of compound 14 (1.50 g, 6.87 mmol) in ethanol (20 mL) was added o LiOH H 2 0 (0.400 g, 9.53 mmol). The mixture was refluxed for lOh. The solvent was evaporated under reduced pressure and the residue diluted with water (40 mL). The aqueous layer was acidified to pH 6.0 with 10% aq. hydrochloric acid and the precipitate was collected by filtration. The residue was washed with water and dried under vacuum to afford 0.400 g (1.95 mmol, 28%) of 4-cyano-7-fluoro- 1 H-indole-2-carboxylic acid as a white solid.

c

Rt (Method G) 1.02 mins, m/z 203 [M-H]

Preparation of 4-cyano-5-fluoro-lH-indole-2-carboxylic acid

Step N: To a solution of compound 15 (5.00 g, 19.4 mmol) in DMF (50 mL) was added NaHCOs (1.59 g, 18.9 mmol) and iodomethane (3 mL). The resulting mixture was stirred overnight at r.t, then diluted with water (50 mL) and extracted with diethyl ether (3x 50 xnL). The combined organic extracts were dried over Na 2 S0 4 , and evaporated under reduced pressure to obtain 4.90 g (18.0 mmol, 90%) of compound 16 as white solid.

Step O: To a stirred solution of compound 16 (4.80 g, 17.6 mmol) in DMF (50 mL) was added CuCN (2.70 g, 30.1 mmol). The mixture was stirred for 4h at l50°C. The mixture was then cooled to r.t, water (100 mL) added. The resulting mixture was extracted with ethyl acetate (4x 100 mL). The combined organic extracts were washed with water (50 mL) and brine (50 mL), dried over Na 2 S0 4 , and evaporated under reduced pressure to give 1.40 g (6.42 mmol, 36%) of compound 17, pure enough for the next step.

Step P: To a solution of compound 17 (1.40 g, 6.42 mmol) in ethanol (20 mL) was added LiOH-H 2 0 (0.350 g, 8.34 mmol). The mixture was refluxed for lOh. The solvent was evaporated under reduced pressure and the residue diluted with water (30 mL). The aqueous layer was acidified to pH 6.0 with 10% aq. hydrochloric acid and the precipitate collected by filtration. The residue was washed with water and dried under vacuum to afford 0.500 g (2.45 mmol, 38%) of 4-cyano-5-fluoro- 1 H-indole-2-carboxylic acid as a white solid.

Rt (Method G) 1.10 mins, m/z 203 [M-H]

Preparation of 4,5,6-trifluoro-lH-indole-2-carboxylic acid

Step Q: To a solution of sodium methoxide (23.0 g, 426 mmol) in methanol (200 mL) at -10°C was added dropwise a solution of compound 18 (15.0 g, 93.7 mmol) and compound 5 (26.0 g, 201 mmol) in methanol (100 mL). The reaction mixture was stirred for 3h, maintaining the temperature below 5°C and then quenched with ice water. The resulting mixture was stirred for 10 min, and the precipitate collected by filtration. The solid was washed with water and dried to afford 12 mmol, 72%) of compound 19 as a white solid.

Step R: A solution of compound 19, obtained in the previous step, (12.0 g, 46.7 mmol) in xylene (250 mL) was refluxed for lh under an argon atmosphere and then evaporated under reduced pressure. The residue was recrystallized form hexane-ethyl acetate mixture (60:40) to give 7.00 g (30.5 mmol, 65%) of compound 20.

Step S: To a solution of compound 20 (7.00 g, 30.5 mmol) in ethanol (50 mL) was added 2 N aqueous sodium hydroxide solution (18 mL). The mixture was stirred for 2h at 60°C. The solvent was evaporated and the residue was acidified to pH 5-6 with aqueous hydrochloric acid. The resulting precipitate was collected by filtration, washed with water, and dried to obtain 5.00 g (23.2 mmol, 76%) 4,5,6-trifluoro-lH-indole-2-carboxylic acid.

1H NMR (400 MHz, d6-dmso) 7.17 (1H, s), 7.22 (1H, dd), 12.3 (1H, br s), 13.3 (1H, br s)

Preparation of 4,6,7-trifluoro-lH-indole-2-carboxylic acid

Step T: To a solution of sodium methoxide (23.0 g, 426 mmol) in methanol (200 mL) at -l0°C was added dropwise a solution of compound 21 (15.0 g, 90.3 mmol) and compound 5 (26.0 g, 201 mmol) in methanol (100 mL). The reaction mixture was stirred for 3h maintaining the temperature below 5°C and then quenched with ice water. The resulting mixture was stirred for 10 min. The precipitate was collected by filtration, washed with water and dried to afford 10.0 g (38.0 mmol, 42%) of compound 22 as a white solid.

Step U: A solution of compound 22, obtained in the previous step, (10.0 g, 38.0 mmol) in xylene (200 mL) was refluxed for lh under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized form hexane-ethyl acetate mixture (60:40) to give 6.00 g

(26.2 mmol, 69%) of compound 23.

Step V; To a solution of compound 23 (7.00 g, 30.5 mmol) in ethanol (40 mL) was added 2 N aqueous sodium hydroxide solution (16 mL). The mixture was stirred for 2h at 60°C. The solvent was evaporated and the residue was acidified to pH 5-6 with aqueous hydrochloric acid. The resulting precipitate was collected by filtration, washed with water, and dried to obtain 4.10 g (19.1 mmol, 62%) of 4,6,7-trifluoro- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.16 mins, m/z 214 [M-H]

Preparation of 4-cyano-6-fluoro-lH-indole-2-carboxylic acid

a

27

Step W: To a solution of sodium methoxide (65.0 g, 1203 mmol) in methanol (500 mL) at -l0°C was added dropwise a solution of compound 24 (60.0 g, 296 mmol) and compound 5 (85.0 g, 658 mmol) in methanol (200 mL). The reaction mixture was stirred for 3h maintaining the temperature below 5°C and then quenched with ice water. The resulting mixture was stirred for 10 min. The precipitate was collected by filtration, washed with water and dried to afford 45.0 g (143 mmol, 48%) of compound 25.

Step X: A solution of compound 25, obtained in the previous step, (35.0 g, 111 mmol) in xylene (250 mL) was refluxed for lh under an argon atmosphere and then evaporated under reduced pressure. The residue was recrystallized form hexane-ethyl acetate mixture (60:40) to give 11.0 g (38.4 mmol, 35%) of compound 26. Step Y: To a stirred solution of compound 26 (11.0 g, 38.4 mmol) in DMF (20 mL) was added CuCN (6.60 g, 73.7 mmol). The mixture was stirred for 4h at 150°C. The mixture was then cooled to r.t, and water (70 mL) added. The mixture was extracted with ethyl acetate (4x 50 mL). The combined organic extracts were washed with water (50 mL) and brine (50 mL), dried over Na 2 S0 4 , and evaporated under reduced pressure to give 2.40 g (10.3 mmol, 27%) of compound 27, pure enough for the next step.

Step Z: To a solution of compound 27 (2.40 g, 6.42 mmol) in ethanol (30 mL) was added LiOH-H 2 0 (0.600 g, 14.3 mmol). The mixture was refluxed for lOh. The mixture was concentrated under reduced pressure and the residue diluted with water (50 mL). The aqueous layer was acidified to pH 6 with 10% aq. hydrochloric acid and the precipitate was collected by filtration. The solid was washed with water and dried under vacuum to afford 1.20 g (5.88 mmol, 57%) of 4-cyano-6-fluoro- 1 H-indole-2-carboxylic acid as a white solid.

Rt (Method G) 1.06 mins, m/z 203 [M-H]

Preparation of 4-ethyi-lH-mdole-2-carboxylic acid

32

Step AA: A solution of compound 28 (70.0 g, 466 mmol) in dry THF (500 mL) was treated with 10 M solution of BH 3 in THF (53 mL, 53.0 mmol of BH 3 ) at 0°C. The reaction mass was stirred at r.t. for 24h before methanol (150 mL) was slowly added thereto. The resulting mixture was stirred for 45 min, and evaporated under reduced pressure to yield 55.0 g (404 mmol, 87%) of compound 29, pure enough for the next step.

Step AB: To a cooled (0°C) solution of compound 29 (55.0 g, 404 mmol) in CH 2 C1 2 (400 mL) was added Dess-Martin periodinane (177 g, 417 mmol) portionwise. After stirring for lh at r.t., the reaction mixture was quenched with saturated aqueous Na 2 S 2 0 3 (300 mL) and saturated aqueous NaHC0 3 (500 mL). The mixture was extracted with CH 2 Cl 2 (3x 300 mL). The combined organic extracts were washed with water and brine, dried over Na 2 S0 4 and concentrated to yield 51.0 g of crude compound 30 as a yellow solid.

Step AC: To a solution of sodium methoxide (107 g, 1981 mmol) in methanol (600 mL) at -l0°C was added dropwise a solution of compound 30, obtained in the previous step, (51.0 g) and compound 5 (126 g, 976 mmol) in methanol (300 mL). The reaction mixture was stirred for 4h maintaining temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min, and the precipitate collected by filtration. The solid was washed with water and dried to afford 35.0 g (151 mmol, 37% over 2 steps) of compound 31.

Step AD: A solution of compound 31 , obtained in the previous step, (35.0 g, 151 mmol) in xylene (500 mL) was refluxed for lh under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized form hexane-ethyl acetate mixture (60:40) to give 21.0 g (103 mmol, 68%) of compound 32.

Step AE: To a solution of compound 32 (21.0 g, 103 mmol) in ethanol (200 mL) was added 2 N aqueous sodium hydroxide solution (47 mL). The mixture was stirred for 2h at 60°C. The mixture was concentrated under reduced pressure, and the residue acidified to pH 5-6 with aqueous hydrochloric acid. The precipitate was collected by filtration, washed with water, and dried to obtain 19 g (100 mmol, 97%) of 4-ethyl- 1 H-indole-2-carboxylic acid.

33 35

34 Step AF: To a degassed suspension of compound 33 (2,00 g, 7.80 mmol), cyclopropylboroni c add (0.754 g, 8.78 mmol), K3PO4 (5.02 g, 23,6 mmol), tricyclohexyl phosphine (0.189 g, 0,675 mmol), and water (2.0 mL) in toluene (60.0 mL) was added palladium (II) acetate (0 076 g, 0.340 mmol). The reaction mixture was stirred at 100°C for 4k The reaction progress was monitored by diluting an aliquot of the reaction mixture with water and extracting with ethyl acetate. The organic layer was spoted over an analytical silica gel TLC plate and visualized using 254 nm IJV light. The reaction progressed to completion with the formation of a polar spot. The R f values of the starting material and product were 0.3 and 0.2, respectively. The reaction mixture was allowed to cool to r.t. and filtered through a pad of celite. The filtrate was concentrated under reduced pressure and the crude product was purified by flash column using 230-400 mesh silica gel and eluted with 10% ethyl acetate in petroleum ether to afford 1.10 g (5.11 mmol, 63%) of compound 34 as a brown liquid. TLC system: 5% ethyl acetate in petroleum ether.

Step AG: A mixture of compound 34 (1.10 g, 5.11 mmol) in ethanol (40 mL) and 2 N aqueous sodium hydroxide (15 mL) was stirred for 2h at 60°C The mixture was concentrated under reduced pressure, and the residue acidified to pH 5-6 with aqueous hydrochloric acid. The precipitate was collected by filtration, washed with water, and dried to yield 1.01 g (5.02 mmol, 92%) of 4-cyclopropyl- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.17 mins, m/z 200 [M-H]

Preparation of 4-chIoro-5-fIuoro-lH-indole-2-carboxylic acid

Step AH: To a solution of sodium methoxide (39.9 g, 738 mmol) in methanol (300 mL) at -l0°C was added dropwise a solution of compound 36 (28.8 g, 182 mmol) and methyl azidoacetate (52.1 g, 404 mmol) in methanol (150 mL). The reaction mixture was stirred for 3h maintaining temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for

10 min. The precipitate was collected by filtration, washed with water and dried to afford 20.0 g (78.2 mmol, 43%) of compound 37.

Step AI: A solution of compound 37 (19.4 g, 76.0 mmol) in xylene (250 mL) was refluxed for lh under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized from hexane-ethyl acetate (50:50) to give 9.00 g (39.5 mmol, 52%) of compound 38. Step AJ: To a solution of compound 38 (8.98 g, 39.4 mmol) in ethanol (100 mL) was added 2 N aqueous sodium hydroxide solution (18 mL). The mixture was stirred for 2h at 60°C. The mixture was concentrated under reduced pressure, and the residue acidified to pH 5-6 with aqueous hydrochloric acid. The resulting precipitate was collected by filtration, washed with water, and dried to obtain 7.75 g (36.3 mmol, 92%) of 4-chloro-5-fluoro-lH-indole-2-carboxylic acid.

Rt (Method G) 1.15 mins, m/z 212 [M-H]

1H NMR (400 MHz, d6-dmso) 7.08 (1H, s), 7.28 (1H, dd) 7.42 (1H, dd), 12.2 (1H, hr s), 13.2 (1H, hr s)

Preparation of 5-fluoro-4-(l-hydroxyethyl)-lH-indole-2-carboxylic acid

Step AK: To a solution of sodium methoxide (50.0 g, 926 mmol) in methanol (300 mL) at -l0°C was added dropwise a solution of compound 39 (45.0 g, 222 mmol) and methyl azidoacetate (59.0 g, 457 mmol) in methanol (100 mL). The reaction mixture was stirred for 3 h maintaining the temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min. The precipitate was collected by filtration, washed with water and dried to afford 35.0 g (133 mmol, 60%) of compound 40 as a white solid.

Step AL: A solution of compound 40, obtained in the previous step, (35.0 g, 133 mmol) in xylene (250 mL) was refluxed for 1 h under an argon atmosphere and then evaporated under reduced pressure. The residue was recrystallized from hexane-ethyl acetate (60:40) to give 21.0 g (77.2 mmol, 58%) of compound 41.

Step AM: To a degassed solution of compound 41 (4.00 g, 14.7 mmol) and tributyl(l- ethoxyvinyl)stannane (5.50 g, 15.2 mmol) in toluene (50 mL) under nitrogen was added bis(triphenylphosphine) palladium(II) dichloride (1.16 g, 1.65 mmol). The reaction mixture was stirred at 60°C for 20 h. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated under under reduced pressure and the residue purified by silica gel chromatography to afford 2.50 g (9.50 mmol, 65%) of compound 42 as a pale yellow solid. Step AN: To a solution of compound 42 (2.40 g, 9.12 mmol) in l,4-dioxane (30 mL) was added 2M hydrochloric acid (15 mL). The resulting mixture was stirred at room temperature for 30 min. The mixture was concentrated under vacuum and the residue partitioned between ethyl acetate and water. The organic extract was washed with water and brine, dried over sodium sulfate, filtered, and evaporated. The residue was triturated with 5% ether in isohexane and dried to afford 1.80 g (7.65 mmol, 84%) of compound 43 as a white solid.

Step AO: A suspension of compound 43 (1.70 g, 7.23 mmol) and NaBH 4 (2.50 g, 66.1 mmol) in ethanol (13 mL) was refluxed for 2 h, then cooled to room temperature, and filtered. The filtrate was concentrated under reduced pressure and the residue dissolved in ethyl acetate. The solution was washed with 1N hydrochloric acid and brine, dried over Na 2 S0 4 , and evaporated under reduced pressure to give 1.60 g (6.74 mmol, 93%) of compound 44 as a colourless oil.

Step AP: To a solution of compound 44 (1.50 g, 6.32 mmol) in methanol (40 mL) was added 2N aqueous NaOH (10 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated under reduced pressure and the residue acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 x 15 mL), and dried to obtain 1.30 g (5.82 mmol, 92%) of 5-fluoro-4-( 1 -hydroxyethyl)- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.00 mins, m/z 222 [M-H]

Preparation of 4-ethyl-5-fluoro-lH-indole-2-carboxylic acid

Step AQ: To a heated (90°C) solution of compound 41 (4.00 g, 14.7 mmol) in anhydrous DMF under nitrogen (10 mL) were added tri-n-butyl(vinyl)tin (3.60 g, 11.4 mmol) and Pd(PPh 3 ) 2 Cl 2 (0.301 g, 0.757 mmol). The resulting mixture was stirred at 90°C for 1 h. The mixture was then cooled to room temperature and purified by silica gel column chromatography (60-80% ethyl acetate in hexane) to give 2.20 g (10.0 mmol, 68%) of compound 45 as yellow solid.

Step AR: A mixture of compound 45 (1.50 g, 6.84 mmol) and Pd/C (0.300 g, 10% wt.) in methanol (20 mL) was stirred under an atmosphere of hydrogen at room temperature for 16 h. The mixture was filtered, then concentrated under reduced pressure to give 1.45 g (6.55 mmol, 96%) of compound 46.

Step AS: To a solution of compound 46 (1.40 g, 6.33 mmol) in methanol (40 mL) was added 2N aqueous NaOH (10 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated under vacuum, then the residue was acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 x 15 mL), and dried to obtain 1.20 g (5.79 mmol, 91%) of target compound 4-ethyl-5-fluoro-lH-indole-2-carboxylic add. Rt (Method G) 1.33 mins, mfz 206 [M-H]

Preparation of 4-ethyl-6-fluoro-lH-mdole-2-carboxylic acid

Step AT: To a solution of sodium metlioxide (50.0 g, 926 mmol) in methanol (300 mL) at -l0°C was added dropwise a solution of compound 47 (45.0 g, 202 mmol) and methyl azidoacetate (59.0 g, 457 mmol) in methanol (100 mL). The reaction mixture was stirred for 3 h maintaining temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min. The precipitate was collected by filtration, washed with water and dried to afford 38.5 g (128 mmol, 63%) of compound 48 as a white solid. Step AU: A solution of compound 48, obtained in the previous step, (38.5 g, 128 mmol) in xylene (250 mL) was refluxed for 1 h under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized hexane-ethyl acetate (60:40) to give 18.0 g (67.3 mmol, 53%) of compound 49.

