Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
5-MEMBERED HETEROARYL CARBOXAMIDE COMPOUNDS FOR TREATMENT OF HBV
Document Type and Number:
WIPO Patent Application WO/2021/216660
Kind Code:
A1
Abstract:
The present disclosure provides, in part, 5-membered heteroaryl carboxamide compounds, and pharmaceutical compositions thereof, useful for dismption of HBV core protein assembly, and methods of treating Hepatitis B (HBV) infection.

Inventors:
HECKRODT THILO (US)
WALKER MICHAEL (US)
ZHONG MIN (US)
Application Number:
PCT/US2021/028327
Publication Date:
October 28, 2021
Filing Date:
April 21, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ASSEMBLY BIOSCIENCES INC (US)
International Classes:
C07D207/34; A61K31/40; A61K31/404; A61K31/437; A61P31/12; C07D209/42; C07D471/04
Domestic Patent References:
WO2014184365A12014-11-20
WO2015011281A12015-01-29
WO2014037480A12014-03-13
WO2014184328A12014-11-20
WO2013006394A12013-01-10
WO2014089296A22014-06-12
WO2014106019A22014-07-03
WO2013102655A12013-07-11
WO2014184350A12014-11-20
WO2014184365A12014-11-20
WO2014161888A12014-10-09
WO2014131847A12014-09-04
WO2014033176A12014-03-06
WO2014033167A12014-03-06
WO2014033170A12014-03-06
WO2013010069A12013-01-17
WO2014074906A12014-05-15
Other References:
THOMAS LAHLALI ET AL: "Novel Potent Capsid Assembly Modulators Regulate Multiple Steps of the Hepatitis B Virus Life Cycle", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 62, no. 10, 16 July 2018 (2018-07-16), US, pages 1 - 15, XP055584508, ISSN: 0066-4804, DOI: 10.1128/AAC.00835-18
CARREIRAKVAERNO: "Classics in Stereoselective Synthesis", 2009, WILEY-VCH
RAUTIOKUMPULAINEN ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 7, 2008, pages 255
WANG Y ET AL.: "Transbody against hepatitis B virus core protein inhibits hepatitis B virus replication in vitro", INT. IMMUNOPHARMACOL, 2014
Attorney, Agent or Firm:
FIX, Amy, H. et al. (US)
Download PDF:
Claims:
CLAIMS:

1. A compound of Formula I Formula I

, or a pharmaceutically acceptable salt thereof, wherein:

L is Ci-4alkylene or haloCi-4alkylene;

L1 is a bond, Ci-6alkylene, O, NRC, C(O), C(0)0, C(0)NRc, S(O), or S(0),NRc;

X1 is NRxl, O or S;

X3 is O, NR4, CR4R8, C(O), or S(O),;

X4 and X6 are independently O or S;

X5 is O, S or NR°;

Ra, Rb and Rc are independently selected for each occurrence from the group consisting of hydrogen, Ci-6 alkyl, haloCi-6 alkyl and C3-6 monocycloalkyl;

Rd is hydrogen, OH, Ci-6 alkyl or Ci-6 alkoxy;

Rxl is hydrogen, C1-4 alkyl, C1-4 alkenyl, C1-4 alkynyl, haloCi-4 alkyl, or C3-6 monocycloalkyl; or Rxl and R2 together form a -CH2CH2CH2-, -CH2CH2CH2CH2-, - CH2CH2O-, -CH2OCH2-, -CH2CH2CH2O- -CH2CH2OCH2-, -CH2CH2-NH- -CH2NHCH2-, - CH2CH2CH2NH- or -CH2CH2NHCH2- group;

R0a is independently selected for each occurrence from the group consisting of hydrogen, halogen, OH, CN, NO2, RaRbN-, Ci-4alkyl and haloCi-4alkyl;

R4a and R6a are independently hydrogen or C1-4 alkyl;

R°, R6 and R11 are independently selected for each occurrence from the group consisting of hydrogen, halogen, OH, CN, NO2, oxo, RdN=, hydrazino, formyl, azido, silyl, siloxy, HOC(O)-, RaRbN-, RaRbNS(0)t-, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, haloCi-6alkyl, hydroxyCi-ealkyl-, RaRbNCi-6alkyl-, HOC(0)Ci-6alkyl-, RaRbNCi-6alkylNRc-, Ci- 6alkylNRaCi-6alkylNRc-, Ci-6alkoxy, haloCi-6alkoxy, hydroxyCi-6alkoxy-, RaRbNCi-6alkoxy-, Ci-6alkoxyCi-6alkyl-, haloCi-6alkoxyCi-6alkyl-, RaRbNC(0)-, Ci-6alkylC(0)-, Ci- 6alkoxyC(0)-, Ci-6alkylC(0)0-, Ci-6alkylS(0)q-, Ci-6alkylS(0),NRc-, Ci-6alkylS(0),Ci-6alkyl- , Ci-6alkylS(0)tNRaCi-6alkyl-, C3-6cycloalkylS(0)tCi-6alkyl-, Ci-6alkylC(0)Ci-6alkyl-, and Ci- 6alkylC(0)0Ci-6alkyl-;

R1 is a phenyl or 5-6 membered monocyclic heteroaryl, wherein the phenyl or 5-6 membered monocyclic heteroaryl is optionally substituted with one, two, or three independently selected R11 groups;

R2, R2a and R8 are independently selected from the group consisting of hydrogen, halo, CN, OH, RaRbN, Ci-4alkyl, haloCi-4alkyl, C3-5monocycloalkyl, Ci-4alkoxy, and haloCi- 4alkoxy; or R2 and R2a together with the carbons to which they are attached form a phenyl, pyridyl, pyridazine, pyrimidine or pyrazine ring; or R2 and R2a together form a -CH2CH2CH2- , -CH2CH2CH2CH2-, -OCH2CH2-, -CH2OCH2-, -OCH2CH2CH2-, -CH2OCH2CH2-, - NHCH2CH2-, -CH2NHCH2-, -NHCH2CH2CH2- or -CH2NHCH2CH2- group;

R9 is R14S(0)q-L-, R14S(0)qNH-L-, or R14C(0)NH-L-;

R14is RaRbN-, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, Ci-6alkoxy, Ci- 6haloalkyl, Ci-6haloalkoxy, or R5-L1-; q, r, t, and w are independently selected for each occurrence from 0, 1 and 2; and v is independently selected for each occurrence from 0, 1, 2 and 3.

2. The compound of claim 1 , or a pharmaceutically acceptable salt thereof, wherein X1 is NRxl and Rxl is hydrogen or methyl.

3. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein Rxl is methyl.

4. The compound according to any one of Claims 1-3, or a pharmaceutically acceptable salt thereof, wherein: R1 is ; R11 is independently selected for each occurrence from the group consisting of halogen, CN, Ci-6 alkyl and haloCi-6 alkyl; and zl is 0, 1, 2 or 3.

5. The compound of Claim 4, or a pharmaceutically acceptable salt thereof, wherein for each occurrence R11 is independently selected from the group consisting of CN, F, Cl, Br and I.