Step AV: To a heated (90°C) solution of compound 49 (4.00 g, 14.7 mmol) in anhydrous DMF under nitrogen (10 mL) were added tri-n-butyl(vinyl)tin (3.60 g, 11.4 mmol) and Pd(PPh 3 ) 2 Cl 2 (0.301 g, 0.757 mmol). The resulting mixture was stirred at 90°C for 1 h. The mixture was then cooled to room temperature and purified by silica gel column chromatography (60-80% ethyl acetate in hexane) to give 2.00 g (9.12 mmol, 62%) of compound 50 as yellow solid.

Step AW: A mixture of compound 50 (1.50 g, 6.84 mmol) and Pd/C (0.300 g, 10% wt.) in methanol (20 mL) was stirred under an atmosphere of hydrogen at room temperature for 16 h. The mixture was filtered and concentrated to give 1.40 g (6.33 mmol, 93%) of compound 51.

Step AX: To a solution of compound 51 (1.10 g, 4.97 mmol) in methanol (40 mL) was added 2N aqueous NaOH (10 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated under reduced pressure, then acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 x 15 mL), and dried to obtain 0.900 g (4.34 mmol, 87%) of target compound 4-ethyl-6-fluoro- 1 H-indole-2 -carboxylic acid.

Rt (Method G) 1.29 mins, m/z 206 [M-H]

Preparation of 6-fluoro-4-(l-hydroxyethyl)-lH-indole-2-carboxyIic acid

54 Step AY: To a degassed solution of compound 49 (4.00 g, 14.7 mmol) and tributyl(l- ethoxyvinyl)stannane (5.50 g, 15.2 mmol) in toluene (50 mL) under nitrogen were added bis(triphenylphosphine) palladium(II) di chloride (1.16 g, 1.65 mmol). The reaction mixture was stirred at 60°C for 20 h. The reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure and the residue purified by silica gel chromatography to give 2.10 g (7.98 mmol, 54%) of compound 52 as a pale yellow solid.

Step AZ: To a solution of compound 52 (2.10 g, 7.98 mmol) in 1,4-dioxane (30 mL) was added 2M hydrochloric acid (15 mL). The resulting mixture was stirred at room temperature for 30 min. The mixture was concentrated under reduced pressure, and residue partitioned between ethyl acetate and water. The organic extract was washed with water and brine, dried over sodium sulfate, filtered, and concentrated. The residue was triturated with 5% ether in isohexane and dried to afford 1.70 g (7.23 mmol, 91%) of compound 53 as a white solid.

Step BA: A suspension of compound 53 (1.70 g, 7.23 mmol) and NaBH 4 (2.50 g, 66.1 mmol) in ethanol (13 mL) was refluxed for 2 h, cooled to room temperature, and filtered. The filtrate was concentrated under reduced pressure and the residue was dissolved in ethyl acetate. The solution was washed with IN hydrochloric acid and brine, dried over Na 2 S0 4 , and concentrated under reduced pressure to give 1.60 g (6.74 mmol, 93%) of compound 54 as a colourless oil.

Step BB: To a solution of compound 54 (1.40 g, 5.90 mmol) in methanol (40 mL) was added 2N aqueous NaOH (10 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated and the residue acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 x 15 mL), and dried to obtain 1.10 g (4.93 mmol, 48%) of target compound 6-fluoro-4-(l-hydroxyethyl)-lH-indole-2-carboxylic acid.

Rt (Method G) 1.00 mins, m/z 222 [M-H]

Preparation of 4-ethyl-7-fluoro-lH-indoIe-2-carboxylic acid

Step BC: To a solution of sodium methoxide (50.0 g, 926 mmol) in methanol (300 mL) -l0°C was added dropwise a solution of compound 55 (45.0 g, 222 mmol) and methyl azidoacetate (59.0 g, 457 mmol) in methanol (100 mL). The reaction mixture was stirred for 3 h maintaining temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min. The precipitate was collected by filtration, washed with water and dried to afford 33.0 g (110 mmol, 50%) of compound 56 as a white solid.

Step BD: A solution of compound 56, obtained in the previous step, (33.0 g, 110 mmol) in i() xylene (250 mL) was refluxed for 1 h under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized from hexane-ethyl acetate (60:40) to give 21.5 g (79.0 mmol, 72%) of compound 57.

Step BE: To a heated (90°C) solution of compound 57 (4.00 g, 14.7 mmol) in anhydrous DMF under nitrogen (10 mL) were added tri-n-butyl(vinyl)tin (3.60 g, 11.4 mmol) and Pd(PPh 3 ) 2 Cl 2 (0.301 g, 0.757 mmol). The resulting mixture was stirred at 90°C for 1 h. The mixture was cooled to room temperature and purified by silica gel column chromatography (60-80% EtOAc in hexane). The combined product fractions of the product were concentrated, washed with water (3 x 100 mL), dried over Na 2 S0 4 , and concentrated to give 1.80 g (8.21 mmol, 56%) of compound 58 as yellow solid.

Step BF: A mixture of compound 58 (1.50 g, 6.84 mmol) and Pd/C (0.300 g, 10% wt.) in methanol (20 mL) was stirred under atmosphere of hydrogen at room temperature for 16 h. The mixture was filtered and concentrated to give 1.25 g (5.65 mmol, 83%) of compound 59. Step BG: To a solution of compound 59 (1.40 g, 6.33 mmol) in methanol (40 mL) was added 2N aqueous NaOH (10 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated under reduced pressure, and the residue acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 x 15 mL), and dried to obtain 1.25 g (6.03 mmol, 95%) of target compound 4-ethyl-7-fluoro- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.27 mins, m/z 206 [M-H]

Preparation of 7-fluoro-4-(l-hydroxyethyl)-lH-indole-2-carboxylic acid

Step BH: To a degassed solution of compound 57 (4.00 g, 14.7 mmol) and tributyl(l- ethoxyvinyl)stannane (5.50 g, 15.2 mmol) in toluene (50 mL) under nitrogen was added bis(triphenylphosphine) palladium(II) dichloride (1.16 g, 1.65 mmol). The reaction mixture was stirred at 60°C for 20 h. The mixture was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure and the residue purified by silica gel chromatography to afford 2.70 g (10.3 mmol, 70%) of compound 60 as a pale yellow solid.

Step BI: To a solution of compound 60 (2.40 g, 9.12 mmol) in l,4-dioxane (30 mL) was added 2M hydrochloric acid (15 mL). The mixture was stirred at room temperature for 30 min. The majority of the solvent was evaporated and the residue was partitioned between ethyl acetate and water. The combined organic extracts were washed with water and brine, dried over sodium sulfate, filtered, and evaporated. The residue was triturated with 5% ether in isohexane and dried to afford 1.90 g (8.08 mmol, 86%) of compound 61 as a white solid.

Step BJ: A suspension of compound 61 (1.70 g, 7.23 mmol) and NaBH 4 (2.50 g, 66.1 mmol) in ethanol (13 mL) was refluxed for 2 h, cooled to room temperature, and filtered. The filtrate was evaporated under reduced pressure and the residue was dissolved in ethyl acetate. The solution was washed with 1N hydrochloric acid and brine, dried over Na 2 S0 4 , and evaporated under reduced pressure to give 1.50 g (6.32 mmol, 87%) of compound 62 as a colourless oil.

Step BK: To a solution of compound 62 (1.50 g, 6.32 mmol) in methanol (40 mL) was added 2N aqueous NaOH (10 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated under reduced pressure and the residue acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 c 15 mL), and dried to obtain 1.35 g (6.05 mmol, 96%) of target compound 7-fluoro-4-( 1 -hydroxyethyl)- 1 H-indole-2-carboxylic acid.

Rt (Method G) 0.90 mins, m/z 222 [M-H]

Preparation of 4-(hydroxymethyl)-lH-indole-2-carboxylic acid

65

Step BL: To a solution of compound 33 (10.0 g, 39.4 mmol) in a mixture of dioxane (200 mL) and water (50 mL) were added potassium vinyltrifluoroborate (11.0 g, 82.1 mmol), triethylamine (30 mL, 248 mmol) and Pd(dppf)Cl 2 (1.0 g, 1.37 mmol). The mixture was stirred at 80°C for 48h. The mixture was concentrated under vacuum, and the residue was dissolved in ethyl acetate. The solution was washed with water and concentrated under reduced pressure. The obtained material was purified by silica gel column chromatography to give 2.50 g (12.4 mmol, 38%) of compound 63.

Step BM: To a mixture of compound 63 (2.50 g, 12.4 mmol), acetone (200 mL), and water (40 mL) were added Os0 4 (0.100 g, 0.393 mmol) and NaI0 4 (13.4 g, 62.6 mmol). The reaction was stirred for 10 h at room temperature. The acetone was distilled off and the remaining aqueous solution extracted with dichloromethane. The organic layer was washed with saturated NaHCQ 3 solution (2 x 50 ml) and brine (2 x 50 mL), dried over Na 2 S04, and concentrated under reduced pressure to obtain 1.50 g (7.40 mmol, 60%) of compound 64.

Step BN: To a cooled (0°C) solution of compound 64 (1.50 g, 7.38 mmol) in THF/methanol mixture (100 mL) was added NaBH 4 (0.491 g, 13.0 mmol). The reaction mixture was stirred for 12 h at room temperature. Then the mixture was cooled to 0°C, treated with 2N hydrochloric acid (40 mL), and concentrated. The residue was extracted with ethyl acetate. The organic extract was washed with water, dried over Na 2 S0 4 , and concentrated under reduced pressure to obtain 1.00 g (4.87 mmol, 65%) of compound 65, pure enough for the next step.

Step BO: To a solution of compound 65, obtained in the previous step, (LOO g, 4.87 mmol) in THF (50 mL), was added IN aqueous LiOH (9 mL). The resulting mixture was stirred for 48 h at room temperature, then concentrated and diluted with IN aqueous NaHS0 4 (9 mL). The mixture was extracted with ethyl acetate. The organic extract was dried over Na 2 S0 4 , andΊ concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 0.250 g (1.30 mmol, 27%) of target compound 4-(hydroxymethyl)- 1 H-indole-2-carboxylic acid.

Rt (Method G) 0.98 mins, m/z 190 [M-H]

Preparation of 4-(2-hydroxypropan-2-yl)-lH-indole-2-carboxylic acid

Steps BP and BQ: To a degassed solution of compound 33 (1.00 g, 3.94 mmol) and tributyl-(l- ethoxyvinyl)stannane (1.58 g, 4.37 mmol) in DMF (25 mL) under argon was added bis(triphenylphosphine)palladium(II) dichloride (0.100 g, 0.142 mmol). The reaction mixture was stirred at room temperature until TLC revealed completion of the reaction (approx. 7 days). The mixture was concentrated under reduced pressure and the residue partitioned between ethyl acetate and water. The organic layer was filtered through a plug of silica gel, dried over MgS04, and concentrated under reduced pressure. The resulting black oil was dissolved in methanol (100 mL), treated with 5N hydrochloric acid (100 mL), and stirred at room temperature overnight. The mixture was concentrated and the residue dissolved in ethyl acetate. The solution was washed with water, dried over Na 2 S0 4 , and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography to give 0.500 g (2.30 mmol, 58%) of compound 67.

Step BR: To a solution of compound 67 (1.00 g, 4.60 mmol) in THF (50 mL), was added 1N aqueous LiOH (7 mL). The resulting mixture was stirred for 48 h at room temperature, then concentrated under reduced pressure and diluted with 1N aqueous NaHS04 (7 mL). The mixture was extracted with ethyl acetate. The organic extract was dried over MgS0 4 , and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 0.900 g (4.43 mol, 96%) of compound 68.

Step BS: To a cooled (0°C) solution of compound 68 (0.900 g, 4.43 mmol) in THF (50 mL) under argon was added a IN solution of MeMgCl (16 mL) in hexane. The resulting mixture was stirred for 48 h at room temperature. The mixture was carefully quenched with 1N NaHS0 4 and extracted with ethyl acetate. The organic extract was dried over Na 2 S0 4 , and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 0.250 g (1.14 mmol, 26%) of target compound 4-(2-hydroxypropan-2-yl)- 1 H-indole-2-carboxylic acid.

Rt (Method G) 0.99 mins, m/z 202 [M-H]

Preparation of 4-(l-hydroxyethyl)-lH-indole-2-carboxyIic acid

Step BS: To a cooled (0°C) solution of compound 66 (1.00 g, 4.60 mmol) in THF/methanol mixture (50 mL) was added NaBH 4 (0.385 g, 10.2 mmol). The reaction mixture was stirred for l2h at room temperature. The mixture was cooled to 0°C, treated with 2N hydrochloric acid (20 mL), and concentrated. The residue was extracted with ethyl acetate. The organic extract was washed with water, dried over Na 2 S0 4 , and evaporated under reduced pressure to obtain 0.800 g (3.65 mmol, 79%) of compound 69, pure enough for the next step.

Step BT: To a solution of compound 69, obtained in the previous step, (0.800 g, 3.65 mmol) in THF (50 mL), was added IN aqueous LiOH (6 mL). The resulting mixture was stirred for 48 h at room temperature, then concentrated and diluted with IN aqueous NaHS0 4 (6 mL). The mixture was extracted with ethyl acetate. The organic extract was dried over MgS0 4 , and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 0.300 g (1.46 mmol, 40%) of target compound 4-(l -hydroxyethyl)- 1 H-indole-2-carboxylic acid.

Rt (Method G) 0.82 mins, m/z 204 [M-H]

Preparation of 4-(propan-2-yl)-lH-indole-2-carboxylic acid

Step BU: To a solution of sodium methoxide (10.0 g, 185 mmol) in methanol (150 mL) at -10°C was added dropwise a solution of compound 70 (15.0 g, 101 mmol) and methyl azidoacetate (12.0 g, 104 mmol) in methanol (100 mL). The reaction mixture was stirred for 3 h maintaining the temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min. The precipitate was then collected by filtration, washed with water and dried to afford 7.00 g (23.3 mmol, 23%) of compound 71 as a white solid.

Step BY: A solution of compound 71, obtained in the previous step, (7.00 g, 23.3 mmol) in xylene (200 mL) was refluxed for lh under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized from hexane-ethyl acetate (60:40) to give 3.50 g (16.1 mmol, 69%) of compound 72. Step BW: To a solution of compound 72 (3.50 g, 16.1 mmol) in methanol (100 mL) was added 2N aqueous NaOH (40 mL). The mixture was stirred for 2 h at 60°C. The mixture was concentrated under reduced pressure, and then residue acidified to pH 5-6 with 10% hydrochloric acid. The precipitate was collected by filtration, washed with water (3 x 50 mL), and dried to obtain 2.70 g (13.3 mmol, 83%) of target compound 4-(propan-2-yl)- lH-indole-2- carboxylic acid.

Rt (Method G) 1.32 mins, m/z 202 [M-H] Preparation of 4-ethenyl-lH-indole-2-carboxylic acid

63

Step BX: To a solution of compound 63 (0.900 g, 4.47 mmol) in THF (50 mL), was added 1N aqueous LiOH (8 mL). The resulting mixture was stirred for 48 h at room temperature, then concentrated under reduced pressure and diluted with 1N aqueous NaHSQ 4 (8 mL). The mixture was extracted with ethyl acetate. The organic extract was dried over MgS0 4 and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 0.500 g (2.67 mmol, 59%) of target compound 4-ethenyl- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.14 mins, m/z 186 [M-H]

Preparation of 4-ethynyl-lH-indole-2-carboxylic acid

33 73

Step BY: To a solution of compound 33 (1.00 g, 3.94 mmol) in THF (50 mL) under argon were added TMS-acetylene (0.68 mL, 4.80 mmol), Cul (0.076 g, 0.399 mmol), triethylamine (2.80 mL, 20.0 mmol), and Pd(dppf)Cl 2 (0.100 g, 0.137 mmol). The mixture was stirred at 60°C until TLC revealed completion of the reaction (approx. 5 days). The mixture was concentrated under reduced pressure, and the residue dissolved in ethyl acetate. The solution was washed with water, dried over Na 2 S0 4 , and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 0.600 g (2.14 mmol, 56%) of compound 73.

Step BZ: To a solution of compound 73 (0.840 g, 3.10 mmol) in THF (50 mL), was added IN aqueous LiOH (7 mL). The resulting mixture was stirred for 48 h at room temperature, then concentrated under reduced pressure and diluted with 1N aqueous NaHS0 4 (7 mL). The mixture was extracted with ethyl acetate. The organic extract was dried over MgS0 4 and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 0.400 g (2.17 mmol, 70%) of target compound 4-ethynyl- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.12 mins, m/z 184 [M-H]

Preparation of 4-(l,l-difluoroethyl)-lH-indoIe-2-carboxylic acid

Step CA: To a mixture of 2-bromoacetophenone (63.0 g, 317 mmol), water (0.5 mL), and dichloromethane (100 mL) was added Morph-DAST (121 mL, 992 mmol). The resulting mixture was stirred for 28 days at room temperature. The reaction mixture was then poured into saturated aqueous NaHC0 3 (1000 mL) and extracted with ethyl acetate (2 x 500 mL). The organic layer was dried over Na 2 S0 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give 16.8 g (76.0 mmol, 12%) of compound 74,

Step CB: To a cooled (-85°C) solution of compound 74 (16.8 g, 76.0 mmol) in THF (300 mL) under Ar was added 2.5M solution of n-BuLi in hexanes (36.5 mL, 91.5 mmol) over 30 min. The resulting mixture was stirred for 1 h at -85 °C. DMF (8.80 mL, 114 mmol) was then added (maintaining temperature below -80°C) and the reaction stirred for a further 45 min. The reaction was quenched with saturated aqueous NH 4 C1 (100 mL) and diluted with water (600 mL). The obtained mixture was extracted with ethyl acetate (2 x 500 mL). The combined organic extracts were dried over Na 2 S04, and concentrated under reduced pressure to obtain 12.5 g (73.6 mmol, 97%) of compound 75 (sufficiently pure for the next step).

Step CC: To a cooled (-30°C) mixture of compound 75 (12.5 g, 73.5 mmol), ethanol (500 mL), and ethyl azidoacetate (28.5 g, 221 mmol) was added a freshly prepared solution of sodium methoxide (prepared by mixing Na (5.00 g, 217 mmol) and methanol (100 mL)) portionwise under Ar (maintaining the temperature below -25°C). The reaction mixture was warmed to 15°C and stirred for 12 h. The obtained mixture was poured into saturated aqueous NH 4 C1 (2500 mL) and stirred for 20 min. The precipitate was collected by filtration, washed with water, and dried to obtain 10.0 g (35.6 mmol, 51%) of compound 76.

Step CD: A solution of compound 76 (10.0 g, 35.6 mmol) in xylene (500 mL) was refluxed until gas evolution ceased (approx. 2 h) and then concentrated under reduced pressure. The orange oil obtained was triturated with hexane/ethyl acetate (5:1), collected by filtration, and dried to obtain 1.53 g (6.04 mmol, 17%) of compound 77.

Step CE: To a solution of compound 77 (1.53 g, 6.04 mmol) in THF/water 9:1 mixture (100 mL) was added LiOH-H 2 0 (0.590 g, 14.1 mmol). The resulting mixture was stirred overnight at r.t. The volatiles were evaporated and the residue mixed with water (50 mL) and IN hydrochloric acid (10 mL). The mixture was extracted with ethyl acetate (2 x 100 mL). The combined organic extracts were dried over Na 2 S0 4 , and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography to give 0.340 g (1.33 mmol, 24%) of 4-( 1 , 1 -difluoroethyl)- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.16 mins, m/z 224 [M-H]

>0

Preparation of 4-(trimethylsilyl)-lH-indole-2-carboxylic acid

78

Step CF: To a cooled (-78°C) solution of 4-bromo- 1 H-indole (5.00 g, 25.5 mmol) in THF (100 mL) under Ar was added a 2.5M solution of n-BuLi in hexanes (23 mL, 57.5 mmol). The resulting mixture was stirred for 30 min. TMSC1 (16 mL, 126 mmol) was added and the reaction mixture warmed to room temperature. After lh the mixture was diluted with MTBE (250 mL), washed with water (2 x 200 mL) and brine (200 mL), then dried over Na 2 S0 4 , and concentrated under reduced pressure. The residue was refluxed in methanol (100 mL) for 1 h. The solvent was then distilled off to obtain 3.60 g (19.0 mmol, 74%) of compound 78.

Step CG: To a cooled (-78°C) solution of compound 78 (1.50 g, 7.92 mmol) in THF (50 mL) under Ar was added a 2.5M solution of n-BuLi in hexanes (3.8 mL, 9.5 mmol). The resulting mixture was stirred for 20 min. C0 2 (2 L) was then bubbled through the mixture for 10 min, and the reaction mixture warmed to room temperature. The volatiles were evaporated and the residue dissolved in THF (50 mL). The solution was cooled to -78°C, and a 1.7M solution of t-BuLi (5.6 mL, 9.50 mmol) was added. The mixture was warmed to -30°C, then again cooled to -78°C. C0 2 (2 L) was bubbled through the solution for 10 min. The obtained solution was allowed to slowly warm to r.t. then concentrated under reduced pressure. The residue was dissolved in water (50 mL), washed with MTBE (2 x 50 mL), then acidified to pH 4, and extracted with ethyl acetate (2x 50 mL). The organic extract was washed with water (2 x 50 mL), and brine (50 mL), dried over Na 2 S0 4 , and evaporated under reduced pressure. The crude product was washed with hexane and dried to obtain 1.24 g (5.31 mmol, 67%) of target compound 4-(trimethylsilyl)- 1 H- indole-2-carboxylic acid.

Rt (Method G) 1.47 mins, m/z 232 [M-H]

Preparation of 6-chloro-5-fluoro-lH-indole-2-carboxylic acid

Step CH: To a solution of (3-chloro-4-fluorophenyl)hydrazine (80.0 g, 498 mmol) in ethanol (200 mL) was added ethyl pyruvate (58.0 g, 499 mmol). The mixture was refluxed for 1 h, then concentrated under reduced pressure, and diluted with water (300 mL). The solid was collected by filtration then dried to obtain 122 g (472 mmol, 95%) of compound 79.

Step Cl: A suspension of compound 79 (122 g, 472 mmol) and pTSA (81.5 g, 473 mmol) in toluene (500 mL) was refluxed for 48 h, then cooled to room temperature. The precipitate was collected by filtration and purified by fractional crystallization from toluene to obtain 4.00 g (16.6 mmol, 4%) of compound 80.

Step CJ: To a refluxing solution of compound 80 (4.00 g, 16.6 mmol) in ethanol (30 mL) was added NaOH (0.660 g, 16.5 mmol). The mixture was refluxed for 1 h, then concentrated under reduced pressure. The residue was triturated with warm water (80°C, 50 mL) and the solution acidified (pH 2) with concentrated hydrochloric acid. The precipitate was collected by filtration, washed with water (2 x 10 mL), and dried to obtain 3.18 g (14.9 mmol, 90%) of target compound 6-chloro-5 -fluoro- 1 H-indole-2-carboxylic acid.

Rt (Method G) 1.23 mins, m/z 212 [M-H]

Preparation of 4-(difluoromethyl)-6-fluoro-lH-indole-2-carboxylic acid

Step CK: To a solution of sodium methoxlde (50.0 g, 926 mmol) in methanol (300 mL) at -10°C was added dropwise a solution of 2-bromo-4-fluorobenzaldehyde (222 mmol) and methyl azidoacetate (59.0 g, 457 mmol) in methanol (100 mL). The reaction mixture was stirred for 3h, maintaining the temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min and the solid collected by filtration. The solid was washed with water to afford compound 81 as a white solid (62% yield).

Step CL: A solution of compound 81 (133 mmol) in xylene (250 mL) was refluxed for lh under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized form hexane-ethyl acetate mixture (60:40) to give compound 82 (58% yield).

Step CM: To a heated (90 °C) solution of compound 82 (14.7 mmol) in anhydrous DMF (10 mL) tri-n-butyl(vinyl)tin (3.60 g, 11.4 mmol) and Pd(PPh3)2Cl2 (0.301 g, 0.757 mmol) were added under nitrogen and the resulting mixture was stirred at 90°C for 1 h. The mixture was cooled to room temperature and purified by silica gel column chromatography (60-80% ethyl acetate in hexane). The combined product fractions were concentrated, washed with water (3 x 100 mL), dried over Na 2 S0 4 , and concentrated under reduced pressure to afford compound 83 as a yellow solid (60% yield).

Step CN: To a mixture of compound 83 (12.4 mmol), acetone (200 mL), and water (40 mL) Os0 4 (0.100 g, 0.393 mmol) and NaI0 4 (13.4 g, 62.6 mmol) were added and the reaction was stirred for 10 h at room temperature. Acetone was distilled off and the aqueous solution was extracted with dichloromethane. The combined organic layer was washed with saturated NaHC0 3 solution (2 x 50 mL) and brine (2 x 50 tnL), dried over Na 2 S0 4 , and concentrated under reduced pressure to afford compound 84 (33% yield).

Step CO: To a solution of compound 84 (11.0 mmol) in dichloromethane (50 mL) was added Morph-DAST (4.10 mL, 33.6 mmol). The resulting mixture was stirred until NMR of an aliquot revealed completion of the reaction (2-5 days). The reaction mixture was added dropwise to a cold saturated NaHC0 3 solution (1000 mL). The mixture obtained was extracted with ethyl acetate. The organic layer was dried over MgS0 4 and concentrated. The residue was purified by column chromatography to give compound 85 as yellow solid (48% yield).

Step CP: To a solution of compound 85 (4.50 mmol) in THF (50 mL), was added 1N aqueous LiOH (8 mL). The resulting mixture was stirred for 48 h at room temperature then concentrated under reduced pressure and diluted with 1N aqueous NaHS0 4 (8 mL). The obtained mixture was extracted with ethyl acetate. The organic extract was dried over MgS0 4 and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain 4-(difluoromethyl)-6- fluoro- 1 H-indole-2-carboxylic acid (87%).

Rt (Method G) 1.22 mins, m/å 228 [M-H]

Preparation of 4-(difIuoromethyl)-7-fluoro-lH-indole-2-carboxylic acid

Prepared as described for 4-(difluoromethyl)-6-fluoro- 1 H-indole-2-carboxylic acid, starting from 2-bromo-5-fluorobeazaldehyde (2.5% overall yield).

Rt (Method G) 1.13 mins, m/z 228 [M-H]

Preparation of 4-(difluoromethyl)-lH-indole-2-carboxylic acid

Prepared as described for 4-(difluoromethyl)-6-iluoro- 1 H-indole-2-carboxylic acid, starting from 4-bromo- 1 H-indole-2-carboxylic acid (11% overall yield).

Rt (Method G) 1.17 mins, m/z 210 [M-H] ~

Preparation of 4-(l,l-difluoroethyl)-6-fluoro-lH-mdole-2-carboxylic acid

Step CQ: To a solution of 2-bromo-5-fluorobenzonitrile (10.0 g, 48.5 mmol) in anhydrous tetrahydrofuran (100 mL) under nitrogen was added methylmagnesium bromide (3.2M in ether, 19 mL, 60.0 mmol). The resulting mixture was heated to reflux for 4 h. The reaction mixture was then cooled, poured into 2N hydrochloric acid (100 mL), and diluted with methanol (100 mL). The organic solvents were removed and the crude product precipitated out. The reaction mixture was extracted with ethyl acetate, dried over MgS0 4 , and concentrated. The residue was purified by column chromatography (heptane/dichloromethane) to give 4.88 g (21.9 mmol, 45%) of compound 86 as a pink oil. Step CM: To a solution of compound 86 (110 mmol) in dichloromethane (50mL) at room temperature was added Morph-DAST (41 mL, 336 mmol) and a few drops of water. The resulting mixture was stirred for 48 days at room temperature; every 7 days an additional portion of Morph-DAST (41 mL, 336 mmol) was added. After the reaction was complete, the mixture was carefully added dropwise to cold saturated aqueous NaHC0 3 . The product was extracted with ethyl acetate and the organic extract dried over MgS0 4 and concentrated. The residue was purified by column chromatography to give 87 as a colorless liquid (37% yield).

Step CS: To a cooled (-80 °C) solution of compound 87 (21.0 mmol) in THF (150 mL) was added slowly a 2.5M solution of n-BuLi in hexanes (10.0 mL, 25.0 mmol of n-BuLi). The mixture was stirred for Ih, then DMF (2.62 mL, 33.8 mmol) was added and the mixture stirred for a further lh. The reaction was quenched with saturated aqueous NH 4 CI (250 mL) and extracted with Et 2 0 (3 x 150 mL). The organic layer was dried over Na 2 S0 4 and concentrated under reduced pressure. The residue was purified by silica gel chromatography (ethyl acetate/hexane 1:9) to give compound 88 (52% yield).

Step t’T: I o a solution of sodium methoxide (50.0 g, 926 mmol) in methanol (300 mL) at -10 °C was added dropwise a solution of compound 88 (222 mmol) and methyl azidoacetate (59.0 g, 457 mmol) in methanol (100 mL). The reaction mixture was stirred for 3h, maintaining the temperature below 5°C, then quenched with ice water. The resulting mixture was stirred for 10 min. The solid obtained was collected by filtration, and washed with water to afford compound 89 as a white solid (66% yield).

Step CU: A solution of compound 89 (120 mmol) in xylene (250 mL) was refluxed for 1 h under an argon atmosphere and then concentrated under reduced pressure. The residue was recrystallized from hexane-ethyl acetate to give compound 90 (70% yield).

Step CV: To a solution of compound 90 (4.40 mmol) in THF (50 mL) was added IN aqueous LiOH (8 mL). The resulting mixture was stirred for 48 h at room temperature, then concentrated under reduced pressure and diluted with IN aqueous NaHS0 4 (8 mL). The residue obtained was extracted with ethyl acetate. The organic extract was dried over MgS0 4 and concentrated under reduced pressure. The residue was recrystallized from MTBE to obtain target compound 4-(l,l- difluoroethyl)-6-fluoro- 1 H-indole-2-carboxylic acid (95% yield). Rt (Method G) 1.26 mins, m/z 242 [M-H]

Preparation of 4-(l,l-difluoroethyl)-7-fIuoro-lH-indole-2-carboxylic acid

Prepared as described for 4-( 1 , 1 -difluoroethyl)-6-fluoro- 1 H-indole-2-carboxylic acid, starting from 2-bromo-4-fluoroacetophenone (3.6% overall yield).

Rt (Method G) 1.23 mins, m/z 242 [M-HJ

Preparation of tert-butyl 2-amino-6-methyl-4H,5H,6H,7H-pyrazolo[l,5-a]pyrazine-5- carboxylate

Step A: To a heated (50°C) mixture of 5-[(tert-butoxy)carbonyl]-6-methyl-4H,5H,6H,7H- pyrazolo[ 1 ,5-a]pyrazine-2-carboxylic acid (3.64 g, 12.9 mmol), DIPEA (2.01 g, 15.6 mmol), and benzyl alcohol (4.20 g, 38.8 mmol) in dioxane (30 mL) was added dropwise DPPA (3.56 g, 12.9 mmol). The reaction mixture was then stirred at 90°C for 3 h. Then the solution was cooled to r.t. and concentrated in vacuo. The residue was partitioned between ethyl acetate and water. The organic layer was washed with water and brine, dried over Na 2 S0 4 , and evaporated in vacuo to provide the crude material, which was triturated with MTBE to afford 2.60 g (6.73 mmol, 52%) of tert-butyl 2-{[(benzyloxy)carbonyl]amino}-6-methyl-4H,5H,6H,7H-pyrazolo [l,5- a]pyrazine-5-carboxylate.

Step B: To a solution of tert-butyl 2- {[(benzyloxy)carbonyl]amino} -6-methyl-4H,5H,6H,7H- pyrazolo[ 1 ,5-a]pyrazine-5-carboxylate (2.60 g, 6.73 mmol) in methanol (30 mL) was added Pd/C (358 mg, 10% wt). The suspension was stirred at 45 °C under an atmosphere of hydrogen atmosphere. The catalyst was removed by filtration, and the solution was evaporated to dryness under reduced pressure to obtain 1.68 g (6.66 mmol, 99%) of target compound tert-butyl 2- amino-6-methyl-4H,5H,6H,7H-pyrazolo[ 1 ,5-a]pyrazine-5-carboxylate.

Rt (Method G) 1.07 mins, m/z 253 [M+H] +

Example 1

5-(lH-lndole-2-carbonyl)-N-(oxoian-3-yl)-4H,5H,6H,7H-pyrazoi o[l,5-a]pyrazin-2-amine

Rt (Method A) 2.75 mins, m/z 352 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 7.63 (d, J = 8.1 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.24 - 7.16 (m, 1H), 7.10 - 7.03 (m, 1H), 6.94 (d, J = 2.1 Hz, 1H), 5.42 (d, J = 6.4 Hz, 1H), 5.40 - 5.36 (m, 1H), 4.99 - 4.79 (m, 2H), 4.20 - 4.13 (m, 2H), 4.04 - 3.97 (m, 2H), 3.97 - 3.89 (m, 1H), 3.83 - 3.73 (m, 2H), 3.71 - 3.62 (m, 1H), 3.48 (dd, J = 8.7, 4.0 Hz, 1H), 2.14 - 2.00 (m, 1H), 1.81 - 1.67 (m, 1H).

Example 2

(lr,4r)-4-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l, 5-a]pyrazin-2- yl] amino } cyclohexan- l-ol

Rt (Method A) 2.74 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.77 - 11.58 (m, 1H), 7.63 (d, I = 7.9 Hz, 1H), 7.43 (d, J = 8.3 Hz, 1H), 7.24 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.96 - 6.91 (m, 1H), 5.34 (s, 1H), 4.99 - 4.76 (m, 3H), 4.48 (d, I = 4.3 Hz, 1H), 4.19 - 4.12 (m, 2H), 4.01 - 3.95 (m, 2H), 3.44 - 3.37 (m, 1H), 3.13 - 3.00 (m, 1H), 1.98 - 1.87 (m, 2H), 1.84 - 1.72 (m, 2H), 1.25 - 1.04 (m, 4H).

Example 3

4-{[5-(4-chloro-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l ,5-a]pyrazin-2- yl] amino } cyclohexan- 1 -ol

Step 1

To 4-hydroxycyclohexan- 1 -one (71.9 mg, 0.629 mmol) was added tert-butyl 2 -amino-6, 7- dihydropyrazolo[ 1 ,5-a]pyrazine-5(4H)-carboxylate (100 mg, 0.420 mmol) and dry THF (1 mL). Titanium (IV) ethoxide (0.262 mL, 0.839 mmol) was then added and the mixture heated at 100 °C for 2h. The mixture was cooled, and sodium cyanoborohydride (52.7 mg, 0.839 mmol) was added. The mixture was then heated at 100 °C for a further hour. The reaction mixture was cooled and poured into 2M NH 3 (3 mL). The solids were removed by filtration and washed with EtOAc (6 mL) and water (3 mL). The layers were separated, and the aqueous fraction was extracted with EtOAc (3 mL). The combined organic extracts were washed with brine (4 mL), dried with Na 2 S0 4 and concentrated, then used in the next step without further purification.

Step 2

To a cooled (0 °C) solution of the product of Step 1 in THF (3 mL) was added 1M HC1 (3 mL). After 15 min at room temperature, the reaction mixture was warmed to 60 °C and stirred overnight. The mixture was cooled, and then concentrated to give 4-((4, 5,6,7- tetrahydropyrazolo[ 1 ,5-a]pyrazin-2-yl)amino)cyclohexan- 1 -ol dihydrochloride which was used in the next step without further purification.