6. The compound of Claim 5, or a pharmaceutically acceptable salt thereof, wherein

7. The compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein r is 0.

8. The compound of claims 7, or a pharmaceutically acceptable salt thereof, wherein X3 is C(O).

9. The compound of claim 7, or a pharmaceutically acceptable salt thereof, und of claim 9, or a pharmaceutically acceptable salt thereof,

11. The compound of claim 9 or 10, or a pharmaceutically acceptable salt thereof, wherein R4 is R6, and R6 is Ci-6 alkylS(0)tCi-6 alkyl- or Ci-6 alkylS(0)tNRaCi-6 alkyl-.

12. The compound of claim 9 or 10, or a pharmaceutically acceptable salt thereof, wherein R4 is R6, and R6 is hydrogen.

13. The compound according to any one of claims 9-12, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen, OH or Ci-6 alkoxy.

14. The compound of claim 13, or a pharmaceutically acceptable salt thereof, wherein R8 is OH.

15. A pharmaceutical composition comprising the compound according to any one of claims 1-14, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

16. A method of treating Hepatitis B (HBV) infection in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a compound according to any one of claims 1-14, or a pharmaceutically acceptable salt thereof.

17. A method of treating Hepatitis B (HBV) infection in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of pharmaceutical composition of claim 15.

Description:
5-MEMBERED HETEROARYL CARBOXAMIDE COMPOUNDS FOR

TREATMENT OF HBV

BACKGROUND

Hepatitis B (HBV) causes viral hepatitis that can further lead to chronic liver disease and increase the risk of liver cirrhosis and liver cancer (hepatocellular carcinoma). Worldwide, about 2 billion people have been infected with HBV, around 360 million people are chronically infected, and every year HBV infection causes more than one half million deaths. HBV can be spread by body fluids: from mother to child, by sex, and via blood products. Children bom to HBV-positive mothers may also be infected, unless vaccinated at birth.

The hepatitis virus particle is composed of a lipid envelope studded with surface protein (HBsAg) that surrounds the viral core. The core is composed of a protein shell, or capsid, built of 120 core protein (Cp) dimers, which in turn contains the relaxed circular DNA (rcDNA) viral genome as well as viral and host proteins. In an infected cell, the genome is found as a covalently closed circular DNA (cccDNA) in the host cell nucleus. The cccDNA is the template for viral RNAs and thus viral proteins. In the cytoplasm, Cp assembles around a complex of full-length viral RNA (the so-called pregenomic RNA or pgRNA and viral polymerase (P). After assembly, P reverse transcribes the pgRNA to rcDNA within the confines of the capsid to generate the DNA-filled viral core.

At present, chronic HBV is primarily treated with nucleos(t)ide analogs (e.g., entecavir) that suppress the vims while the patient remains on treatment, but do not eliminate the infection, even after many years of treatment. Once a patient starts taking nucleos(t)ide analogs, most must continue taking them or risk the possibility of a life-threatening immune response due to viral rebound. Further, nucleotide therapy may lead to the emergence of antiviral drug resistance.

The only FDA approved alternative to nucleos(t)ide analogs is treatment with interferon a or pegylated interferon a. Unfortunately, the adverse event incidence and profile of interferon a can result in poor tolerability, and many patients are unable to complete therapy. Moreover, only a small percentage of patients are considered appropriate for interferon therapy, as only a small subset of patients is likely to have a sustained clinical response to a course of interferon therapy. As a result, interferon-based therapies are used in only a small percentage of all diagnosed patients who elect treatment.

Thus, current HBV treatments can range from palliative to watchful waiting. Nucleotide analogs suppress virus production, treating the symptom, but leave the infection intact. Interferon a has severe side effects and less tolerability among patients and is successful as a finite treatment strategy in only a small minority of patients. There is a clear on-going need for more effective treatments for HBV infections.

SUMMARY

The present disclosure provides, in part, 5-membered heteroaryl carboxamide compounds and pharmaceutical compositions thereof, useful for disruption of HB V core protein assembly, and methods of treating HBV infections.

In one aspect, the disclosure provides a compound of Formula I: or a pharmaceutically acceptable salt thereof, where the variables are described in the detailed description.

In another aspect, the disclosure provides pharmaceutical compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

In another aspect, the disclosure provides a method of treating an HBV infection in a subject in need thereof, comprising: administering to the subject a therapeutically effective amount of compound of Formula I, or a pharmaceutically acceptable salt thereof.

In another aspect, the disclosure provides a method of treating an HBV infection in a subject in need thereof, comprising: administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

BRIEF DESCRIPTION OF DRAWINGS

FIGURE 1 shows the ORTEP plot for compound CP-AIA-227-2.

FIGURE 2 shows the relative stereochemistry scheme of compound CP-AIA-227-2.

DETAILED DESCRIPTION

The features and other details of the disclosure will now be more particularly described. Before further description of the present disclosure, certain terms employed in the specification, examples and appended claims are collected here. These definitions should be read in light of the remainder of the disclosure and as understood by a person of skill in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art.

I. Definitions

The term “alkenyl” as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond. Exemplary alkenyl groups include, but are not limited to, a straight or branched group of 2-6 carbon atoms, referred to herein as C2-6alkenyl. Exemplary alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, and pentenyl, etc.

The term “alkoxy” as used herein refers to a straight or branched alkyl group attached to oxygen (i.e., alkyl-O-). Exemplary alkoxy groups include, but are not limited to, alkoxy groups of 1-6 or 1-4 carbon atoms, referred to herein as Ci- 6 alkoxy and Ci-4alkoxy, respectively. Exemplary alkoxy groups include, but are not limited to methoxy, ethoxy, and isopropoxy, etc. The term “alkoxyalkyl” as used herein refers to an alkyl group substituted with an alkoxy group. Examples include, but are not limited to, CH3CH2OCH2-, CH3OCH2CH2-, and CH3OCH2-, etc.

The term “alkyl” as used herein refers to a saturated straight or branched hydrocarbon. Exemplary alkyl groups include, but are not limited to, straight or branched hydrocarbons of

1-6 or 1-4 carbon atoms, referred to herein as Ci-6 alkyl and C 1 -4 alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, 2- methyl-1 -butyl, 3-methyl-2-butyl, 2-methyl- 1-pentyl, 3-methyl- 1-pentyl, 4-methyl- 1 -pentyl,

2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl- 1 -butyl, 3,3- dimethyl- 1 -butyl, 2-ethyl- 1 -butyl, n-butyl, isobutyl, t-butyl, n-pentyl, isopentyl, neopentyl, and n-hexyl, etc. The term “alkylene” as used herein refers to a biradical alkyl group.

The term “alkynyl” as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond. Exemplary alkynyl groups include, but are not limited to, straight or branched groups of 2-6 carbon atoms, referred to herein as C2-6alkynyl. Exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and methylpropynyl, etc.

The term “carbonyl” as used herein refers to the biradical -C(O)-.

The term “cyano” as used herein refers to the radical -CN.

The term “cycloalkyl” as used herein refers to a saturated monocyclic hydrocarbon group of, for example, 3-6 carbons, referred to herein as C3-6 monocycloalkyl, or bicyclic hydrocarbon ring structure of, for example, 8-12 carbons, referred to herein as Cs- nbicycloalkyl. For bicyclic cycloalkyl groups, the two rings may be attached through the same or different carbons. Exemplary monocyclic cycloalkyl groups include, but are not limited to, cyclohexyl, cyclopentyl, cyclopentenyl, cyclobutyl and cyclopropyl. Exemplary bicyclic cycloalkyl groups include, but are not limited to, spiro[2.5]octanyl, spiro[3.5]nonanyl, bicyclo[2.2.2]octanyl, bicyclo[4.1.0]heptanyl, octahydropentalenyl, bicyclo[4.2.0]octanyl, bicyclo[l.l.l]pentanyl, bicyclo[2.2.1]heptanyl, and bicyclo[2.2.2]octanyl. The term “cycloalkenyl” as used herein refers to a partially unsaturated monocyclic hydrocarbon group of, for example, 4-6 carbons, referred to herein as C4-6monocycloalkenyl, or bicyclic hydrocarbon ring structure of, for example, 8-12 carbons, referred to herein as Cs- i 2 bicycloalkenyl. For bicyclic cycloalkenyl groups: 1) either one or both rings may contain one or more double bonds, and 2) the two rings may be attached through the same or different ring carbons. Exemplary monocyclic cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptenyl. Exemplary bicyclic cycloalkenyl groups include, but are not limited to, spiro[2.5]oct-5-enyl, spiro[2.5]oct-4-enyl, spiro[3.5]non-5-enyl, spiro[3.5]non-6-enyl, bicyclo[4.1.0]hept-3-enyl, bicyclo[4.1.0]hept-2-enyl, and bicyclo[2.2.2]oct-2-enyl.

The term “carbocyclyl” as used herein refers to a bicyclic ring system formed by fusing a phenyl ring to a C3-6monocycloalkyl or C4-6monocycloalkenyl ring. Examples of carbocyclyls include, but are not limited to, 2,3-dihydro- lH-indenyl, 1, 2,3,4- tetrahydronaphthalenyl, and lH-indenyl.

The terms “halo” or “halogen” as used herein refer to F, Cl, Br, or I.

The term “haloalkyl” as used herein refers to an alkyl group substituted with one or more halogen atoms. For example, haloCi- 6 alkyl refers to a straight or branched alkyl group of 1-6 carbon atoms substituted with one or more halogen atoms. Examples include, but are not limited to, CH 2 F-, CHCk-, -CHF 2 , CF3-, CF CH 2 -, CH CF 2 , CF CC1 2 - and CF CF 2 -.

The term “haloalkoxy” as used herein refers to an alkoxy group substituted with one or more halogen atoms. Examples include, but are not limited to, CCI3O-, CF3O-, CHF 2 0- CF 3 CH 2 0-, and CF CF 2 0-.

The terms “heteroaryl” as used herein refers to a 5-6 membered monocyclic or 8-12 membered bicyclic aromatic ring system containing one to four independently selected heteroatoms, such as nitrogen, oxygen and sulfur. Where possible, the heteroaryl ring may be linked to the adjacent radical though carbon or nitrogen. Examples of 5-6 membered monocyclic heteroaryl groups include, but are not limited to, furanyl, thiophenyl (also referred to as thienyl), pyrrolyl, thiazolyl, oxazolyl, isothiazolyl, isoxazolyl, imidazolyl, pyrazolyl, lH-l,2,3-triazolyl, 2H-l,2,3-triazolyl, 1,2,4-triazolyl, pyridinyl (also referred to as pyridyl), pyridazinyl, pyrimidinyl, pyrazinyl, 1,3,5-triazinyl, 1,2,4-triazinyl, 1,2,3-triazinyl,

1.2.4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl,

1.2.5-thiadiazolyl and tetrazolyl. Examples of 8-12 membered bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, isobenzofuranyl, benzol /;|thiophenyl, benzo[c]thiophenyl, indolyl, isoindolyl, benzol c/|isoxazolyl, benzo[c]isoxazolyl, benzol c/|oxazolyl, benzo|c/|isothiazolyl, benzo[c]isothiazolyl, benzo|c/|thiazolyl, indazolyl, benzol c/|imidazolyl, benzo|c/|imidazolyl, and benzol d\\ 1 ,2,3 |triazolyl .

The term “heterocycloalkyl” refers to a saturated 3-6 membered monocyclic or 8-12 membered bicyclic ring system, referred to herein as C3-6monoheterocycloalkyl and Cs- nbiheterocycloalkyl, containing one to four independently selected heteroatoms, such as nitrogen, oxygen, and sulfur (including its oxidation states: S(O) and SO2). Where possible, heterocycloalkyl rings may be linked to the adjacent radical through carbon or nitrogen. Examples of C3-6monoheterocycloalkyl groups include, but are not limited to, aziridinyl, oxiranyl, thiiranyl 1,1 -dioxide, oxetanyl, azetidinyl, thietanyl 1,1 -dioxide, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydro-2H-pyranyl, morpholinyl, thiomorpholinyl, and piperazinyl. Examples of Cs-nbiheterocycloalkyl groups include, but are not limited to, 1,4- dioxaspiro[4.5]decanyl and l,5-dioxaspiro[5.5]undecanyl.

The term “heterocycloalkenyl” refers to a partially unsaturated 3-6 membered monocyclic or 8-12 membered bicyclic ring system, referred to herein as C3- 6 monoheterocycloalkenyl and Cs-nbiheterocycloalkenyl, containing one to four independently selected heteroatoms, such as nitrogen, oxygen, and sulfur (including its oxidation states: S(O) or S(0) 2 . Where possible, heterocycloalkenyl rings may be linked to the adjacent radical through carbon or nitrogen. For bicyclic heterocycloalkenyl groups: 1) either one or both rings may contain one or more double bonds, and 2) the two rings may be attached through the same or different ring atoms. Examples of C3-6monoheterocycloalkenyl groups include, but are not limited to, 2,3-dihydro-lH-pyrrolyl, 2,5-dihydro- lH-pyrrolyl, 4,5- dihydro- lH-pyrazolyl, 2,3-dihydro- lH-pyrazolyl, 4,5-dihydro-lH-imidazolyl, 2,3-dihydro- lH-imidazolyl, 2,3-dihydrothiophenyl, 2,5-dihydrothiophenyl, 4,5-dihydrothiazolyl, 2,3- dihydrothiazolyl, 4,5-dihydroisothiazolyl, 2,3-dihydroisothiazolyl, 2,3-dihydrofuranyl, 2,5- dihydrofuranyl, 4,5-dihydrooxazolyl, 2,3-dihydrooxazolyl, 4,5-dihydroisoxazolyl, 2,3- dihydroisoxazolyl, 3,4-dihydropyridinyl, 2,3-dihydropyridinyl, 2,3,4,5-tetrahydropyridinyl,

1.6-dihydropyridazinyl, 4,5-dihydropyridazinyl, 3,4,5,6-tetrahydropyridazinyl, 4,5- dihydropyrimidinyl, 1,2,5,6-tetrahydropyrimidinyl, 1,2-dihydropyrimidinyl, 1,2- dihydropyrazinyl, 2,3-dihydropyrazinyl, 1,2,3,6-tetrahydropyrazinyl, 4H-l,4-oxazinyl, 3,4- dihydro-2H-l,4-oxazinyl, 4H-l,4-thiazinyl, and 3,4-dihydro-2H-l,4-thiazinyl. Examples of Cs nbiheterocycloalkenyl groups include, but are not limited to, 6,7-dihydroindolyl, 4,5- dihydroindolyl, 7,8-dihydroimidazo[l,2-a]pyridinyl, 5,6-dihydroimidazo[l,2-a]pyridinyl, 4,5- dihydrobenzo[d]imidazolyl, 6,7-dihydro-lH-indazolyl, 4,5-dihydro-lH-indazolyl, 4,5- dihydropyrazolo [ 1 , 5-a]pyridinyl, and 6,7 -dihydropyrazolo [ 1 ,5 -a ] pyridi ny 1.