Step 3

(4-((4,5 ,6,7 -tetrahydropyrazolo [ 1 ,5-a]pyrazin-2-yl)amino)cyclohexan- 1 -ol dihydrochloride

(50 mg, 0.162 mmol) and DABCO (181 mg, 1.617 mmol)) were dissolved in dry N,N- dimethylformamide (3 mL). To a solution of 4-chloro- 1 H-indole-2-carboxylic acid (31.6 mg, 0.162 mmol) in dry N,N -dimethylformamide (1 mL) was added HATU (73.8 mg, 0.194 mmol). The mixtures were stirred for 10 minutes, then combined and stirred for lh, and then concentrated. The residue was suspended in DMSO. The suspension was filtered and the filtrate was washed with DMSO to give ~2 mL solution. This residue was purified by reverse phase column chromatography, to give 4-{[5-(4-chloro-lH-indole-2-carbonyl)-4H,5H,6H,7H- pyrazolo [1,5 -a]pyrazin-2-yl] amino } cyclohexan- 1 -ol as a white solid (31.4 mg, 47% yield).

Rt (Method A) 2,93 / 2,99 mins, m/z 414,1 / 416,1 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.06 (s, 2H), 7.42 (d, J = 8.1 Hz, 2H), 7.21 (t, J = 7.8 Hz, 2H), 7.15 (d, J = 7.3 Hz, 2H), 6.91 (s, 2H), 5.38 - 5.33 (m, 2H), 5.05 - 4.78 (m, 6H), 4.51 - 4.45 (m, 1H), 4.35 - 4.28 (m, 1H), 4.19 - 4.12 (m, 4H), 4.01 - 3.94 (m, 4H), 3.69 - 3.58 (m, 1H), 3.43 - 3.34 (m, 1H), 3.27 - 3.16 (m, 1H), 3.14 - 2.97 (m, 1H), 1.97 - 1.86 (m, 2H), 1.84 - 1.74 (m, 2H), 1.63 - 1.53 (m, 6H), 1.50 - 1.37 (m, 2H), 1.14 (q, J = 24.0, 12.2 Hz, 4H).

Example 4 N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2-yl]azetidine-3- carboxamide hydrochloride

Rt (Method B) 2,16 mins, m/z 365 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.72 - 11.67 (m, 1H), 10.67 (s, 1H), 9.05 (s, 1H), 8.81 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.45 (d, I = 8.3 Hz, 1H), 7.22 (t, I - 7.6 Hz, 1H), 7.07 (t, J = 7.5

Hz, 1H), 7.01 - 6.96 (m, 1H), 6.48 (s, 1H), 5.12 - 4.91 (m, 2H), 4.27 - 4.12 (m, 4H), 4.10 - 3.99 (m, 4H), 3.80 - 3.69 (m, 1H).

Example 5

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.74 mins, m/z 386 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 9.90 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.21 (t, J = 7.6 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 7.00 - 6.95 (m, 1H), 5.97 (s, 1H), 5.15 - 4.76 (m, 2H), 4.29 - 4.08 (m, 4H), 2.71 - 2.62 (m, 1H), 0.99 - 0.91 (m, 4H).

Example 6

tert-butyl 4-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyra zin-2- yl] carbamoyl } piperidine- 1 -carboxylate

Rt (Method A) 3.4 mins, m/z 491 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.38 (s, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.21 (t, J = 7.7 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.98 (s, 1H), 6.41 (s, 1H), 5.19 - 4.85 (m, 2H), 4.30 - 4.10 (m, 4H), 4.04 - 3.87 (m, 2H), 2.88 - 2.61 (m, 2H), 1.75 - 1.66 (m, 2H), 1.50 - 1.36 (m, 11H).

Example 7

tert-butyl 3-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyra zin-2- yl] carbamoyl } azetidine- 1 -carboxylate

Rt (Method A) 3.31 mins, m/z 463 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.54 (s, 1H), 7.64 (d, J - 7.8 Hz, 1H), 7.44 (d,

J - 8.3 Hz, 1H), 7.21 (t, J = 7.5 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 7.01 - 6.95 (m, 1H), 6.47 (s, 1H), 5.23 - 4.78 (m, 2H), 4.30 - 4.07 (m, 4H), 4.04 - 3.76 (m, 4H), 3.54 - 3.40 (m, 1H), 1.38 (s, 9H).

Example 8

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2-yl]methanesulfonamide

Rt (Method A) 2.44 mins, m/z 360 [M+H]+ 1H NMR (400 MHz, DMSO-d6) d 11.74 - 11.64 (m, 1H), 9.90 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, I = 8.2 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.98 (d, I = 2.0 Hz, 1H), 5.94 (s, 1H), 5.16 - 4.75 (m, 2H), 4.29 - 4.10 (m, 4H), 3.02 (s, 3H).

Example 9

5-(4-chloro- 1 H-indole-2-carbonyl)-6-methyl-4H,5H,6H,7H-pyrazolo[ 1 ,5-a]pyrazin-2-amine

Rt (Method A) 2.99 mins, m/z 330 / 332 [M+H]+

lH NMR (400 MHz, DMSO-d6) d 12.04 (s, 1H), 7.42 (d, J = 8.1 Hz, 1H), 7.26 - 7.10 (m, 2H), 6.91 (s, 1H), 5.37 (s, 1H), 5.25 - 5.07 (m, 2H), 4.75 - 4.43 (m, 3H), 4.12 - 3.96 (m, 1H), 3.92 - 3. s 6 (m, 111) , 1.26 (d, J . S 11 iz, ^311^ .

Example 10

N-[5-(lH-indole-2-carbonyl)-6-methyl-4H,5H,6H,7H-pyrazoio[l, 5-a]pyrazin-2-yi]oxoiane-3- carboxamide

Rt (Method A) 2.92 mins, m/z 394 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 10.52 (s, 1H), 7.65 (d, J = 8.0 Hz, 1H), 7.45 (d, J = 8.7 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.11 - 7.04 (m, 1H), 6.98 (s, 1H), 6.47 (s, 1H), 5.32 (d, J = 17.4 Hz, 1H), 5.29 - 5.20 (m, 1H), 4.79 - 4.52 (m, 1H), 4.32 - 4.16 (m, 1H), 3.99 (d, J = 12.6 Hz, 1H), 3.90 (td, J = 8.2, 4.1 Hz, 1H), 3.80 - 3.72 (m, 1H), 3.72 - 3.63 (m, 2H), 3.15 (p, J = 7.7 Hz, 1H), 2.09 - 1.98 (m, 2H), 1.25 (d, J = 6.8 Hz, 3H).

Example 11

N-[5-(4-chloro-lH-indole-2-carbonyi)-6-methyl-4H,5H,6H,7H-py razolo[l,5-a]pyrazin-2- yl]oxolane-3-carboxamide

Rt (Method A) 3.13 mins, m/z 428 / 430 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.06 (s, 1H), 10.52 (s, 1H), 7.43 (d, J = 8.1 Hz, 1H), 7.21 (t,

J = 7.8 Hz, 1H), 7.15 (d, J = 7.4 Hz, 1H), 6.95 (s, 1H), 6.48 (s, 1H), 5.31 (d, J = 17.3 Hz, 1H), 5.27 - 5.16 (m, 1H), 4.92 - 4.48 (m, 1H), 4.36 - 4.14 (m, 1H), 4.00 (d, J = 12.6 Hz, 1H), 3.90 (td, J = 8.1, 4.0 Hz, 1H), 3.80 - 3.72 (m, 1H), 3.72 - 3.62 (m, 2H), 3.15 (p, J = 7.7 Hz, 1H), 2.08 - 1.98 (m, 2H), 1.29 - 1.21 (m, 3H).

Example 12

5-(lH-indole-2-carbonyl)-6-methyl-N-[(oxolan-3-yl)methyl]-4H ,5H,6H,7H-pyrazolo[l,5- a]pyrazin-2-amine

Rt (Method A) 3.00 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.65 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.47 - 7.41 (m, 1H), 7.24 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.94 (s, 1H), 5.40 (s, 1H), 5.33 (t, J = 6.1 Hz, 1H), 5.26

- 5.13 (m, 2H), 4.74 - 4.31 (m, 1H), 4.13 - 4.01 (m, 1H), 3.86 (d, J = 12.4 Hz, 1H), 3.75 - 3.66 (m, 2H), 3.60 (q, J = 7.7 Hz, 1H), 3.46 - 3.39 (m, 1H), 3.04 - 2.89 (m, 2H), 2.48 - 2.38 (m, 1H), 1.98 - 1.87 (m, 1H), 1.60 - 1.49 (m, 1H), 1.27 (d, J = 6.9 Hz, 3H).

Example 13

5-(4-chloro-lH-indole-2-carbonyl)-6-methyl-N-[(oxolan-3-yl)m ethyl]-4H,5H,6H,7H- pyrazolo[ 1 ,5-a]pyrazin-2-amine

Rt (Method A) 3.22 mins, m/z 414 / 416 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.04 (s, 1H), 7.42 (d, J = 8.1 Hz, 1H), 7.21 (t, J = 7.8 Hz, 1H), 7.15 (d, J = 7.3 Hz, 1H), 6.91 (s, 1H), 5.41 (s, 1H), 5.34 (t, J = 6.1 Hz, 1H), 5.25 - 5.10 (m, 2H), 4.16 - 3.99 (m, 1H), 3.87 (d, J = 12.3 Hz, 1H), 3.75 - 3.66 (m, 2H), 3.60 (q, J = 7.7 Hz, 1H), 3.46 - 3.39 (m, 1H), 3.04 - 2.89 (m, 2H), 2.48 - 2.40 (m, 1H), 1.99 - 1.87 (m, 1H), 1.60 - 1.50 (m, 1H), 1.27 (d, J = 6.8 Hz, 3H).

Example 14

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2-yl]acetamide

Rt (Method A) 2.72 mins, m/z 324 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.36 (s, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.26 - 7.17 (m, 1H), 7.11 - 7.03 (m, 1H), 6.98 (s, 1H), 6.41 (s, 1H), 5.31 - 4.64 (m, 2H), 4.33 - 4.03 (m, 4H), 1.98 (s, 3H).

Example 15

5-(lH-indoie-2-carbonyl)-N-[(oxan-4-yl)methyl]-4H,5H,6H,7H-p yrazolo[l,5-a]pyrazin-2-amine

Rt (Method A) 2.98 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.78 - 11.57 (m, 1H), 7.63 (d, I = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.30 - 7.16 (m, 1H), 7.11 - 7.02 (m, 1H), 6.98 - 6.91 (m, 1H), 5.36 (s, 1H), 5.24 (t, J = 6 2 Hz, 1H), 5.15 - 4.59 (m, 2H), 4.26 - 4.09 (m, 2H), 4.06 - 3.91 (m, 2H), 3.88 - 3.76 (m, 2H), 3.30 - 3.18 (m, 2H), 2.88 (t, I = 6.4 Hz, 2H), 1.81 - 1.67 (m, 1H), 1.67 - 1.55 (m, 2H), 1.14 (qd, J = 12.0, 4.4 Hz, 2H).

Example 16

5-(lH-indole-2-carbonyl)-6-methyl-4H,5H,6H,7H-pyrazolo[l,5-a ]pyrazin-2-amine

Rt (Method A) 2.76 mins, m/z 296 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.65 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (dd, J = 8.3, 1.0 Hz, 1H), 7.30 - 7.15 (m, 1H), 7.15 - 7.00 (m, 1H), 7.00 - 6.89 (m, 1H), 5.36 (s, 1H), 5.28 - 5.11 (m, 2H), 4.61 (s, 3H), 4.05 (dd, J = 12.5, 4.4 Hz, 1H), 3.82 (dd, J = 12.5, 1.4 Hz, 1H), 1.26 (d, J = 6.9 Hz, 3H

Example 17

5-(4-ethyl-lH-indole-2-carbony!)-N-[(oxolan-3-yl)methyl]-4H, 5H,6H,7H-pyrazolo[l,5- a]pyrazin-2-amine

Rt (Method A) 3.16 mins, m/z 394 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.62 (s, 1H), 7.26 (d, J = 8.3 Hz, 1H), 7.17 - 7.09 (m, 1H), 6.98 (s, 1H), 6.88 (d, J = 7.4 Hz, 1H), 5.42 - 5.28 (m, 2H), 5.10 - 4.70 (m, 2H), 4.25 - 4.10 (m,

2H), 4.07 - 3.93 (m, 2H), 3.75 - 3.65 (m, 2H), 3.64 - 3.55 (m, 1H), 3.42 (dd, J = 8.4, 5.5 Hz, 1H), 3.03 - 2.85 (m, 4H), 2.48 - 2.39 (m, 1H), 1.98 - 1.86 (m, 1H), 1.60 - 1.49 (m, 1H), 1.28 (t, J = 7.5 Hz, 3H).

Example 18

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2-yl]oxane-4-carboxamide

Rt (Method A) 2.84 mins, m/z 392 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.35 (s, 1H), 7.67 - 7.61 (m, 1H), 7.47 - 7.41 (m, 1H), 7.25 - 7.17 (m, 1H), 7.1 1 - 7.03 (m, 1H), 7.01 - 6.95 (m, 1H), 6.43 (s, 1H), 5.23 - 4.78 (m, 2H), 4.27 - 4.09 (m, 4H), 3.92 - 3.83 (m, 2H), 3.32 - 3.24 (m, 2H), 2.65 - 2.54 (m, 1H), 1.68 - 1.54 (m, 4H).

Example 19

N-[5-(4-chloro-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l, 5-a]pyrazin-2-yl]oxolane-3- carboxamide

Step 1

To a cooled (0° C) solution of tert-butyl 2-amino-6,7-dihydropyrazolo[ 1 ,5-a]pyrazine-5(4H)- carboxylate (500 mg, 2.098 mmol) in dichloromethane (14 mL), were added DMAP (25.6 mg, 0.210 mmol) and TEA (0.379 mL, 2.73 mmol). A solution of oxolane-3-carbonyl chloride (296 mg, 2.203 mmol) in dichloromethane (2 mL) was then added. After lh, the reaction was quenched by addition of sat. aq. NaHC0 3 . The layers were seperated and the aqueous layer was extracted with DCM. The combined organic extracts were dried (Na 2 S0 4 ), concentrated and purified by flash column chromatography (24 g silica, 50%-100% EtOAc in heptane) to give tert-butyl 2-(oxolane-3-amido)-4H,5H,6H,7H-pyrazolo[l,5-a]pyrazine-5-ca rboxylate as a white solid (672 mg, 87% yield).

Step 2

To tert-butyl 2-(tetrahydrofuran-3-carboxamido)-6,7-dihydropyrazolo[l,5-a] pyrazine-5(4H)- carboxylate (180 mg, 0.535 mmol) was added 4M HC1 in dioxane (3 mL, 12 mmol). The mixture was stirred at r.t. for lh. The reaction mixture was concentrated under reduced pressure and stripped twice with DCM, then used in the next step without further purification.

Step 3

To a solution of 4-chloro-l H-indole-2-carboxylic acid (17.44 mg, 0.089 mmol) in N,N-dry dimethylformamide (0.5 mL), was added HATU (44.1 mg, 0.116 mmol). In a separate vial, N- (4,5,6,7-tetrahydropyrazolo[l,5-a]pyrazin-2-yl)tetrahydrofur an-3-carboxamide hydrochloride (24.32 mg, 0.089 mmol) was suspended in dry N,N-dimethylformamide (0.5 mL) to which was added TEA (0.062 mL, 0.446 mmol). After 5 min the reaction mixtures were combined and stirred overnight. A few drops of water were then added, and the mixture filtered and then purified by chromatography to give N-[5-(4-chloro-lH-indole-2-carbonyl)-4H,5H,6H,7H- pyrazolo[ 1 ,5-a]pyrazin-2-yl]oxolane-3-carboxamide as a white powder (25 mg, 67% yield).

Rt (Method A) 3.01 mins, m/z 414 / 416 [M+H]+

1H NMR (400 MHz, DMSO-d6) 8 12.07 (s, 1H), 10.51 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.21 (t, J = 7.8 Hz, 1H), 7.16 (d, J = 7.3 Hz, 1H), 6.95 (s, 1H), 6.44 (s, 1H), 5.36 - 4.70 (m, 2H), 4.34 - 4.07 (m, 4H), 3.89 (t, J = 8.2 Hz, 1H), 3.80 - 3.71 (m, 1H), 3.71 - 3.62 (m, 2H), 3.14 (p, J - 7.7 Hz, 1H), 2.07 - 1.98 (m, 2H)

Example 20

N-[5-(4-chloro-5-fluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H-py razolo[l,5-a]pyrazin-2- yl]oxolane-3-carboxamide

To a solution of 4-chloro-5-fluoro- 1 H-indole-2-carboxylic acid (19.05 mg, 0.089 mmol) in dry N,N -dimethylformamide (0.5 mL), was added HATU (44.1 mg, 0.116 mmol). In a separate vial, N-(4,5,6,7-tetrahydropyrazolo[l,5-a]pyrazin-2-yl)tetrahydrof uran-3-carboxamide hydrochloride (24.32 mg, 0.089 mmol) was suspended in dry N,N-dimethylformamide (0.5 mL) to which was added TEA (0.062 mL, 0.446 mmol). After 5 min the reaction mixtures were combined and stirred overnight. A few drops of water were added, the mixture was filtered and then purified by chromatography to give N-[5-(4-chloro-5-fluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H- pyrazolo [1,5 -a]pyrazin-2-yl] oxolane-3 -carboxamide as a white powder (21 mg, 54% yield).

Rt (Method A) 3.05 mins, /z 432 / 434 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.16 (s, 1H), 10.51 (s, 1H), 7.43 (dd, J = 8.9, 4.0 Hz, 1H), 7.29 - 7.21 (m, 1H), 6.99 (s, 1H), 6.44 (s, 1H), 5.33 - 4.73 (m, 2H), 4.27 - 4.09 (m, 4H), 3.89 (t, J = 8.2 Hz, 1H), 3.80 - 3.71 (m, 1H), 3.71 - 3.62 (m, 2H), 3.14 (p, J = 7.7 Hz, 1H), 2.07 - 1.98 (m, 2H).