The term “heterocyclyl” as used herein refers to a bicyclic ring system formed by either (1) fusing a phenyl ring to a 3-6 membered monocyclic heterocycloalkyl or 4-7 membered monocyclic heterocycloalkenyl ring, or (2) fusing a 5-6 membered monocyclic heteroaryl ring to a C3-6 cycloalkyl, C4-7 cycloalkenyl, 3-6 membered monocyclic heterocycloalkyl or 4-6 membered monocyclic heterocycloalkenyl ring. Where possible, the rings may be linked to the adjacent radical though carbon or nitrogen. Examples of heterocyclyls include, but are not limited to isochromanyl, 2H-quinolinyl, 6,7,8,9-tetrahydro- 5H-[l,2,4]triazolo[4,3-a]azepine, 5,6,8,9-tetrahydro-[l,2,4]triazolo[4,3-d][l,4]oxazepane,

6.7-dihydro-5H,9H-[l,2,4]triazolo[3,4-c][l,4]oxazepane, 5,6,8,9-tetrahydro-712- [l,2,4]triazolo[4,3-d][l,4]diazepine, 8,9-dihydro-5H-[l,2,4]triazolo[4,3-a]azepine, 6,9- dihydro-5H-[l,2,4]triazolo[4,3-a]azepine, 5,6,7,8-tetrahydro-[l,2,4]triazolo[4,3-a]pyridine, 5,6-dihydro-8H-[l,2,4]triazolo[3,4-c][l,4]oxazine, 5,6,7,8-tetrahydroimidazo[l,2-a]pyridine, and 5H,9H-[l,2,4]triazolo[3,4-c][l,4]oxazepine.

The terms “hydroxy” and “hydroxyl” as used herein refers to the radical -OH.

The term “hydroxyalkyl” as used herein refers to an alkyl group substituted with one or more hydroxy groups. Examples include, but are not limited to, HOCH2-, HOCH2CH2-, CH 3 CH(OH)CH 2 - and HOCH 2 CH(OH)CH 2 -. The term “hydroxyalkoxy” as used herein refers to an alkoxy group substituted with one or more hydroxy groups. Examples include but are not limited to HOCH2O-, HOCH2CH2O-, CH 3 CH(0H)CH 2 0- and H0CH 2 CH(0H)CH 2 0-.

The term “R a R b NCi- 6 alkyl-,” as used herein refers to an alkyl group substituted with a R a R b N- group, as defined herein. Examples include but are not limited to NH2CH2-, NH(CH 3 )CH 2 -, N(CH 3 ) 2 CH 2 CH2- and CH 3 CH(NH 2 )CH 2 -.

The term “R a R b NCi- 6 alkoxy,” as used herein refers to an alkoxy group substituted with a R a R b N- groups, as defined herein. Examples include but are not limited to NH2CH2-, NH(CH 3 )CH 2 0-, N(CH 3 ) 2 CH 2 CH 2 0- and CH 3 CH(NH 2 )CH 2 0-.

The term “oxo” as used herein refers to the radical =0.

As used herein, when a bicyclic ring is shown with a floating point of attachment and/or floating substituents, for example signifies that the bicyclic ring can be attached via a carbon atom on either ring, and that the substituents (e.g., the R 33 group(s)) can be independently attached to either or both rings.

The terms “Individual,” “patient,” or “subject” are used interchangeably and include any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans. The compounds or pharmaceutical compositions of the disclosure can be administered to a mammal, such as a human, but can also be administered to other mammals such as an animal in need of veterinary treatment, e.g., domestic animals (e.g., dogs, cats, and the like), farm animals (e.g., cows, sheep, pigs, horses, and the like) and laboratory animals (e.g., rats, mice, guinea pigs, dogs, primates, and the like). The mammal treated in the methods of the disclosure is desirably a mammal in which treatment of HBV infection is desired.

The term “modulation” includes antagonism (e.g., inhibition), agonism, partial antagonism and/or partial agonism.

The term “Pharmaceutically acceptable” include molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate. For human administration, preparations should meet sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biologies standards.

The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” as used herein refers to any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, fillers, and the like, that are compatible with pharmaceutical administration· The use of such media and agents for pharmaceutically active substances is well known in the art. The compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.

The term “pharmaceutical composition” as used herein refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable excipients.

The term "pharmaceutically acceptable salt(s)" as used herein refers to salts of acidic or basic groups that may be present in compounds used in the compositions. Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. The acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including, but not limited to, malate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate (i.e.,

1 , 1 , -methylene-/ /.v-(2-hydroxy-3-naphthoate)) salts. Compounds included in the present compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include alkali metal or alkaline earth metal salts, particularly calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts. Compounds included in the present compositions that include a basic or acidic moiety may also form pharmaceutically acceptable salts with various amino acids. The compounds of the disclosure may contain both acidic and basic groups; for example, one amino and one carboxylic acid group. In such a case, the compound can exist as an acid addition salt, a zwitterion, or a base salt.

The term “therapeutically effective amount” or “effective amount” as used herein refers to the amount of the subject compound that will elicit the biological or medical response of a tissue, system or animal, (e.g., mammal or human) that is being sought by the researcher, veterinarian, medical doctor or other clinician. The compounds or pharmaceutical compositions of the disclosure are administered in therapeutically effective amounts to treat a disease. Alternatively, a therapeutically effective amount of a compound is the quantity required to achieve a desired therapeutic and/or prophylactic effect.

The term “treating” includes any effect, e.g., lessening, reducing, modulating, or eliminating, via disruption of HBV core protein assembly, that results in the improvement of the disease. “Disruption” includes inhibition of HBV viral assembly and infection.

The compounds of the disclosure may contain one or more chiral centers and, therefore, exist as stereoisomers. The term “stereoisomers” when used herein consist of all enantiomers or diastereomers. These compounds may be designated by the symbols “(+),” “(- ),” “R” or “S,” depending on the configuration of substituents around the stereogenic carbon atom, but the skilled artisan will recognize that a structure may denote a chiral center implicitly. The present disclosure encompasses various stereoisomers of these compounds and mixtures thereof. Mixtures of enantiomers or diastereomers may be designated “(±)” in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.

The compounds of the disclosure may contain one or more double bonds and, therefore, exist as geometric isomers resulting from the arrangement of substituents around a carbon-carbon double bond. The symbol denotes a bond that may be a single, double or triple bond as described herein. Substituents around a carbon-carbon double bond are designated as being in the “Z’ or “E" configuration wherein the terms “Z’ and “£” are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting double bonds encompass both the “E” and “Z” isomers. Substituents around a carbon-carbon double bond alternatively can be referred to as “cis” or “trans,” where “cis” represents substituents on the same side of the double bond and “trans” represents substituents on opposite sides of the double bond.

Compounds of the disclosure may contain a carbocyclic or heterocyclic ring and therefore, exist as geometric isomers resulting from the arrangement of substituents around the ring. The arrangement of substituents around a carbocyclic or heterocyclic ring are designated as being in the “Z” or “E” configuration wherein the terms “Z” and “E” are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting carbocyclic or heterocyclic rings encompass both “Z” and “E” isomers. Substituents around a carbocyclic or heterocyclic ring may also be referred to as “cis” or “trans”, where the term “cis” represents substituents on the same side of the plane of the ring and the term “trans” represents substituents on opposite sides of the plane of the ring. Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated “cis/trans.”

Individual enantiomers and diastereomers of compounds of the present disclosure can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, (3) direct separation of the mixture of optical enantiomers on chiral liquid chromatographic columns, or (4) kinetic resolution using stereoselective chemical or enzymatic reagents. Racemic mixtures can also be resolved into their component enantiomers by well-known methods, such as chiral-phase liquid chromatography or crystallizing the compound in a chiral solvent. Stereoselective syntheses, a chemical or enzymatic reaction in which a single reactant forms an unequal mixture of stereoisomers during the creation of a new stereocenter or during the transformation of a pre-existing one, are well known in the art. Stereoselective syntheses encompass both enantiomeric and diastereoselective transformations and may involve the use of chiral auxiliaries. For examples, see Carreira and Kvaemo, Classics in Stereoselective Synthesis, Wiley-VCH: Weinheim, 2009.

The compounds disclosed herein can exist in solvated as well as unsolvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the disclosure embrace both solvated and unsolvated forms. In one embodiment, the compound is amorphous. In one embodiment, the compound is a single polymorph. In another embodiment, the compound is a mixture of polymorphs. In another embodiment, the compound is in a crystalline form.

The disclosure also embraces isotopically labeled compounds of the disclosure which are identical to those recited herein, except that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, and 36 C1, respectively. For example, a compound of the disclosure may have one or more H atom replaced with deuterium.

Certain isotopically-labeled disclosed compounds (e.g., those labeled with 3 H and 14 C) are useful in compound and/or substrate tissue distribution assays. Tritiated (/. <? ., 3 H) and carbon-14 (/. <? ., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (/. <? ., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labeled compounds of the disclosure can generally be prepared by following procedures analogous to those disclosed in the examples herein by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

The term “prodrug” refers to compounds that are transformed in vivo to yield a disclosed compound or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms (such as by esterase, amidase, phosphatase, oxidative and or reductive metabolism) in various locations (such as in the intestinal lumen or upon transit of the intestine, blood or liver). Prodrugs are well known in the art (for example, see Rautio, Kumpulainen, et al., Nature Reviews Drug Discovery 2008, 7, 255).

II. 5-Membered Heteroaryl Carboxamide Compounds

In one aspect, the present disclosure provides a compound of Formula I Formula I

, or a pharmaceutically acceptable salt thereof, wherein:

L is Ci-4alkylene or haloCi-4alkylene;

L 1 and L 2 are independently a bond, Ci- 6 alkylene, O, NR C , C(O), C(0)0, C(0)NR c , S(O), or S(0),NR c ;

X 1 is NR xl , O or S;

X 2 is O, NR 13 , CR 13 R 8 , C(O), or S(O),;

X 3 is O, NR 4 , CR 4 R 8 , C(O), or S(O),;

X 4 and X 6 are independently O or S;

X 5 is O, S or NR°;

R a , R b and R c are independently selected for each occurrence from the group consisting of hydrogen, Ci- 6 alkyl, haloCi- 6 alkyl and C3-6 monocycloalkyl;

R d is hydrogen, OH, Ci-6 alkyl or Ci-6 alkoxy;

R xl is hydrogen, C 1-4 alkyl, C 1-4 alkenyl, C 1-4 alkynyl, haloCi- 4 alkyl, or C 3-6 monocycloalkyl; or R xl and R 2 together form a -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, - CH2CH2O-, -CH2OCH2-, -CH2CH2CH2O- -CH2CH2OCH2-, -CH2CH2-NH- -CH2NHCH2-, - CH 2 CH 2 CH 2 NH- or -CH 2 CH 2 NHCH 2 - group;

R 0a is independently selected for each occurrence from the group consisting of hydrogen, halogen, OH, CN, NO2, R a R b N-, Ci-4alkyl and haloCi-4alkyl;

R 4a and R 6a are independently hydrogen or C 1-4 alkyl; R°, R 6 and R 11 are independently selected for each occurrence from the group consisting of hydrogen, halogen, OH, CN, NO2, oxo, R d N=, hydrazino, formyl, azido, silyl, siloxy, HOC(O)-, R a R b N-, R a R b NS(0) t -, Ci- 6 alkyl, C2-6alkenyl, C2-6alkynyl, haloCi- 6 alkyl, hydroxyCi-ealkyl-, R a R b NCi- 6 alkyl-, HOC(0)Ci- 6 alkyl-, R a R b NCi- 6 alkylNR c -, Ci- 6 alkylNR a Ci- 6 alkylNR c -, Ci- 6 alkoxy, haloCi- 6 alkoxy, hydroxyCi- 6 alkoxy-, R a R b NCi- 6 alkoxy-, Ci- 6 alkoxyCi- 6 alkyl-, haloCi- 6 alkoxyCi- 6 alkyl-, R a R b NC(0)-, Ci- 6 alkylC(0)-, Ci- ealkoxyC(O)-, Ci- 6 alkylC(0)0-, Ci- 6 alkylS(0) q -, Ci- 6 alkylS(0),NR c -, Ci- 6 alkylS(0),Ci- 6 alkyl- , Ci- 6 alkylS(0) t NR a Ci- 6 alkyl-, C 3-6 cycloalkylS(0) t Ci- 6 alkyl-, Ci- 6 alkylC(0)Ci- 6 alkyl-, and Ci- 6 alkylC(0)0Ci- 6 alkyl-;

R 1 is a phenyl or 5-6 membered monocyclic heteroaryl, wherein the phenyl or 5-6 membered monocyclic heteroaryl is optionally substituted with one, two, or three independently selected R 11 groups;

R 2 , R 2a and R 8 are independently selected from the group consisting of hydrogen, halo, CN, OH, R a R b N, Ci-4alkyl, haloCi-4alkyl, C3-5monocycloalkyl, Ci-4alkoxy, and haloCi- 4alkoxy; or R 2 and R 2a together with the carbons to which they are attached form a phenyl, pyridyl, pyridazine, pyrimidine or pyrazine ring; or R 2 and R 2a together form a -CH2CH2CH2- , -CH2CH2CH2CH2-, -OCH2CH2-, -CH2OCH2-, -OCH2CH2CH2-, -CH2OCH2CH2-, - NHCH2CH2-, -CH2NHCH2-, -NHCH2CH2CH2- or -CH2NHCH2CH2- group;

R 4 is R 5 -L 1 -, R 6 or R 9 ; or R 4 and R 8 together with the carbon atom to which they are attached form

R 14 is R a R b N-, Ci- 6 alkyl, C2-6alkenyl, C2-6alkynyl, Ci- 6 haloalkyl, Ci- 6 alkoxy, Ci-

6 haloalkyl, Ci- 6 haloalkoxy, or R 5 -L 1 -; q, r, t, and w are independently selected for each occurrence from 0, 1 and 2; and v is independently selected for each occurrence from 0, 1, 2 and 3.

The following embodiments further describe a compound of Formula I, or a pharmaceutically acceptable salt thereof. It will be appreciated that all chemically allowable combinations of the embodiments described herein are envisioned as further embodiments of the invention.