Example 21

N-[5-(4-chloro-6-fluoro- 1 H-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[ 1 ,5-a]pyrazin-2- yl]oxolane-3-carboxamide

Rt (Method A) 3.1 mins, m/z 432 / 434 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.15 (s, 1H), 10.51 (s, 1H), 7.27 - 7.10 (m, 2H), 6.98 (s, 1H), 6.44 (s, 1H), 5.34 - 4.67 (m, 2H), 4.37 - 4.01 (m, 4H), 3.89 (t, J = 8.2 Hz, 1H), 3.75 (q, J = 7.3 Hz, 1H), 3.71 - 3.62 (m, 2H), 3.14 (p, J = 7.7 Hz, 1H), 2.06 - 1.97 (m, 2H).

Example 22

N-[5-(4,6-difluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazol o[l,5-a]pyrazin-2-yl]oxolane-3- carboxamide

Rt (Method A) 2.9 mins, m/z 416 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.11 (s, 1H), 10.51 (s, 1H), 7.10 - 7.02 (m, 2H), 6.92 (td, J =

10.4, 2.1 Hz, 1H), 6.43 (s, 1H), 5.29 - 4.73 (m, 2H), 4.36 - 4.09 (m, 4H), 3.89 (t, J = 8.2 Hz, 1H), 3.79 - 3.71 (m, 1H), 3.71 - 3.61 (m, 2H), 3.14 (p, J = 7.7 Hz, 1H), 2.07 - 1.97 (m, 2H).

Example 23

N-[5-(4-ethyl-6-fluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyr azolo[l,5-a]pyrazm-2- yl]oxolane-3 -carboxamide

Rt (Method A) 3.15 mins, m/z 426 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.72 (s, 1H), 10.52 (s, 1H), 7.07 (s, 1H), 6.97 (dd, J = 9.8, 2.2 Hz, 1H), 6.78 (dd, J = 10.8, 2.3 Hz, 1H), 6.44 (s, 1H), 5.26 - 4.73 (m, 2H), 4.32 - 4.05 (m, 4H), 3.89 (t, J = 8.2 Hz, 1H), 3.75 (q, J = 7.2 Hz, 1H), 3.71 - 3.62 (m, 2H), 3.14 (p, I = 7.7 Hz, 1H), 2.91 (q, J - 7.5 Hz, 2H), 2.07 - 1.97 (m, 2H), 1.28 (t, J = 7.5 Hz, 3H).

Example 24

N-[5-(4-ethyl-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5 -a]pyrazin-2-yl]oxolane-3- carboxamide

Rt (Method A) 3.08 mins, m/z 408 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.64 (s, 1H), 10.52 (s, 1H), 7.26 (d, J = 8.1 Hz, 1H), 7.16 - 7.10 (m, 1H), 7.03 (s, 1H), 6.88 (d, J = 7.0 Hz, 1H), 6.44 (s, 1H), 5.24 - 4.71 (m, 2H), 4.27 - 4.11 (m, 4H), 3.89 (t, I = 8.2 Hz, 1H), 3.75 (q, J = 7.2 Hz, 1H), 3.71 - 3.62 (m, 2H), 3.14 (p, J = 7.7 Hz, 1H), 2.91 (q, J = 7.6 Hz, 2H), 2.07 - 1.97 (m, 2H), 1.29 (t, J = 7.5 Hz, 3H).

Example 25

3-yl)methyl]-4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin-2-amine

Rt (Method A) 3.13 mins, m/z 418 / 420 [M+H]+

lH NMR (400 MHz, DMSO-d6) 6 12.15 (s, 1H), 7.42 (dd, J = 8.8, 4.0 Hz, !H), 7.24 (dd, J = 10.0, 8.9 Hz, 1H), 6.95 (s, 1H), 5.47 - 5.28 (m, 2H), 4.98 - 4.82 (m, 2H), 4.29 - 4.07 (m, 2H), 4.06 - 3.92 (m, 2H), 3.75 - 3.65 (m, 2H), 3.60 (q, J = 7.7 Hz, 1H), 3.42 (dd, J = 8.4, 5.5 Hz, 1H), 3.03 - 2.89 (m, 2H), 2.48 - 2.40 (m, 1H), 1.98 - 1.86 (m, 1H), 1.60 - 1.48 (m, 1H).

Example 26

5-(4-chloro-lH-indole-2-carbonyl)-N-[(oxolan-3-yl)methyl]-4H ,5H,6H,7H-pyrazolo[l,5- a]pyrazin-2-amine

Rt (Method A) 3.1 mins, m/z 400 / 402 [M+H]+

1H NMR (400 MHz, DMSCM6) d 12.06 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.21 (t, J = 7.8 Hz, 1H), 7.15 (d, I = 7.3 Hz, 1H), 6.91 (s, 1H), 5.42 - 5.30 (m, 2H), 5.14 - 4.60 (m, 2H), 4.29 - 4.08 (m, 2H), 4.06 - 3.90 (m, 2H), 3.76 - 3.65 (m, 2H), 3.60 (q, I = 7.7 Hz, 1H), 3.42 (dd, J = 8.4, 5.5 Hz, 1H), 3.03 - 2.88 (m, 2H), 2.48 - 2.39 (m, 1H), 1.98 - 1.86 (m, 1H), 1.60 - 1.48 (m, 1H).

Example 27 5-(4-chloro-6-fluoro-lH-indole-2-carbonyl)-N-[(oxolan-3-yl)m ethyl]-4H,5H,6H,7H- pyrazolo[l ,5-a]pyrazin-2-amine

Rt (Method A) 3.19 mins, m/z 418 / 420 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.14 (s, 1H), 7.20 - 7.15 (m, 2H), 6.94 (s, 1H), 5.38 (s, 1H),

5.34 (t, J = 6.1 Hz, 1H), 5.04 - 4.76 (m, 2H), 4.21 - 4.10 (m, 2H), 4.03 - 3.93 (m, 2H), 3.75 - 3.65 (m, 2H), 3.60 (q, J = 7.7 Hz, 1H), 3.42 (dd, I = 8.4, 5.5 Hz, 1H), 3.03 - 2.89 (m, 2H), 2.48 - 2.39 (m, 1H), 1.98 - 1.86 (m, 1H), 1.60 - 1.48 (m, 1H). Example 28

5-(4,6-difluoro- 1 H-indole-2-carbonyl)-N-[(oxolan-3-yl)methyl]-4H,5H,6H,7H-pyr azolo[ 1 ,5- a]pyrazin-2-amine

Rt (Method A) 3.06 mins, m/z 402 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.09 (s, 1H), 7.08 - 7.00 (m, 2H), 6.92 (td, J = 10.4, 2.1 Hz, 1H), 5.44 - 5.25 (m, 2H), 5.17 - 4.57 (m, 2H), 4.28 - 4.07 (m, 2H), 4.06 - 3.91 (m, 2H), 3.75 - 3.65 (m, 2H), 3.60 (q, J = 7.7 Hz, 1H), 3.42 (dd, J = 8.4, 5.5 Hz, 1H), 3.03 - 2.88 (m, 2H), 2.48 - 2.39 (m, 1H), 1.98 - 1.86 (m, 1H), 1.60 - 1.48 (m, 1H).

Example 29

5-(4-ethyl-6-fluoro-lH-indole-2-carbonyl)-N-[(oxolan-3-yl)me thyl]-4H,5H,6H,7H-pyrazolo[l,5- a]pyrazin-2-amine

Step 1

To a solution of tert-butyl 2-(tetrahydrofiiran-3-carboxamido)-6,7-dihydropyrazolo[ 1 ,5- a]pyrazine-5(4H)-carboxylate (345 mg, 1.026 mmol) in dry THF (7 mL) was added borane-THF complex (1M solution in THF, 5.13 mL, 5.13 mmol). The mixture was heated to 60 °C and stirred for 48h. The mixture was cooled to 0 °C, and 1M aq. HC1 (3 mL) was added. The mixture was stirred for 15 minutes at room temperature, then re-cooled to 0 °C. Saurated aq NaHCOs was added until pH 8. EtOAc was added, and the layers were seperated. The aqueous fraction was extracted with EtOAc (twice). The combined organic extracts were dried (Na S0 4 ), concentrated under reduced pressure, and purified by flash chromatography (silica, 12g 0%-l0% MeOH:DCM) to give tert-butyl 2-(((tetrahydrofuran-3 -yl)methyl)amino)-6,7- dihydropyrazolo[ 1 ,5-a]pyrazine-5(4H)-carboxylate (279 mg, 0.822 mmol, 80 % yield).

Step 2

To tert-butyl 2-(((tetrahydrofuran-3-yl)methyl)amino)-6,7-dihydropyrazolo[ l,5-a]pyrazine- 5(4H)-carboxylate (279 mg, 0.865 mmol) was added HC1 (4M in dioxane, 3 mL, 12 mmol). The mixture was stirred for 1H, then concentrated under reduced pressure and used directly without further purification.

Step 3

To a solution of 4-ethyl-6-fluoro- 1 H-indole-2-carboxylic acid (25.5 mg, 0.123 mmol) in dry N,N -dimethylformamide (0.5 mL) was added HATU (60.9 mg, 0.160 mmol). In a separate vial, to a suspension of N-((tetrahydrofuran-3-yl)methyl)-4,5,6,7-tetrahydropyrazolo[ l,5-a]pyrazin-2- amine dihydrochloride (36.4 mg, 0.123 mmol) in dry N,N -dimethylformamide (0.5 mL) was added TEA (0.086 mL, 0.617 mmol). After 5 mins the two reaction mixtures were combined and stirred for 48h. A few drops of water were added, the solution was filtered and then purified by chromatography to give 5-(4-ethyl-6-fluoro- 1 H-indole-2-carbonyl)-N-[(oxolan-3-yl)methyl]- 4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin-2-amine as a white solid (18 mg, 35% yield).

5 Rt (Method A) 3.23 mins, m/z 412 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.71 (s, 1H), 7.03 (s, 1H), 6.97 (dd, I = 9.8, 2.3 Hz, 1H), 6.77 (dd, J = 10.8, 2.3 Hz, 1H), 5.44 - 5.29 (m, 2H), 5.09 - 4.70 (m, 2H), 4.28 - 4.07 (m, 2H), 4.04 - 3.91 (m, 2H), 3.75 - 3.65 (m, 2H), 3.60 (q, I = 7.7 Hz, 1H), 3.42 (dd, I = 8.5, 5.5 Hz, 1H), 3.03 - 2.86 (m, 4H), 2.48 - 2.40 (m, 1H), 1.98 - 1.86 (m, 1H), 1.60 - 1.49 (m, 1H), 1.28 (t, J = 7.5 I Hz, 3H).

Example 30

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2-yl]oxolane-3-carboxamide

Rt (Method A) 2.8 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.50 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (dd, J = 8.1, 1.0 Hz, 1H), 7.25 - 7.19 (m, 1H), 7.11 - 7.04 (m, 1H), 6.98 (s, 1H), 6.43 (s, 1H), 5.18 - 4.78 (m, 2H), 4.30 - 4.07 (m, 4H), 3.89 (t, J = 8.2 Hz, 1H), 3.75 (q, J = 7.2 Hz, 1H), 3.71 - 3.62 (m, 2H), 3.20 - 3.09 (m, 1H), 2.09 - 1.97 (m, 2H).

Example 31

5-(lH-indole-2-carbonyl)-N-[(oxolan-3-yl)methyl]-4H,5H,6H,7H -pyrazolo[l,5-a]pyrazin-2- amine

;, - Step 1: To a solution of tert-butyl 2-(tetrahydrofuran-3 -carboxamido)-6, 7 -dihydrothiazolo [5,4- c]pyridine-5(4H)-carboxylate (0.085 g, 0.253 mmol) in DCM (2 mL) was added a solution of LiAlH4 (2.4M solution in THF). The mixture was stirred for 30 minutes, then quenched by the careful addition of water. Product was extracted with DCM, and the combined organic extracts were dried, concentrated, and used in the next step without further purification.

Step 2: To a DCM (1 mL) solution of the product of Step 1 was added TFA. The mixture was stirred for Ih, then concentrated under vacuum. Excess TFA was removed by co-evaporation with additional DCM (twice). The product was used in the next step without further purification.

Step 3: To a solution of indole-2-carboxylic acid (0.0203 g, 0.126 mmol) in dry DMF (1.0 mL) was added HATU (0.0575 g, 0.151 mmol). The reaction mixture was stirred for 5 minutes, then a solution of th product of Step 2 (N-((tetrahydrofuran-3-yl)methyl)-4, 5,6,7- tetrahydropyrazolo[ 1 ,5-a]pyrazin-2-amine bis(2,2 ,2-trifluoroacetate), 56.7 mg, 0.126 mmol) and triethylamine (0.088 ml, 0.630 mmol) in dry DMF (1.0 ml) was added. The mixture was stirred for lh, then few drops of water were added and the resulting solution was purified directly by reverse phase HPLC to give the desired product (0.0210 g, 52% yield).

Rt (Method A) 2.89 mins, m/z 366 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 7.63 (d, I = 8.0 Hz, 1H), 7.46 - 7.40 (m, 1H), 7.24 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.94 (s, 1H), 5.37 (s, 1H), 5.33 (t, J = 6.1 Hz, 1H), 5.00 - 4.77 (m, 2H), 4.24 - 4.09 (m, 2H), 4.06 - 3.93 (m, 2H), 3.75 - 3.65 (m, 2H), 3.60 (q, J = 7.6 Hz, 1H), 3.42 (dd, J = 8.4, 5.5 Hz, 1H), 3.03 - 2.89 (m, 2H), 2.48 - 2.39 (m, 1H), 1.98 - 1.87 (m, 1H), 1.60 - 1.49 (m, 1H).

Example 32

5-(4-chloro-lH-mdole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a] pyrazin-2-amme

To a solution of 4-chloro- 1 H-indole-2-carboxylic acid (0.0246 g, 0.126 mmol) in dry DMF (0.65 mL) was added HATU (0.0575 g, 0.151 mmol). The reaction mixture was stirred for 5 minutes, then a solution of 4,5,6,7-tetrahydropyrazolo[l,5-a]pyrazin-2-amine dihydrochloride (0.0266 g, 0.126 mmol) and triethylamine (0.088 ml, 0.630 mmol) in dry DMF (0.650 ml) was added. The mixture was stirred for lh, then few drops of water were added and the resulting solution was purified directly by reverse phase HPLC to give the desired product (0.0210 g, 52% yield).

Rt (Method A) 2.88 mins, m/z 316 / 318 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.06 (s, 1H), 7.42 (d, J = 8.1 Hz, 1H), 7.21 (t, J = 7.8 Hz, 1H), 7.15 (d, J = 7.3 Hz, 1H), 6.91 (s, 1H), 5.34 (s, 1H), 5.08 - 4.69 (m, 2H), 4.61 (s, 2H), 4.24 - 4.08 (m, 2H), 4.04 - 3.88 (m, 2H).

Example 33

5-(4,6-difluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l ,5-a]pyrazin-2-amlne

To a solution of 4,6-difluoro- 1 H-indole-2-carboxylic acid (0.0248 g, 0.126 mmol) in dry DMF (0.65 mL) was added HATU (0.0575 g, 0.151 mmol). The reaction mixture was stirred for 5 minutes, then a solution of 4,5,6,7-tetrahydropyrazolo[ 1 ,5-a]pyrazin-2-amine dihydrochloride (0.0266 g, 0.126 mmol) and triethylamine (0.088 ml, 0.630 mmol) in dry DMF (0.650 ml) was added. The mixture was stirred for lh, then few drops of water were added and the resulting solution was purified directly by reverse phase HPLC to give the desired product (0.0217 g, 54% yield).

Rt (Method A) 2.84 mins, m/z 318 [M+HJ+

*H NMR (400 MHz, DMSO-d6) 6 12.10 (s, 1 H), 7.07 - 7.00 (m, 2H), 6.92 (td, J = 10.4, 2.1 Hz, 1H), 5.34 (s, 1H), 5.05 - 4.75 (m, 2H), 4.61 (s, 2H), 4.18 - 4.10 (m, 2H), 4.02 - 3.90 (m, 2H).

Example 34

5-(lH-indole-2-cafbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin- 2-amine

To a solution of indole-2-carboxylic acid (0.020 g, 0.126 mmol) in dry DMF (0.65 ml) was added and HATU (0.057 g, 0.151 mmol). The mixture was stirred for 5 minutes, then a solution of 4,5,6,7-tetrahydropyrazolo[ 1 ,5-a]pyrazin-2-amine dihydrochloride (0.026 g, 0.126 mmol) and triethylamine (0.088 ml, 0.63 mmol) in dry DMF (0.650 ml) was added. The mixture was stirred for lh, water was added until the suspension became a solution, and the mixture was purified directly by reverse phase HPLC to yield the desired product (0.025 g, 70% yield).

Rt (Method A) 2.64 mins, m/z 282 [M+H]+

*H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.43 (d, I = 8.2 Hz,

1H), 7.25 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.94 (s, 1H), 5.33 (s, 1H), 5.08 - 4.70 (m, 2H), 4.61 (s, 2H), 4.24 - 4.09 (m, 2H), 4.04 - 3.90 (m, 2H).

Example 35

N-[5-(4,6-difluoro- 1 H-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[ 1 ,5-a]pyrazin-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.78 mins, m/z 422 [M+H]+

1H NMR (400 MHz, DMSO-d6) 6 12.11 (s, 1H), 9.92 (s, 1H), 7.08 - 7.02 (m, 2H), 6.97 - 6.89 (m, 1H), 5.97 (s, 1H), 5.21 - 4.69 (m, 2H), 4.31 - 3.93 (m, 4H), 2.71 - 2.60 (m, 1H), 1.00 - 0.89 (m, 4H).

Example 36

N-[5-(4-chloro-5-fluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H -pyrazolo[l,5-a]pyrazin-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.87 mins, m/z 438 / 440 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.15 (s, 1H), 10.63 - 9.19 (m, 1H), 7.43 (dd, J = 9.1, 3.9 Hz, 1H), 7.26 (t, J = 9.4 Hz, 1H), 6.99 (s, 1H), 5.98 (s, 1H), 5.16 - 4.83 (m, 2H), 4.24 - 4.12 (m, 4H), 2.71 - 2.61 (m, 1H), 0.99 - 0.91 (m, 4H).