In certain embodiments, X 1 is NR xl . In certain embodiments, X 1 is NR xl and R xl is hydrogen of methyl.

In certain embodiments, X 1 is NR xl and R xl is methyl.

In certain embodiments, X 2 is CR 13 R 8 .

In certain embodiments, X 3 is CR 4 R 8 .

In certain embodiments, L 1 is a bond.

In certain embodiments, L 1 is a Ci- 6 alkylene.

In certain embodiments, r is 0.

^Q-(R 11 ) Z1

In certain embodiments, R 1 is ; R 11 is independently selected for each occurrence from the group consisting of halogen, CN, Ci- 6 alkyl and haloCi- 6 alkyl; and zl is 0, 1, 2 or 3.

In certain embodiments, R 11 is independently selected for each occurrence from the group consisting of halogen and CN.

In certain embodiments, R 11 is independently selected for each occurrence from the group consisting of F, Cl, Br and I.

In certain embodiments, R 1 is selected from the group consisting of:

,

In certain embodiments,

In certain embodiments, X 1 is NR xl , R xl is hydrogen or methyl, and R 1 is

In certain embodiments, R 1 is a 5-6 membered monocyclic heteroaryl optionally substituted with one, two, or three substituents independently selected from the group consisting of halogen, CN, Ci- 6 alkyl, and haloCi- 6 alkyl.

In certain embodiments, R 1 is ; R 11 is independently selected for each occurrence from the group consisting of halogen, CN, Ci- 6 alkyl and haloCi- 6 alkyl; and zl is 0, 1, 2 or 3.

In certain embodiments, R 2 is R a R b N; In certain embodiments, R 2 is R a R b N, and R a and R b are independently selected the group consisting of hydrogen and Ci-6alkyl.

In certain embodiments, R 2 is NH 2 .

In certain embodiments, X 1 is NR xl , R xl is hydrogen or methyl, R 1 is and R 2 is NH 2 .

In certain embodiments,

In certain embodiments,

In certain embodiments,

In certain embodiments,

In certain embodiments, In certain embodiments,

In certain embodiments,

In certain embodiments,

In certain embodiments,

In certain embodiments,

In certain embodiments, In certain embodiments,

In certain embodiments, R 4 is R 5 -L 1

In certain embodiments, R 4 is R 5 .

In certain embodiments, R 4 is R 6 .

In certain embodiments, R 4 is R 9 .

In certain embodiments, or R 4 and R 8 together with the carbon atom to which they are

In certain embodiments, R 6 is hydrogen, Ci- 6 alkylS(0) t Ci- 6 alkyl- or Ci-alkylS(0) t NR a C 1-ealkyl- .

In certain embodiments, R 6 is Ci- 6 alkylS(0) t Ci- 6 alkyl- or Ci- 6 alkylS(0) t NR a Ci- 6 alkyl-.

In certain embodiments, R 6 is hydrogen. In certain embodiments, R 8 is hydrogen, OH or Ci- 6 alkoxy.

In certain embodiments, R 8 is OH.

In certain embodiments, R 6 is hydrogen and R 8 is OH.

In certain embodiments, R 14 is R a R b N-, Ci-6alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci- 6haloalkyl, Ci-6alkoxy, Ci-6haloalkyl, or Ci-6haloalkoxy.

In certain embodiments, R 14 is R 5 -L 1

In certain embodiments, R 14 is R 5 .

In certain embodiments, X 1 is NR xl ; R xl is hydrogen or methyl; R 1 is · In certain embodiments, X 1 is NR xl ; R xl is hydrogen or methyl; R 1 is · · > s hydrogen, Ci- 6 alkylS(0) t Ci- 6 alkyl- or Ci- 6 alkylS(0) t NR a Ci- 6 alkyl-; and R 8 is hydrogen, OH or Ci- 6 alkoxy.

In certain embodiments, X 1 is NR xl ; R xl is hydrogen or methyl; R 1 is s hydrogen; and R 8 is OH.

In certain embodiments, X 1 is NR xl ; R xl is hydrogen or methyl; R 1 is R 8 is hydrogen, OH or Ci- 6 alkoxy. In certain embodiments, X 1 is NR xl ; R xl is hydrogen or methyl; R 1 is

III. Pharmaceutical Compositions and Kits

In another aspect, the disclosure provides pharmaceutical compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In particular, the present disclosure provides pharmaceutical compositions comprising compounds as disclosed herein formulated together with one or more pharmaceutically acceptable carriers. These formulations include those suitable for oral, rectal, topical, buccal, parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous), rectal, vaginal, or aerosol administration, although the most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used. For example, disclosed compositions may be formulated as a unit dose, and/or may be formulated for oral or subcutaneous administration·

In another aspect, the disclosure provides a pharmaceutical composition comprises a compound of Table 1, or a pharmaceutically acceptable salt and/or stereoisomer thereof.

Exemplary pharmaceutical compositions of this disclosure may be used in the form of a pharmaceutical preparation, for example, in solid, semisolid or liquid form, which contains one or more compounds of the disclosure, as an active ingredient, in admixture with an organic or inorganic carrier or excipient suitable for external, enteral or parenteral applications. The active ingredient may be compounded, for example, with the usual non toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, solutions, emulsions, suspensions, and any other form suitable for use. The active object compound is included in the pharmaceutical composition in an amount sufficient to produce the desired effect upon the process or condition of the disease.

For preparing solid compositions such as tablets, the principal active ingredient may be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the disclosure, or a non toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.

In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the subject composition is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.

A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the subject composition moistened with an inert liquid diluent.

Tablets, and other solid dosage forms, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art.

Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the subject composition, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, cyclodextrins and mixtures thereof.

Suspensions, in addition to the subject composition, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing a subject composition with one or more suitable non irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the body cavity and release the active agent.

Dosage forms for transdermal administration of a subject composition include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants· The active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.

The ointments, pastes, creams and gels may contain, in addition to a subject composition, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays may contain, in addition to a subject composition, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays may additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

Compositions and compounds of the present disclosure may alternatively be administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A non-aqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers may be used because they minimize exposing the agent to shear, which may result in degradation of the compounds contained in the subject compositions. Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of a subject composition together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular subject composition, but typically include non-ionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.

Pharmaceutical compositions of this disclosure suitable for parenteral administration comprise a subject composition in combination with one or more pharmaceutically- acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

Examples of suitable aqueous and non-aqueous carriers which may be employed in the pharmaceutical compositions of the disclosure include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate and cyclodextrins. Proper fluidity may be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

In another aspect, the disclosure provides enteral pharmaceutical formulations including a disclosed compound and an enteric material; and a pharmaceutically acceptable carrier or excipient thereof. Enteric materials refer to polymers that are substantially insoluble in the acidic environment of the stomach, and that are predominantly soluble in intestinal fluids at specific pHs. The small intestine is the part of the gastrointestinal tract (gut) between the stomach and the large intestine, and includes the duodenum, jejunum, and ileum. The pH of the duodenum is about 5.5, the pH of the jejunum is about 6.5 and the pH of the distal ileum is about 7.5. Accordingly, enteric materials are not soluble, for example, until a pH of about 5.0, of about 5.2, of about 5.4, of about 5.6, of about 5.8, of about 6.0, of about 6.2, of about 6.4, of about 6.6, of about 6.8, of about 7.0, of about 7.2, of about 7.4, of about 7.6, of about 7.8, of about 8.0, of about 8.2, of about 8.4, of about 8.6, of about 8.8, of about 9.0, of about 9.2, of about 9.4, of about 9.6, of about 9.8, or of about 10.0. Exemplary enteric materials include cellulose acetate phthalate (CAP), hydroxypropyl methylcellulose phthalate (HPMCP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose acetate succinate (HPMCAS), cellulose acetate trimellitate, hydroxypropyl methylcellulose succinate, cellulose acetate succinate, cellulose acetate hexahydrophthalate, cellulose propionate phthalate, cellulose acetate maleate, cellulose acetate butyrate, cellulose acetate propionate, copolymer of methylmethacrylic acid and methyl methacrylate, copolymer of methyl acrylate, methylmethacrylate and methacrylic acid, copolymer of methylvinyl ether and maleic anhydride (Gantrez ES series), ethyl methyacrylate-methylmethacrylate- chlorotrimethylammonium ethyl acrylate copolymer, natural resins such as zein, shellac and copal collophorium, and several commercially available enteric dispersion systems (e. g. , Eudragit L30D55, Eudragit FS30D, Eudragit L100, Eudragit S100, Kollicoat EMM30D, Estacryl 30D, Coateric, and Aquateric). The solubility of each of the above materials is either known or is readily determinable in vitro. The foregoing is a list of possible materials, but one of skill in the art with the benefit of the disclosure would recognize that it is not comprehensive and that there are other enteric materials that would meet the objectives of the present disclosure.

Advantageously, the disclosure also provides kits for use by a e.g. a consumer in need of HBV infection treatment. Such kits include a suitable dosage form such as those described above and instructions describing the method of using such dosage form tomediate, reduce or prevent HBV infection. The instructions would direct the consumer or medical personnel to administer the dosage form according to administration modes known to those skilled in the art. Such kits could advantageously be packaged and sold in single or multiple kit units. An example of such a kit is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of the tablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are sealed in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.

It may be desirable to provide a memory aid on the kit, e.g., in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested. Another example of such a memory aid is a calendar printed on the card, e.g., as follows “First Week, Monday, Tuesday, . . . etc. . . . Second Week, Monday, Tuesday, . . . ” etc. Other variations of memory aids will be readily apparent. A “daily dose” can be a single tablet or capsule or several pills or capsules to be taken on a given day. Also, a daily dose of a first compound can consist of one tablet or capsule while a daily dose of the second compound can consist of several tablets or capsules and vice versa. The memory aid should reflect this.

IV. Methods

In a further aspect, a method for treating a hepatitis B infection in a patient in need thereof is provided, comprising administering to a subject or patient an effective amount of a disclosed compound, and/or administering a first disclosed compound and optionally, an additional, different disclosed compound(s). In another embodiment, a method for treating a hepatitis B infection in a patient in need thereof is provided, comprising administering to a subject or patient a therapeutically effective amount of a disclosed pharmaceutical composition or a pharmaceutical composition comprising a disclosed compound, or two or more disclosed compounds, and a pharmaceutically acceptable excipient.

For use in accordance with this aspect, the appropriate dosage is expected to vary depending on, for example, a particular compound employed, the mode of administration, and the nature and severity of the infection to be treated as well as the specific infection to be treated and is within the purview of the treating physician. Usually, an indicated administration dose may be in the range between about 0.1 to about 1000 pg/kg body weight. In some cases, the administration dose of the compound may be less than 400 pg/kg body weight. In other cases, the administration dose may be less than 200 pg/kg body weight. In yet other cases, the administration dose may be in the range between about 0.1 to about 100 pg/kg body weight. The dose may be conveniently administered once daily, or in divided doses up to, for example, four times a day or in sustained release form.

A compound of the present disclosure may be administered by any conventional route, in particular: enterally, topically, orally, nasally, e.g., in the form of tablets or capsules, via suppositories, or parenterally, e.g., in the form of injectable solutions or suspensions, for intravenous, intra-muscular, sub-cutaneous, or intra-peritoneal injection. Suitable formulations and pharmaceutical compositions will include those formulated in a conventional manner using one or more physiologically acceptable carriers or excipients, and any of those known and commercially available and currently employed in the clinical setting. Thus, the compounds may be formulated for oral, buccal, topical, parenteral, rectal or transdermal administration or in a form suitable for administration by inhalation or insufflation (either orally or nasally).

For oral administration, pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate). Tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). Preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.

Preparations for oral administration may also be suitably formulated to give controlled-release or sustained release of the active compound(s) over an extended period.

For buccal administration the compositions may take the form of tablets or lozenges formulated in a conventional manner known to the skilled artisan. A disclosed compound may also be formulated for parenteral administration by injection e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain additives such as suspending, stabilizing and/or dispersing agents. Alternatively, the compound may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen- free water, before use. Compounds may also be formulated for rectal administration as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.

Also contemplated herein are methods and compositions that include a second active agent, or administering a second active agent. For example, in addition to being infected with HBV, a subject or patient can further have HBV infection-related co-morbidities, i.e., diseases and other adverse health conditions associated with, exacerbated by, or precipitated by being infected with HBV. Contemplated herein are disclosed compounds in combination with at least one other agent that has previously been shown to treat these HBV-infection- related conditions.

In some cases, a disclosed compound may be administered as part of a combination therapy in conjunction with one or more antivirals. Example antivirals include nucleoside analogs, interferon a, and other assembly effectors, for instance heteroaryldihydropyrimidines (HAPs) such as methyl 4-(2-chloro-4-fluorophenyl)-6-methyl- 2-(pyridin-2-yl)-l,4-dihydropyrimidine-5-carboxylate (HAP-1). For example, provided herein is a method of treating a patient suffering from hepatitis B infection comprising administering to the patient a first amount of a disclosed compound and a second amount of an antiviral, or other anti HBV agent, for example a second amount of a second compound selected from the group consisting of: a HBV capsid assembly promoter, for example, GLS4, BAY 41-4109, AT-130, DVR-23 (e.g., as depicted below), NVR 3-778, NVR1221 (by code); and N890 (as depicted below): other capsid inhibitors such as those disclosed in the following patent applications hereby incorporated by reference: W02014037480, WO2014184328, W02013006394, WO2014089296, W02014106019, WO2013102655, WO2014184350, WO2014184365, WO2014161888, WO2014131847, WO2014033176, WO2014033167, and W02014033170; Nucleos(t)ide analogs interfering with viral polymerase, such as entecavir (Baraclude), Lamivudine, (Epivir-HBV), Telbivudine (Tyzeka, Sebivo), Adefovir dipivoxil (Hepsera), Tenofovir (Viread), Tenofovir alafenamide fumarate (TAF), prodrugs of tenofavir (e.g. AGX-1009), L-FMAU (Clevudine), FB80380 (Besifovir) and: viral entry inhibitors such as Myrcludex B and related lipopeptide derivatives; HBsAg secretion inhibitors such as REP 9AC’ and related nucleic acid-based amphipathic polymers, HBF-0529 (PBHBV-001), PBHBV-2-15 as depicted below:

22: H8F-0529 23: PBHBV-2-15 and BM601 as depicted below: disruptors of nucleocapsid formation or integrity such as NZ-4/W28F: cccDNA formation inhibitors such as BSBI-25, CCC-0346, CCC-0975 (as depicted below):