Example 37

N-[5-(4-chloro-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo [l,5-a]pyrazin-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.82 mins, m/z 420 / 422 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.07 (s, 1H), 9.92 (s, 1H), 7.42 (d, I = 8.1 Hz, 1H), 7.22 (t, I = 7.8 Hz, 1H), 7.16 (d, 1 = 7.4 Hz, 1H), 6.95 (s, 1H), 5.98 (s, 1H), 5.23 - 4.79 (m, 2H), 4.25 - 4.12 (m, 4H), 2.74 - 2.62 (m, 1H), 0.99 - 0.91 (m, 4H).

Example 38

N-[5-(4-chloro-6-fluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H -pyrazolo[l,5-a]pyrazin-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.93 mins, m/z 438 / 440 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 12.41 - 11.91 (m, 1H), 10.12 - 9.60 (m, 1H), 7.22 - 7.16 (m, 2H), 6.98 (s, 1H), 5.98 (s, 1H), 5.15 - 4.83 (m, 2H), 4.25 - 4.12 (m, 4H), 2.71 - 2.62 (m, 1H), 0.99 - 0.90 (m, 4H).

Example 39

N-[5-(4-ethyl-6-fluoro-lH-indole-2-carbonyl)-4H,5H,6H,7H- pyrazolo[l,5-a]pyrazin-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.99 mins, m/z 432 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.72 (s, 1H), 9.93 (s, 1H), 7.09 - 7.04 (m, 1H), 6.98 (dd, J = 9.6, 1.9 Hz, 1H), 6.78 (dd, I = 10.8, 2.2 Hz, 1H), 5.98 (s, 1H), 5.12 - 4.87 (m, 2H), 4.26 - 4.12 (m, 4H), 2.91 (q, I = 7.6 Hz, 2H), 2.70 - 2.62 (m, 1H), 1.29 (t, I = 7.5 Hz, 3H), 0.99 - 0.91 (m, 4H).

Example 40 N-[5-(4-ethyl-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5 -a]pyrazin-2- yl] cyclopropanesulfonamide

Rt (Method A) 2.91 mins, m/z 414 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.64 (s, 1H), 9.88 (s, 1H), 7.26 (d, J = 8.2 Hz, 1H), 7.13 (t, I = 7.7 Hz, 1H), 7.04 - 7.00 (m, 1H), 6.89 (d, I = 7.0 Hz, 1H), 5.98 (s, 1H), 5.09 - 4.90 (m, 2H), 4.26 - 4.13 (m, 4H), 2.91 (q, J = 7.6 Hz, 2H), 2.72 - 2.62 (m, 1H), 1.29 (t, I = 7.5 Hz, 3H), 0.99 - 0.91 (m, 4H).

Example 41

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]piperidine-4- carboxamide hydrochloride

Rt (Method A) 2.73 mins, m/z 393 [M+H]+

‘H NMR (400 MHz, DMSO-d6) d 11.69 (s, 1H), 10.50 (s, 1H), 8.96 - 8.75 (m, 1H), 8.66 - 8.46 (m, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.10 - 7.04 (m, 1H), 6.98 (d, J = 2.2 Hz, 1H), 6.42 (s, 1H), 5.08 - 4.86 (m, 2H), 4.31 - 4.08 (m, 4H), 3.34 - 3.25 (m, 2H), 2.95 - 2.80 (m, 2H), 2.71 - 2.58 (m, 1H), 2.03 - 1.86 (m, 2H), 1.85 - 1.65 (m, 2H).

Example 42 l-acetyl-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l, 5-a]pyrazin-2-yl]piperidine-4- carboxamide

Rt (Method A) 2.69 mins, m/z 435 [M+H]+

'H NMR (400 MHz, DMSO-d6) d 1 1.67 (s, 1H), 10.38 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.21 (t, I = 7.6 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.98 (s, 1H), 6.42 (s, 1H), 5.11 - 4.86 (m, 2H), 4.40 - 4.32 (m, 1H), 4.26 - 4.07 (m, 4H), 3.87 - 3.79 (m, 1H), 3.07 - 2.97 (m, 1H), 2.63 - 2.53 (m, 2H), 1.99 (s, 3H), 1.81 - 1.68 (m, 2H), 1.63 - 1.49 (m, 1H), 1.46 - 1.33 (m, 1H).

Example 43

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l - phenylmethanesulfonamide

Rt (Method B) 3.21 mins, m/z 436 [M+H]+

1H NMR (400 MHz, DMSO-46) 6 11.70 (s, 1H), 9.97 (s, 1H), 7.65 (d, J = 7.9 Hz, 1H), 7.45 (d, J = 8.3 Hz, 1H), 7.40 - 7.29 (m, 5H), 7.26 - 7.18 (m, 1H), 7.08 (t, I = 7.5 Hz, 1H), 7.02 - 6.97 (m, 1H), 5.90 (s, 1H), 4.98 (m, 2H), 4.46 (s, 2H), 4.30 - 4.15 (m, 4H).

Example 44

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razm-2-yl]-l- methylcyclopropane- 1 -sulfonamide

Step 1

To a stirred solution of tert-butyl 2-atnino-6,7-dihydropyrazolo[ 1 ,5-a]pyrazine-5(4H)- carboxylate (1.009 g, 4.23 mmol) in Dichloromethane (25 mL) was added 4M hydrochloric acid in 1 ,4-dioxane (16 mL, 64.0 mmol). The resulting suspension was stirred at r.t. overnight. The mixture was concentrated to give 4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin-2-amine dihydrochloride a white solid that was used in the next step without further purification.

Step 2

Indole-2-carboxylic acid (459 mg, 2.85 mmol) and HATU (1.084 g, 2.85 mmol) were dissolved in dry N,N-dimethylformamide (20 mL), and the mixture was stirred for 10 minutes. A suspension of 4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin-2-amine dihydrochloride (599 mg) and TEA (1.3 mL, 9.35 mmol) in dry N,N-dimethylformamide (30 mL) was then added and the resulting reaction mixture was stirred under N 2 at r.t. for lh. of stirring. The reaction mixture was concentrated, then partitioned between EtOAc (100 mL) and water (100 mL). The aqueous phase was extracted with EtOAc (70 mL). The combined organic extracts were washed succesively with sat. NaHC0 3 solution (100 mL) and brine (100 mL), dried over sodium sulfate and concentrated. Solid NaCl was added to the combined aqueous fractions was added until complete saturation, after which the aqueous phase was extracted with EtOAc (100 and 80 mL). The combined organic phases were washed with brine (80 mL), dried over sodium sulfate, concentrated, then purified by flash chromatography (80 g silica; 0.1-10% MeOH in DCM) to give 5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin- 2-amine as a light beige foam (615 mg, 81% yield). Step 3

A solution of 5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyrazin- 2-amine in pyridine (0.8 mL, 0.21M solution, 0.168 mmol) was added to 1 -methylcyclopropane- 1 -sulfonyl chloride (55 pL, 0.252 mmol). The resulting solution was stirred at r.t. for 2 days. To the mixture was added KHSO 4 solution (0.5M, 2 mL) and DCM (2 mL). The resulting mixtures were stirred vigurously for 10 minutes before the organic phases were separated over a phase separator and rinsed with DCM. The organic phase was concentrated, and purified by chromatography to give N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2- yl] - 1 -methylcyclopropane- 1 - sulfonamide (5.6 mg, 8% yield).

Rt (Method A) 2.86 mins, m/z 400 [M+H]+ l º NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 9.95 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.25 - 7.17 (m, 1H), 7.07 (t, J = 7.4 Hz, 1H), 7.00 - 6.94 (m, 1H), 5.95 (s, 1H), 4.96 (s, 2H), 4.31 - 4.05 (m, 4H), 1.42 (s, 3H), 1.19 - 1.07 (m, 2H), 0.81 - 0.69 (m, 2H).

Example 45

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]ethane-l-sulfonamide

Rt (Method A) 2.55 mins, m/z 374 [M+H]+

Example 46

N-[5-(lH-mdole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyr azin-2-yl]-3,5-dimethyl-l,2- oxazole-4-sulfonamide

Rt (Method A) 2.5 mins, m/z 441 [M+H]+

1H NMR (400 MHz, DMSO-d6) 6 11.67 (s, 1H), 10.76 (s, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.44 (d, I = 8.3 Hz, 1H), 7.25 - 7.17 (m, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.98 - 6.93 (m, 1H), 5.90 (s, 1H), 4.94 (s, 2H), 4.26 - 4.04 (m, 4H), 2.57 - 2.52 (m, 3H), 2.32 - 2.25 (m, 3H).

Example 47

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razm-2-yl]propane-2-sulfonamide

Rt (Method A) 2.78 mins, m/z 388 [M+H]+ 1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 9.88 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.21 (t, J = 7.6 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 7.00 - 6.95 (m, 1H), 5.94 (s, 1H), 4.96 (s, 2H), 4.31 - 4.06 (m, 4H), 3.39 - 3.34 (m, 1H), 1.25 (d, J = 6.8 Hz, 6H).

Example 48

-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyr azin-2-yl]-l,2-dimethyl-lH- imidazole-4-sulfonamide

Rt (Method A) 2 4 mins, m/z 440 [M+H]+

Example 49

N- [5-( 1 H-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[ 1 ,5-a]pyrazin-2-yl] - 1 -methyl- 1 H-pyrazole- 4-sulfonamide

Rt (Method A) 2.35 mins, m/z 426 [M+H]+

Example 50

N-[5-(lH-mdole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyr azin-2-yl]pyridine-3-sulfonamide

Rt (Method A) 2.35 mins, m/z 423 [M+H]+

Example 51

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2- yl] cyclohexanesulfonamide

Rt (Method B) 3.23 mins, m/z 428 [M+H]+

1H NMR (400 MHz, DMSO-d6) 5 11.67 (s, 1H), 9.92 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, I = 8.2 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.97 (d, J = 2.1 Hz, 1H), 5.87 (s, 1H), 4.94 (m, 2H), 4.44 - 3.90 (m, 4H), 3.11 - 2.77 (m, 1H), 2.12 - 1.96 (m, 2H), 1.84 - 1.64 (m, 2H), 1.64 - 1.52 (m, 1H), 1.48 - 1.29 (m, 2H), 1.29 - 1.02 (m, 3H).

Example 52

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l-

(methoxymethyl)cyclopropane- 1 -sulfonamide

Rt (Method A) 2.84 mins, m/z 430 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 9.95 (s, 1H), 7.64 (d, I = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.10 - 7.04 (m, 1H), 7.00 - 6.95 (m, 1H), 5.94 (s, 1H), 4.96 (s, 2H), 4.29 - 4.05 (m, 4H), 3.66 (s, 2H), 3.20 (s, 3H), 1.25 - 1.13 (m, 2H), 1.00 - 0.88 (m, 2H).

Example 53

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]oxane-4-sulfonamide

Rt (Method B) 2.89 mins, m/z 430 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 10.05 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.11 - 7.03 (m, 1H), 7.00 - 6.94 (m, 1H), 5.93 (s, 1H), 4.96 (m, 2H), 4.28 - 4.06 (m, 4H), 3.96 - 3.86 (m, 2H), 3.42 - 3.33 (m, 1H), 3.30 - 3.22 (m, 2H), 1.93 - 1.81 (m, 2H), 1.72 - 1.56 (m, 2H).

Example 54

5-(lH-indole-2-carbonyl)-N-{[l-(methoxymethyl)cyclopropyl ]methyl}-4H,5H,6H,7H- pyrazolo[ 1 ,5-a]pyrazin-2-amine

Rt (Method A) 3.07 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.66 (s, 1H), 7.63 (d, I = 7.9 Hz, 1H), 7.43 (d, J = 8.3 Hz,

1H), 7.24 - 7.17 (m, lH), 7.10 - 7.03 (m, 1H), 6.96 - 6.91 (m, 1H), 5.37 (s, 1H), 5.05 (t, J = 6.2 Hz, 1H), 4.97 - 4.78 (m, 2H), 4.25 - 4.09 (m, 2H), 4.05 - 3.89 (m, 2H), 3.24 - 3.20 (m, 5H),

3.00 (d, J = 6.2 Hz, 2H), 0.48 - 0.42 (m, 2H), 0.37 - 0.30 (m, 2H).

Example 55

5-(lH-indole-2-carbonyl)-N-[(oxan-3-yl)methyl]-4H,5H,6H,7 H-pyrazolo[l,5-a]pyrazin-2-amine

Rt (Method A) 2.91 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.21 (t, J = 7.5 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.94 (s, 1H), 5.35 (s, 1H), 5.23 (t, J = 5.9 Hz, 1H), 4.99 - 4.75 (m, 2H), 4.21 - 4.10 (m, 2H), 4.04 - 3.93 (m, 2H), 3.86 - 3.77 (m, 1H), 3.75 - 3.66 (m, 1H), 3.29 - 3.23 (m, 1H), 3.10 - 3.00 (m, 1H), 2.89 - 2.81 (m, 2H), 1.83 - 1.71 (m, 2H), 1.62 - 1.50 (m, 1H), 1.49 - 1.36 (m, 1H), 1.26 - 1.12 (m, 1H).

Example 56

N-benzyl-5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l, 5-a]pyrazin-2-amine

Rt (Method B) 3.3 mins, m/z 372 [M+H]+

1H NMR (400 MHz, DMSCM6) d 11.65 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.37 - 7.25 (m, 4H), 7.24 - 7.16 (m, 2H), 7.09 - 7.03 (m, 1H), 6.93 (d, J = 1.5 Hz, 1H), 5.77 (t, J = 6.3 Hz, 1H), 5.38 (s, 1H), 5.06 - 4.69 (m, 2H), 4.24 - 4.11 (m, 4H), 4.02 - 3.94 (m, 2H).

Example 57

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l-(pyridm-4- yl)piperidine-4-carboxamide

Rt (Method B) 2.51 mins, m/z 470 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 10.40 (s, 1H), 8.16 - 8.09 (m, 2H), 7.64 (d, I = 8.0 Hz, 1H), 7.44 (d, I = 8.2 Hz, 1H), 7.26 - 7.17 (m, 1H), 7.10 - 7.04 (m, 1H), 7.01 - 6.95 (m, 1H), 6.84 - 6.76 (m, 2H), 6.42 (s, 1H), 5.16 - 4.79 (m, 2H), 4.30 - 4.09 (m, 4H), 4.02 - 3.89 (m, 2H), 2.91 - 2.80 (m, 2H), 2.70 - 2.59 (m, 1H), 1.86 - 1.74 (m, 2H), 1.68 - 1.54 (m, 2H).

Example 58

Methyl 4-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyra zin-2- yl] carbamoyl } piperidine- 1 -carboxylate

Rt (Method B) 3.09 mins, m/z 451 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 10.38 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.24 - 7.18 (m, 1H), 7.10 - 7.03 (m, 1H), 6.98 (s, 1H), 6.41 (s, 1H), 5.15 - 4.77 (m, 2H), 4.28 - 4.10 (m, 4H), 4.07 - 3.91 (m, 2H), 3.59 (s, 3H), 2.91 - 2.72 (m, 2H), 2.60 - 2.52 (m, 1H), 1.79 - 1.68 (m, 2H), 1.54 - 1.40 (m, 2H).

Example 59

3-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]p yrazin-2-yl]amino}-2,2- dimethylpropan- 1 -ol

Rt (Method A) 2.97 mins, m/z 368 [M+H]+

1H NMR (400 MHz, DMSO-d6) 5 11.67 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.3 Hz, 1H), 7.25 - 7.17 (m, 1H), 7.09 - 7.02 (m, 1H), 6.94 (s, 1H), 5.38 (s, 1H), 5.23 (t, J = 6.5 Hz, 1H), 5.01 - 4.69 (m, 3H), 4.24 - 4.08 (m, 2H), 4.02 - 3.91 (m, 2H), 3.10 (s, 2H), 2.87 (d, J = 6.5 Hz, 2H), 0.80 (s, 6H).

Example 60

5-(lH-indole-2-carbonyl)-N-[(l-methoxycyclobutyl)methyl]- 4H,5H,6H,7H-pyrazolo[l,5- a]pyrazin-2-amine

Rt (Method B) 3.21 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) 5 11.67 (s, 1H), 7.64 (d, I = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.21 (t, J = 7.7 Hz, 1H), 7.07 (t, I = 7.4 Hz, 1H), 6.95 (s, 1H), 5.44 (s, 1H), 5.03 - 4.75 (m, 3H), 4.22 - 4.11 (m, 2H), 4.05 - 3.94 (m, 2H), 3.23 (d, J = 6.0 Hz, 2H), 3.07 (s, 3H), 2.06 - 1.94 (m, 2H), 1.94 - 1.83 (m, 2H), 1.74 - 1.48 (m, 2H).

Example 61

N- [5-( 1 H-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l ,5-a]pyrazin-2-yl] -2- (trifluoromethyl)piperidine-4-carboxamide

Rt (Method B) 2.54 mins, m/z 461 [M+H]+

Example 62

1 -(2-hydroxyethyl)-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-py razolo[ 1 ,5-a]pyrazin-2- yl]piperidine-4-carboxamide

Rt (Method B) 2.39 mins, m/z 437 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.69 (s, 1H), 10.30 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.25 - 7.17 (m, 1H), 7.10 - 7.04 (m, 1H), 7.00 - 6.95 (m, 1H), 6.42 (s, 1H), 5.14 - 4.76 (m, 2H), 4.46 - 4.30 (m, 1H), 4.30 - 4.07 (m, 4H), 3.51 - 3.43 (m, 2H), 2.95 - 2.80 (m, 2H), 2.40 - 2.33 (m, 2H), 2.33 - 2.24 (m, 1H), 2.01 - 1.84 (m, 2H), 1.72 - 1.49 (m, 4H).

Example 63

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l-(2,2,2- trifluoroethyl)piperidine-4-carboxamide

Rt (Method B) 2.95 mins, m/z 475 [M+H]+

Example 64

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l- methanesulfonylpiperidine-4-carboxamide

Rt (Method B) 3.09 mins, m/z 471 [M+H]+

Example 65

N4-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]p yrazin-2-yl]piperidine-l,4- dicarboxamide

Rt (Method B) 2.85 mins, m/z 436 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.70 (s, 1H), 10.37 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.11 - 7.04 (m, 1H), 6.98 (s, 1H), 6.42 (s, 1H), 5.92 (s, 2H), 5.15 - 4.81 (m, 2H), 4.31 - 4.07 (m, 4H), 4.02 - 3.88 (m, 2H), 2.71 - 2.59 (m, 2H), 2.57 - 2.51 (m, 1H), 1.73 - 1.60 (m, 2H), 1.53 - 1.35 (m, 2H).

Example 66 l-acetyl-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l, 5-a]pyrazin-2-yl]azetidme-3- carboxamide

Rt (Method B) 3.18 mins, m/z 407 [M+H]+

Example 67 l-[5-(lH-indole-2-carbonyI)-4H,5H,6H,7H-pyrazolo[l,5-a]pyraz in-2-yl]-3-methylurea

Rt (Method A) 2.69 mins, m/z 339 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.69 (s, 1H), 8.79 (s, 1H), 7.64 (d, I = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.26 - 7.17 (m, 1H), 7.11 - 7.04 (m, 1H), 7.00 - 6.93 (m, 1H), 6.74 - 6.57 (m, 1H), 6.01 (s, 1H), 5.25 - 4.64 (m, 2H), 4.32 - 3.98 (m, 4H), 2.65 (d, I = 4.6 Hz, 3H).

Example 68

N- [5-( 1 H-indole-2-carbony!)-4H,5H,6H,7H-pyrazolo[ 1 ,5 -a]pyrazin-2-yl] -5H,6H,7H,8H- imidazo [1,2 -a]pyridine- 7-carboxamide

Rt (Method B) 2.4 mins, m/z 430 [M+H]+ 1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.57 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.45 (d, J = 8.3 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.11 - 7.04 (m, 1H), 7.02 - 6.95 (m, 2H), 6.81 (d, J = 1.2 Hz, 1H), 6.46 (s, 1H), 5.16 - 4.78 (m, 2H), 4.30 - 4.11 (m, 4H), 4.11 - 4.01 (m, 1H), 3.93 - 3.80 (m, 1H), 3.01 - 2.88 (m, 2H), 2.86 - 2.73 (m, 1H), 2.21 - 2.10 (m, 1H), 2.05 - 1.90 (m, 1H).

Example 69

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l-(oxan-4- yl)piperidine-4-carboxamide

Rt (Method A) 2.81 mins, m/z 477 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.69 (s, 1H), 10.30 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.26 - 7.18 (m, 1H), 7.12 - 7.03 (m, 1H), 7.01 - 6.94 (m, 1H), 6.42 (s, 1H), 5.25 - 4.74 (m, 2H), 4.31 - 4.07 (m, 4H), 3.94 - 3.81 (m, 2H), 3.27 - 3.20 (m, 2H), 2.98 - 2.83 (m, 2H), 2.46 - 2.36 (m, 1H), 2.36 - 2.24 (m, 1H), 2.17 - 2.03 (m, 2H), 1.81 - 1.49 (m, 6H), 1.49 - 1.33 (m, 2H).

Example 70

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l-methylazetidine-3- carboxamide

Rt (Method B) 2.31 mins, m/z 397 [M+H]+ 1H NMR (400 MHz, DMSO-d6) 6 11.67 (s, 1H), 10.33 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d,

I = 8.2 Hz, 1H), 7.25 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 7.00 - 6.92 (m, 1H), 6.44 (s, 1H), 5.17 - 4.79 (m, 2H), 4.31 - 4.03 (m, 4H), 3.46 - 3.38 (m, 1H), 3.31 - 3.20 (m, 2H), 3.09 (t, J = 6.5 Hz, 2H), 2.17 (s, 3H).

Example 71 l-({[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyr azin-2- yl] amino }methyl)cyclobutan- 1 -ol

Rt (Method B) 3.19 mins, m/z 366 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.21 (t, J = 7.5 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.95 (s, 1H), 5.44 (s, 1H), 5.17 (s, 1H), 5.00 - 4.73 (m, 3H), 4.24 - 4.12 (m, 2H), 4.03 - 3.95 (m, 2H), 3.09 (d, J = 6.0 Hz, 2H), 2.05 - 1.95 (m, 2H), 1.95 - 1.83 (m, 2H), 1.73 - 1.56 (m, 1H), 1.53 - 1.38 (m, 1H).

Example 72

3-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]p yrazin-2-yl]amino}-2- methylpropan- 1 -ol

Rt (Method A) 2.78 mins, m/z 354 [M+H]+ *H NMR (400 MHz, DMSO-d6) d 11.66 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.25 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.96 - 6.91 (m, 1H), 5.36 (s, 1H), 5.16 (t, J = 6.1 Hz, 1H), 5.01 - 4.72 (m, 2H), 4.57 - 4.43 (m, 1H), 4.23 - 4.10 (m, 2H), 4.04 - 3.91 (m, 2H), 3.31 - 3.20 (m, 2H), 3.04 - 2.94 (m, 1H), 2.88 - 2.77 (m, 1H), 1.83 - 1.69 (m, 1H), 0.84 (d, J = 6.8 Hz, 3H).

Kxsiuplc 73

3,3-difluoro-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyra zolo[l,5-a]pyrazin-2-yl]piperidine-

4-carboxamide

Rt (Method B) 2.28 mins, m/z 429 [M+H]+

1H NMR (400 MHz, DMSO-d6) 6 11.69 (s, 1H), 10.49 (s, 1H), 7.64 (d, I = 8.0 Hz, 1H), 7.44 (d,

J = 8.2 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.10 - 7.04 (m, 1H), 7.01 - 6.95 (m, 1H), 6.49 - 6.38 (m, 1H), 5.27 - 4.73 (m, 2H), 4.39 - 4.01 (m, 4H), 3.32 - 3.20 (m, 1H), 3.22 - 3.02 (m, 2H), 2.97 - 2.86 (m, 1H), 2.80 - 2.68 (m, 1H), 1.93 - 1.80 (m, 1H), 1.80 - 1.69 (m, 1H).

Example 74 l-cyclopropyl-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazo lo[l,5-a]pyrazin-2- yl]piperidine-4-carboxamide

Rt (Method B) 2.31 mins, m/z 433.2 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.69 (s, 1H), 10.31 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.1 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.10 - 7.03 (m, 1H), 6.98 (s, 1H), 6.41 (s, 1H), 4.96 (s, 2H), 4.27 - 4.09 (m, 5H), 3.01 - 2.89 (m, 2H), 2.37 - 2.28 (m, 1H), 2.18 - 2.07 (m, 3H), 1.71 - 1.62 (m, 2H), 1.60 - 1.45 (m, 3H), 0.43 - 0.34 (m, 2H), 0.31 - 0.25 (m, 2H).

Example 75

5-(l H-indole-2-carbonyl)-N- [(3 -methyloxolan-3 -yl)methyl] -4H,5H,6H,7H-pyrazolo [1,5- a]pyrazin-2-amine

Rt (Method A) 2.92 mins, m/z 380.1 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.20 (t, J = 7.4 Hz, 1H), 7.06 (t, J = 7.5 Hz, 1H), 6.94 (s, 1H), 5.38 (s, 1H), 5.26 (t, J = 6.4 Hz, 1H), 5.04 - 4.72 (m, 2H), 4.25 - 4.07 (m, 2H), 4.07 - 3.91 (m, 2H), 3.80 - 3.65 (m, 2H), 3.55 (d, J = 8.3 Hz, 1H), 3.26 (d, I = 8.2 Hz, 1H), 3.01 (d, J = 6.5 Hz, 2H), 1.88 - 1.75 (m, 1H), 1.60 - 1.45 (m, 1H), 1.06 (s, 3H).

Example 76

(lr,3r)-3-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo [l,5-a]pyrazin-2- yl] amino } eyclobutan- 1 -ol

Rt (Method A) 2.62 mins, m/z 352 [M+H]+

Example 77

(lR,3S)-3-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo [l,5-a]pyrazin-2- yl] amino } cyclohexan- l-ol

Rt (Method A) 2.76 mins, m/z 380 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 7.63 (d, I = 8.0 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.21 (t, J = 7.6 Hz, 1H), 7.06 (t, I = 7.5 Hz, 1H), 6.96 - 6.90 (m, 1H), 5.34 (s, 1H), 4.99 (d, J = 8.4 Hz, 1H), 4.95 - 4.78 (m, 2H), 4.53 (d, J = 4.2 Hz, 1H), 4.21 - 4.11 (m, 2H), 4.03 - 3.93 (m, 2H), 3.42 - 3.35 (m, 1H), 3.17 - 3.06 (m, 1H), 2.18 - 2.06 (m, 1H), 1.92 - 1.82 (m, 1H), 1.81 - 1.71 (m, 1H), 1.68 - 1.58 (m, 1H), 1.26 - 1.12 (m, 1H), 1.08 - 0.85 (m, 3H).

Example 78

5-(lH-indole-2-carbonyl)-N-{[l-(propan-2-yloxy)cyclobutyl ]methyl}-4H,5H,6H,7H- pyrazo!o[ 1 ,5-a]pyrazin-2-amine

Rt (Method A) 3.37 mins, m/z 408 [M+H]+

Example 79

[l-({[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a ]pyrazin-2- yl] amino}methyl)cyclobutyl]methanol

Rt (Method A) 2.99 mins, m/z 380 [M+H]+

Example 80

5-(lH-indole-2-carbonyl)-N-(4-methoxycyclohexyl)-4H,5H,6H ,7H-pyrazolo[l,5-a]pyrazin-2- amine

Rt (Method A) 3.04 mins, m/z 394 [M+H]+

Example 81

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-l -methyl-2- (trifluoromethyl)piperidine-4-carboxamide

Rt (Method B) 2 51 mins, m/z 457 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.43 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.21 (t, J = 7.7 Hz, 1H), 7.07 (t, J = 7.4 Hz, 1H), 6.98 (s, 1H), 6.42 (s, 1H), 4.97 (s, 2H), 4.26 - 4.10 (m, 4H), 2.95 - 2.86 (m, 1H), 2.84 - 2.71 (m, 1H), 2.48 - 2.42 (m, 1H), 2.35 -

2.21 (m, 4H), 1.94 - 1.86 (m, 1H), 1.78 - 1.69 (m, 1H), 1.63 - 1.49 (m, 2H).

Example 82

N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]py razin-2-yl]-2- azabicyclo[2.2.1 ]heptane-5-carboxamide dihydrochloride

Rt (Method B) 2.29 mins, m/z 405 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.72 - 11.64 (m, 1H), 10.56 (s, 1H), 8.84 - 8.65 (m, 1H), 8.48 - 8.32 (m, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.25 - 7.18 (m, 1H), 7.07 (t, J = 7.5 Hz, 1H), 7.00 - 6.95 (m, 1H), 6.41 (s, 1H), 5.09 - 4.86 (m, 2H), 4.31 - 4.01 (m, 5H), 3.10 - 2.99 (m, 1H), 2.93 - 2.83 (m, 1H), 2.75 - 2.66 (m, 2H), 2.06 - 1.92 (m, 2H), 1.82 - 1.72 (m, 1H), 1.59 - 1.51 (m, 1H), 1.28 - 1.21 (m, 1H).

Example 83

3,3-difluoro-l-({[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-py razolo[l,5-a]pyrazin-2- y!] amino } methyl)cyclobutan- 1 -ol

Rt (Method B) 3 mins, m/z 402 [M+H]+

1H NMR (400 MHz, DMSO-d6) 8 11.67 (s, 1H), 7.63 (d, I = 8.0 Hz, 1H), 7.43 (d, I = 8.1 Hz, 1H), 7.24 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.94 (s, 1H), 5.68 (s, 1H), 5.43 (s, 1H), 5.26 (t, J = 6.3 Hz, 1H), 5.01 - 4.76 (m, 2H), 4.27 - 4.11 (m, 2H), 4.07 - 3.93 (m, 2H), 3.17 (d, I = 6.2 Hz, 2H), 2.82 - 2.68 (m, 2H), 2.48 - 2.39 (m, 2H).

Example 84

5-(lH-indole-2-carbonyl)-N-(l-phenylcyclopropyl)-4H,5H,6H ,7H-pyrazolo[l,5-a]pyrazin-2- amine

Rt (Method B) 3.4 mins, m/z 398 [M+H]+ 1H NMR (400 MHz, DMSO-d6) d 11.63 (s, 1H), 7.61 (d, J = 8.0 Hz, 1H), 7.42 (d, J = 8.2 Hz, 1H), 7.25 - 7.16 (m, 5H), 7.12 - 7.02 (m, 2H), 6.91 (s, 1H), 6.38 (s, 1H), 5.29 (s, 1H), 4.83 (s, 2H), 4.23 - 4.08 (m, 2H), 4.05 - 3.91 (m, 2H), 1.19 - 1.07 (m, 4H).

Example 85

3-fluoro-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo [l,5-a]pyrazm-2-yl]piperidine-4- carboxamide trihydrochloride

Rt (Method B) 2,29 & 2,33 mins, m/z 411 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.76 - 11.63 (m, 2H), 10.80 (s, 1H), 10.62 (s, 1H), 9.51 - 8.56 (m, 3H), 7.64 (d, J = 8.1 Hz, 2H), 7.45 (d, J = 8.3 Hz, 2H), 7.22 (t, J = 7.6 Hz, 2H), 7.07 (t, I = 7.5 Hz, 2H), 7.00 - 6.96 (m, 2H), 6.47 - 6.40 (m, 2H), 5.36 (d, J = 46.5 Hz, 1H), 5.16 - 4.77 (m, 4H), 4.40 - 3.99 (m, 8H), 3.67 - 3.56 (m, 3H), 3.32 - 3.07 (m, 4H), 3.06 - 2.85 (m, 4H), 2.22 - 0.74 (m, 6H).

Example 86

(3R,4R)-3-fluoro-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H- pyrazolo[l,5-a]pyrazin-2- yl]piperidine-4-carboxamide dihydrochloride

Rt (Method B) 2.28 mins, m/z 411 [M+H]+ l E NMR (400 MHz, DMSO-d6) 6 11.68 (s, 1H), 10.79 (s, 1H), 9.29 - 8.87 (m, 2H), 7.64 (d, J = 7.9 Hz, 1H), 7.45 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 7.6 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 7.01 - 6.95 (m, 1H), 6.45 (s, 1H), 5.23 - 4.88 (m, 3H), 4.20 (d, J = 24.7 Hz, 4H), 3.63 - 3.52 (m, 2H), 3.20 - 3.12 (m, 1H), 3.05 - 2.86 (m, 2H), 2.12 - 2.03 (m, 1H), 1.93 - 1.75 (m, 1H), 1.31 - 1.13 (m,

1H).

Example 87

3,3-difluoro-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyra zolo[l,5-a]pyrazin-2-yl]-l- methylpiperidine-4-carboxamide

Rt (Method B) 2.31 mins, m/z 443 [M+H]+

1H NMR (400 MHz, DMSO-d6) 6 11.67 (s, 1H), 10.51 (s, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.44 (d,

J = 8.3 Hz, 1H), 7.21 (t, J = 7.5 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.98 (s, 1H), 6.44 (s, 1H), 4.98 (s, 2H), 4.35 - 4.09 (m, 4H), 3.03 - 2.93 (m, 2H), 2.76 - 2.71 (m, 1H), 2.54 (s, 1H), 2.36 - 2.22 (m, 4H), 2.20 - 2.07 (m, 1H), 2.00 - 1.88 (m, 1H), 1.85 - 1.73 (m, 1H).

Example 88

3-fluoro-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo [l,5-a]pyrazin-2-yl]-l- methylpiperidine-4-carboxamide

Rt (Method B) 2,21 & 2,26 mins, m/z 425 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.68 (s, 2H), 10.56 (s, 1H), 10.38 (s, 1H), 7.64 (d, J = 7.9 Hz, 2H), 7.44 (d, I = 8.3 Hz, 2H), 7.21 (t, J = 7.5 Hz, 2H), 7.07 (t, I = 7.4 Hz, 2H), 6.98 (s, 2H), 6.49 - 6.39 (m, 2H), 5.13 - 4.88 (m, 5H), 4.85 - 4.63 (m, 1H), 4.29 - 4.08 (m, 8H), 3.17 - 3.08 (m, 1H), 3.08 - 2.99 (m, 1H), 2.83 - 2.69 (m, 2H), 2.62 - 2.56 (m, 1H), 2.26 - 2.06 (m, 7H), 2.02 - 1.77 (m, 6H), 1.62 - 1.53 (m, 2H).

Example 89

(3R,4R)-3-fluoro-N-[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H- pyrazolo[l,5-a]pyrazm-2-yl]-l- methylpiperidine-4-carboxamide

Rt (Method B) 2.24 mins, m/z 425 [M+H]+ l H NMR (400 MHz, DMSO-d6) d 11.68 (s, 1H), 10.56 (s, 1H), 7.64 (d, I = 8.0 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.21 (t, I = 7.5 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.98 (s, 1H), 6.45 (s, 1H), 5.10 - 4.86 (m, 2H), 4.84 - 4.61 (m, 1H), 4.19 (d, J = 26.3 Hz, 4H), 3.16 - 3.07 (m, 1H), 2.77 - 2.69 (m, 1H), 2.22 (s, 3H), 1.95 - 1.74 (m, 3H), 1.66 - 1.48 (m, 1H), 1.42 - 1.10 (m, 1H).

Example 90 (l-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l,5-a]pyr azin-2- yl] amino } cyc!obutyl)methano!

Rt (Method B) 2.72 mins, m/z 366 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.66 (s, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.43 (d, J = 8.1 Hz, 1H), 7.21 (t, J = 7.5 Hz, 1H), 7.06 (t, J = 7.4 Hz, 1H), 6.94 (s, 1H), 5.38 (s, 1H), 5.12 (s, 1H), 5.03 - 4.59 (m, 3H), 4.28 - 4.09 (m, 2H), 4.09 - 3.89 (m, 2H), 3.51 (s, 2H), 2.18 - 2.03 (m, 2H), 2.03 - 1.89 (m, 2H), 1.85 - 1.55 (m, 2H).

Example 91

(ls,3s)-3-{[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo [l,5-a]pyrazin-2- yl] amino } cyclobutan- 1 -ol

Rt (Method B) 2.49 mins, m/z 352 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.43 (d, I = 8.3 Hz, 1H), 7.24 - 7.17 (m, 1H), 7.10 - 7.02 (m, 1H), 6.96 - 6.91 (m, 1H), 5.36 - 5.28 (m, 2H), 5.01 - 4.78 (m, 3H), 4.20 - 4.10 (m, 2H), 4.03 - 3.93 (m, 2H), 3.82 - 3.70 (m, 1H), 3.30 - 3.21 (m, 1H), 2.59 - 2.51 (m, 2H), 1.67 - 1.56 (m, 2H). Example 92

(1 - {[5-(lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l ,5-a]pyrazin-2- yl] amino } cy clopropyl)methanol

Rt (Method B) 2.58 mins, m/z 352 [M+H]+ l R NMR (400 MHz, DMSO-d6) 8 11.67 (s, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.44 (d, J = 8.2 Hz, 1H), 7.26 - 7.17 (m, 1H), 7.11 - 7.04 (m, 1H), 6.95 (d, J = 1.6 Hz, 1H), 5.55 (s, 1H), 5.44 (s, 1H), 5.13 - 4.73 (m, 2H), 4.59 (t, J = 5.6 Hz, 1H), 4.30 - 4.09 (m, 2H), 4.09 - 3.93 (m, 2H), 3.42 (d, J = 5.6 Hz, 2H), 0.67 - 0.60 (m, 2H), 0.57 - 0.52 (m, 2H).

Example 93 l-({[5-(4-ethyl-lH-indole-2-carbonyl)-4H,5H,6H,7H-pyrazolo[l ,5-a]pyrazin-2- yl] amino } methyl)cyclobutan- 1 -ol

Rt (Method A) 3.08 mins, m/z 394 [M+H]+

1H NMR (400 MHz, DMSO-d6) d 11.61 (s, 1H), 7.26 (d, J = 8.3 Hz, 1H), 7.13 (dd, J = 8.3, 7.1 Hz, 1H), 6.98 (s, 1H), 6.88 (d, J = 6.9 Hz, 1H), 5.44 (s, 1H), 5.19 - 5.12 (m, 1H), 4.96 - 4.83 (m, 3H), 4.22 - 4.13 (m, 2H), 4.03 - 3.95 (m, 2H), 3.10 (d, I = 6.1 Hz, 2H), 2.90 (q, J = 7.6 Hz, 2H), 2.04 - 1.95 (m, 2H), 1.95 - 1.84 (m, 2H), 1.68 - 1.57 (m, 1H), 1.52 - 1.38 (m, 1H), 1.29 (t, J = 7.5 Hz, 3H). Example 94

5-(lH-indole-2-carbonyl)-N-[(3-methoxyoxolan-3-yl)methyl] -4H,5H,6H,7H-pyrazolo[l,5- a]pyrazin-2-amine

Rt (Method B) 2.79 mins, m/z 396 [M+HJ+

1H NMR (400 MHz, DMSO-d6) d 11.67 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 8.3 Hz, 1H), 7.21 (t, J = 7.7 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.95 (s, 1H), 5.43 (s, 1H), 5.07 (t, J = 5.7

Hz, 1H), 5.01 - 4.71 (m, 2H), 4.31 - 4.08 (m, 2H), 4.08 - 3.90 (m, 2H), 3.84 - 3.63 (m, 3H), 3.56 (d, J = 9.5 Hz, 1H), 3.30 - 3.20 (m, 2H), 3.16 (s, 3H), 2.05 - 1.94 (m, 1H), 1.88 - 1.77 (m, 1H).

Selected compounds of the invention were assayed in capsid assembly and HBV replication assays, as described below and a representative group of these active compounds is shown in Table 1.

Biochemical capsid assembly assay

The screening for assembly effector activity was done based on a fluorescence quenching assay published by Zlotnick et al. (2007). The C-terminal truncated core protein containing 149 amino acids of the N-terminal assembly domain fused to a unique cysteine residue at position 150 and was expressed in E. coli using the pET expression system (Merck Chemicals, Darmstadt). Purification of core dimer protein was performed using a sequence of size exclusion chromatography steps. In brief, the cell pellet from 1 L BL21 (DE3) Rosetta2 culture expressing the coding sequence of core protein cloned Ndel/ Xhol into expression plasmid pET2lb was treated for 1 h on ice with a native lysis buffer (Qproteome Bacterial Protein Prep Kit; Qiagen, Hilden). After a centrifugation step the supernatant was precipitated during 2 h stirring on ice with 0.23 g/ml of solid ammonium sulfate. Following further centrifugation the resulting pellet was resolved in buffer A (lOOmM Tris, pH 7.5; lOOmM NaCl; 2mM DTT) and was subsequently loaded onto a buffer A equilibrated CaptoCore 700 column (GE HealthCare, Frankfurt). The column flow through containing the assembled HBV capsid was dialyzed against buffer N (50mM NaHC03 pH 9.6; 5mM DTT) before urea was added to a final concentration of 3M to dissociate the capsid into core dimers for 1.5 h on ice. The protein solution was then loaded onto a 1L Sephacryl S300 column. After elution with buffer N core dimer containing fractions were identified by SDS-PAGE and subsequently pooled and dialyzed against 50mM HEPES pH 7.5; 5mM DTT. To improve the assembly capacity of the purified core dimers a second round of assembly and disassembly starting with the addition of 5 M NaCl and including the size exclusion chromatography steps described above was performed. From the last chromatography step core dimer containing fractions were pooled and stored in aliquots at concentrations between 1.5 to 2.0 mg/ml at -80°C.

Immediately before labelling the core protein was reduced by adding freshly prepared DTT in a final concentration of 20 mM. After 40 min incubation on ice storage buffer and DTT was removed using a Sephadex G-25 column (GE HealthCare, Frankfurt) and 50 mM HEPES, pH 7.5. For labelling 1.6 mg/ml core protein was incubated at 4°C and darkness overnight with BODIPY-FL maleimide (Invitrogen, Karlsruhe) in a final concentration of 1 mM. After labelling the free dye was removed by an additional desalting step using a Sephadex G-25 column. Labelled core dimers were stored in aliquots at 4°C. In the dimeric state the fluorescence signal of the labelled core protein is high and is quenched during the assembly of the core dimers to high molecular capsid structures. The screening assay was performed in black 384 well microtiter plates in a total assay volume of 10 mΐ using 50 mM HEPES pH 7.5 and 1.0 to 2.0 mM labelled core protein. Each screening compound was added in 8 different concentrations using a 0.5 log-unit serial dilution starting at a final concentration of 100 mM, 31.6 mM or 10 mM, In any case the DMSO concentration over the entire microtiter plate was 0.5%. The assembly reaction was started by the injection of NaCl to a final concentration of 300 mM which induces the assembly process to approximately 25% of the maximal quenched signal. 6 min after starting the reaction the fluorescence signal was measured using a Clariostar plate reader (BMG Labtech, Ortenberg) with an excitation of 477 ran and an emission of 525 nm. As 100% and 0% assembly control HEPES buffer containing 2.5 M and 0 M NaCl was used. Experiments were performed thrice in triplicates. EC 50 values were calculated by non-linear regression analysis using the Graph Pad Prism 6 software (GraphPad Software, La Jolla, USA).

Determination of HBV DNA from the supernatants of HepAD38 cells The anti-HBV activity was analysed in the stable transfected cell line HepAD38, which has been described to secrete high levels of HBV virion particles (Ladner et al., 1997). In brief, HepAD38 cells were cultured at 37°C at 5% C0 2 and 95% humidity in 200 mΐ maintenance medium, which was Dulbecco's modified Eagle’s medium/ Nutrient Mixture F-12 (Gibco, Karlsruhe), 10% fetal bovine serum (PAN Biotech Aidenbach) supplemented with 50 pg/ml penicillin/ streptomycin (Gibco, Karlsruhe), 2 mM L- glutamine (PAN Biotech, Aidenbach), 400 pg/ml G418 (AppliChem, Darmstadt) and 0.3 pg/ml tetracycline. Cells were subcultured once a week in a 1:5 ratio, but were usually not passaged more than ten times. For the assay 60,000 cells were seeded in maintenance medium without any tetracycline into each well of a 96-well plate and treated with serial half-log dilutions of test compound. To minimize edge effects the outer 36 wells of the plate were not used but were filled with assay medium. On each assay plate six wells for the virus control (untreated HepAD38 cells) and six wells for the cell control (HepAD38 cells treated with 0.3 pg/ml tetracycline) were allocated, respectively. In addition, one plate set with reference inhibitors like BAY 41-4109, entecavir, and lamivudine instead of screening compounds were prepared in each experiment. In general, experiments were performed thrice in triplicates. At day 6 HBV DNA from 100 pl filtrated cell culture supernatant (AcroPrep Advance 96 Filter Plate, 0.45 pM Supor membran, PALL GmbH, Dreieich) was automatically purified on the MagNa Pure LC instrument using the MagNA Pure 96 DNA and Viral NA Small Volume Kit (Roche Diagnostics, Mannheim) according to the instructions of the manufacturer. EC50 values were calculated from relative copy numbers of HBV DNA In brief, 5 pl of the 100 pl eluate containing HBV DNA were subjected to PCR LC480 Probes Master Kit (Roche) together with 1 pM antisense primer tgcagaggtgaagcgaagtgcaca, 0.5 pM sense primer gacgtcctttgtttacgtcccgtc, 0.3 pM hybprobes acggggcgcacctctctttacgcgg-FL and LC640- ctccccgtctgtgccttctcatctgc-PH (TIBMolBiol, Berlin) to a final volume of 12.5 pl. The PCR was performed on the Light Cycler 480 real time system (Roche Diagnostics, Mannheim) using the following protocol: Pre-incubation for 1 min at 95°C, amplification: 40 cycles x (10 sec at 95°C, 50 sec at 60°C, 1 sec at 70°C), cooling for 10 sec at 40°C. Viral load was quantitated against known standards using HBV plasmid DNA of pCH-9/309l (Nassal et al., 1990, Cell 63: 1357— 1363) and the LightCycler 480 SW 1.5 software (Roche Diagnostics, Mannheim) and EC 50 values were calculated using non-linear regression with GraphPad Prism 6 (GraphPad Software Inc., La Jolla, USA).

Cell Viability Assay Using the AlamarBlue viability assay cytotoxicity was evaluated in HepAD38 cells in the presence of 0.3 pg/ml tetracycline, which blocks the expression of the HBV genome. Assay condition and plate layout were in analogy to the anti-HBV assay, however other controls were used. On each assay plate six wells containing untreated HepAD38 cells were used as the 100% viability control, and six wells filled with assay medium only were used as 0% viability control. In addition, a geometric concentration series of cycloheximide starting at 60 mM final assay concentration was used as positive control in each experiment. After six days incubation period Alamar Blue Presto cell viability reagent (ThermoFisher, Dreieich) was added in 1/11 dilution to each well of the assay plate. After an incubation for 30 to 45 min at 37°C the fluorescence signal, which is proportional to the number of living cells, was read using a Tecan Spectrafluor Plus plate reader with an excitation filter 550 nm and emission filter 595 nm, respectively. Data were normalized into percentages of the untreated control (100% viability) and assay medium (0% viability) before CC50 values were calculated using non-linear regression and the GraphPad Prism 6.0 (GraphPad Software, La Jolla, USA). Mean EC50 and CC50 values were used to calculate the selectivity index (SI = CC50/EC50) for each test compound.

In vivo efficacy models

HBV research and preclinical testing of antiviral agents are limited by the narrow species- and tissue-tropism of the virus, the paucity of infection models available and the restrictions imposed by the use of chimpanzees, the only animals fully susceptible to HBV infection. Alternative animal models are based on the use of HBV-related hepadnaviruses and various antiviral compounds have been tested in woodchuck hepatitis vims (WHV) infected woodchucks or in duck hepatitis B virus (DHBV) infected ducks or in woolly monkey HBV (WM-HBV) infected tupaia (overview in Dandri et al., 2017, Best Pract Res Clin Gastroenterol 31, 273-279). However, the use of surrogate viruses has several limitations. For example is the sequence homology between the most distantly related DHBV and HBV is only about 40% and that is why core protein assembly modifiers of the HAP family appeared inactive on DHBV and WHV but efficiently suppressed HBV (Campagna et al., 2013, J. Virol. 87, 6931-6942). Mice are not HBV permissive but major efforts have focused on the development of mouse models of HBV replication and infection, such as the generation of mice transgenic for the human HBV (HBV tg mice), the hydrodynamic injection (HDI) of HBV genomes in mice or the generation of mice having humanized livers and/ or humanized immune systems and the intravenous injection of viral vectors based on adenoviruses containing HBV genomes (Ad-HBV) or the adenoassociated virus (AAV-HBV) into immune competent mice (overview in Dandri et al., 2017, Best Pract Res Clin Gastroenterol 31, 273-279).. Using mice transgenic for the full HBV genome the ability of murine hepatocytes to produce infectious HBV virions could be demonstrated (Guidotti et al, 1995, I. Virol., 69: 6158-6169). Since transgenic mice are immunological tolerant to viral proteins and no liver injury was observed in HBV-producing mice, these studies demonstrated that HBV itself is not cytopathic. HBV transgenic mice have been employed to test the efficacy of several anti-HBV agents like the polymerase inhibitors and core protein assembly modifiers (Weber et al., 2002, Antiviral Research 54 69-78; Julander et al., 2003, Antivir. Res., 59: 155- 161), thus proving that HBV transgenic mice are well suitable for many type of preclinical antiviral testing in vivo.

As described in Paulsen et al., 2015, PLOSone, 10: eO 144383 HBV-transgenic mice (Tg [HBV 1.3 fsX 3’5’]) carrying a frameshift mutation (GC) at position 2916/2917 could be used to demonstrate antiviral activity of core protein assembly modifiers in vivo. In brief, The HBV- transgenic mice were checked for HBV-specific DNA in the serum by qPCR prior to the experiments (see section“Determination of HBV DNA from the supernatants of HepAD38 cells”). Each treatment group consisted of five male and five female animals approximately 10 weeks age with a titer of 10 -10 virions per ml serum. Compounds were formulated as a suspension in a suitable vehicle such as 2% DMSO / 98% tylose (0.5% Methylcellulose / 99.5% PBS) or 50% PEG400 and administered per os to the animals one to three times/day for a 10 day period. The vehicle served as negative control, whereas 1 pg/kg entecavir in a suitable vehicle was the positive control. Blood was obtained by retro bulbar blood sampling using an Isoflurane Vaporizer. For collection of terminal heart puncture six hours after the last treatment blood or organs, mice were anaesthetized with isoflurane and subsequently sacrificed by C0 2 exposure. Retro bulbar (100-150 mΐ) and heart puncture (400-500 mΐ) blood samples were collected into a Microvette 300 LH or Microvette 500 LH, respectively, followed by separation of plasma via centrifugation (10 min, 2000g, 4°C). Liver tissue was taken and snap frozen in liquid N 2 . All samples were stored at -80°C until further use. Viral DNA was extracted from 50 mΐ plasma or 25 mg liver tissue and eluted in 50 mΐ AE buffer (plasma) using the DNeasy 96 Blood & Tissue Kit (Qiagen, Hilden) or 320 mΐ AE buffer (liver tissue) using the DNeasy Tissue Kit (Qiagen, Hilden) according to the manufacturer’s instructions. Eluted viral DNA was subjected to qPCR using the LightCycler 480 Probes Master PCR kit (Roche, Mannheim) according to the manufacturer’s instructions to determine the HBV copy number. HBV specific primers used included the forward primer 5’-CTG TAC CAA ACC TTC GGA CGG-3’, the reverse primer 5’- AGG AGA AAC GGG CTG AGG C-3’ and the FAM labelled probe FAM-CCA TCA TCC TGG GCT TTC GGA AAA TT-BBQ. One PCR reaction sample with a total volume of 20 mΐ contained 5 mΐ DNA eluate and 15 mΐ master mix (comprising 0.3 mM of the forward primer, 0.3mM of the reverse primer, 0.15mM of the FAM labelled probe). qPCR was carried out on the Roche LightCyclerl480 using the following protocol: Pre-incubation for 1 min at 95°C, amplification: (10 sec at 95°C, 50 sec at 60°C, 1 sec at 70°C) x 45 cycles, cooling for 10 sec at 40°C. Standard curves were generated as described above. All samples were tested in duplicate. The detection limit of the assay is ~50 HBV DNA copies (using standards ranging from 250-2.5 x 107 copy numbers). Results are expressed as HBV DNA copies / 10m1 plasma or HBV DNA copies / lOOng total liver DNA (normalized to negative control).

It has been shown in multiple studies that not only transgenic mice are a suitable model to proof the antiviral activity of new chemical entities in vivo the use of hydrodynamic injection of HBV genomes in mice as well as the use of immune deficient human liver chimeric mice infected with HBV positive patient serum have also frequently used to profile drugs targeting HBV (Li et al., 2016, Hepat. Mon. 16: e34420; Qiu et al., 2016, J. Med. Chem. 59: 7651-7666; Lutgehetmann et al., 2011, Gastroenterology, 140: 2074-2083). In addition chronic HBV infection has also been successfully established in immunecompetent mice by inoculating low doses of adenovirus- (Huang et al., 2012, Gastroenterology 142: 1447-1450) or adeno-associated virus (AAV) vectors containing the HBV genome (Dion et al., 2013, J Virol. 87: 5554-5563). This models could also be used to demonstrate the in vivo antiviral activity of novel anti-HBV agents.

Table 1: Biochemical and antiviral activities

In Table 1, "+++" represents an EC50 < 1 mM; "++" represents 1 mM < ECso < 10 mM; "+" represents EC50 < 100 mM (Cell activity assay)

In Table 1, "A" represents an IC50 < 5 mM; "B" represents 5 mM < IC 5 o < 10 mM; "C" represents IC50 < 100 mM (Assembly assay activity)