J

HBc directed transbodies such as those described in Wang Y, et al, Transbody against hepatitis B vims core protein inhibits hepatitis B vims replication in vitro, Int. Immunopharmacol (2014), located at//dx.doi.org/10.1016/j.intimp.2015.01.028; antiviral core protein mutant (such as Cpl83-V124W and related mutations as described in WO/2013/010069, W02014/074906, each incorporated by reference); inhibitors of HBx- interactions such as RNAi, antisense and nucleic acid based polymers targeting HBV RNA;, e.g., RNAi (for example ALN-HBV, ARC-520, TKM-HBV, ddRNAi), antisense (ISIS- HBV), or nucleic acid based polymer(REP 2139-Ca); immunostimulants such as Interferon alpha 2a (Roferon), Intron A (interferon alpha 2b), Pegasys (peginterferon alpha 2a), Pegylated IFN 2b, IFN lambda la and PEG IFN lambda la, Wellferon, Roferon, Infergen, lymphotoxin beta agonists such as CBE11 and BS1); Non- Interferon Immune enhancers such as Thymosin alpha- 1 (Zadaxin) and Interleukin-7 (CYT107); TLR-7/9 agonists such as GS- 9620, CYT003, Resiquimod; Cyclophilin inhibitors such as NVP018; OCB-030; SCY-635; Alisporivir; NIM811 and related cyclosporine analogs; vaccines such as GS-4774, TG1050, Core antigen vaccine; SMAC mimetics such as birinapant and other IAP- antagonists; Epigenetic modulators such as KMT inhibitors (EZH1/2, G9a, SETD7, Suv39 inhibitors), PRMT inhibitors, HDAC inhibitors, SIRT agonists, HAT inhibitors, WD antagonists (e.g. OICR-9429), PARP inhibitors, APE inhibitors, DNMT inhibitors, LSD1 inhibitors, JMJD HDM inhibitors, and Bromodomain antagonists; kinase inhibitors such as TKB1 antagonists, PLK1 inhibitors, SRPK inhibitors, CDK2 inhibitors, ATM & ATR kinase inhibitors; STING Agonists; Ribavirin; N-acetyl cysteine ; NOV-205 (BAM205); Nitazoxanide (Alinia), Tizoxanide; SB 9200 Small Molecule Nucleic Acid Hybrid (SMNH); DV-601; Arbidol; FXR agonists (such as GW 4064 and Fexaramin); antibodies, therapeutic proteins, gene therapy, and biologies directed against viral components or interacting host proteins.

In some embodiments, the disclosure provides a method of treating a hepatitis B infection in a patient in need thereof, comprising administering a first compound selected from any one of the disclosed compounds, and one or more other HBV agents each selected from the group consisting of HBV capsid assembly promoters, HBF viral polymerase interfering nucleosides, viral entry inhibitors, HBsAg secretion inhibitors, disruptors of nucleocapsid formation, cccDNA formation inhibitors, antiviral core protein mutant, HBc directed transbodies, RNAi targeting HBV RNA, immunostimulants, TFR-7/9 agonists, cyclophilin inhibitors, HBV vaccines, SMAC mimetics, epigenetic modulators, kinase inhibitors, and STING agonists. In some embodiments, the disclosure provides a method of treating a hepatitis B infection in a patient in need thereof, comprising administering an amount of a disclosed compound, and administering another HBV capsid assembly promoter.

In some embodiments, the first and second amounts together comprise a pharmaceutically effective amount. The first amount, the second amount, or both may be the same, more, or less than effective amounts of each compound administered as monotherapies. Therapeutically effective amounts of a disclosed compound and antiviral may be co administered to the subject, i.e., administered to the subject simultaneously or separately, in any given order and by the same or different routes of administration. In some instances, it may be advantageous to initiate administration of a disclosed compound first, for example one or more days or weeks prior to initiation of administration of the antiviral. Moreover, additional drugs may be given in conjunction with the above combination therapy.

In another embodiment, a disclosed compound may be conjugated (e.g., covalently bound directly or through molecular linker to a free carbon, nitrogen (e.g., an amino group), or oxygen (e.g., an active ester) of a disclosed compound), with a detection moiety, for e.g., a fluorophore moiety (such a moiety may for example re-emit a certain light frequency upon binding to a vims and/or upon photon excitation). Contemplated fluorophores include AlexaFluor ® 488 (Invitrogen) and BODIPY FL (Invitrogen), as well as fluorescein, rhodamine, cyanine, indocarbocyanine, anthraquinones, fluorescent proteins, aminocoumarin, methoxycoumarin, hydroxycoumarin, Cy2, Cy3, and the like. Such disclosed compounds conjugated to a detection moiety may be used in e.g., a method for detecting HBV or biological pathways of HBV infection, e.g., in vitro or in vivo ; and/or methods of assessing new compounds for biological activity.

V. Examples

The compounds described herein can be prepared in a number of ways based on the teachings contained herein and synthetic procedures known in the art. In the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, can be chosen to be the conditions standard for that reaction, unless otherwise indicated. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated. The starting materials for the examples are either commercially available or are readily prepared by standard methods from known materials.

At least some of the compounds identified as “intermediates” herein are contemplated as compounds of the disclosure.

Abbreviations:

DCM Dichloromethane EtOAc Ethyl acetate MeOH Methanol DMSO Dimethyl sulfoxide

ACN Acetonitrile

DIAD Diisopropyl azodicarboxylate

DIEA Diisopropyl ethylamine nBuLi n-Butyllithium iPrOH Isopropanol

AcOH Acetic acid

B0C2O Di-tert-butyl dicarbonate

Et3N Triethylamine

DMF N,N-Dimethylformamide

THF Tetrahydrofuran

TEA Triethylamine

TFA Trifluoroacetic acid rt Room temperature h, hr Hour(s)

TLC Thin-layer chromatography

LC-MS Liquid chromatography-mass spectrometry

HPLC High performance liquid chromatography

XPhos 2-Dicyclohexylphosphino-2’ ,4’ ,6’ -triisopropylbiphenyl

DPPF 1 , G -Bis(diphenylphosphino)ferrocene

NMO N-Methylmorpholine-N-Oxide

HATU Hexafluorophosphate Azabenzotriazole Tetramethyl Uronium) SFC Supercritical Fluid Chromatography

NBS N-Bromosuccinimide Scheme I

General procedure for amidation:

Method A (amide coupling using EDC HC1): To a stirred solution of carboxylic acid (1 eq.) in 1,4-dioxane (5.84 mL/mmol) were added EDC.HC1 (1.1 eq.), HOBt (1.1 eq.) and corresponding amine (1 eq.) at 0 °C and stirred for 5 min. To this solution, DIPEA (3 eq.) was added and the reaction resulting reaction mixture was stirred at 90 °C for overnight. After completion, the reaction mixture was diluted with ice water and extracted with ethyl acetate. The organic layer was washed with sat. NaHCCF solution, water, dried over sodium sulfate, filtered and concentrated in vacuo to afford crude compound which was purified by silica gel column chromatography/prep. HPLC to afford the desired compound.

Method B (amide coupling using HATU): To a stirred solution of acid compound (1.1 -1.2 eq.) in DMF/DCM (1.01 mL/mmol) at 0 °C, DIPEA (2-3 eq.) and HATU (1.5-2.5 eq.) were added and stirred for 5 min. To this solution, corresponding amine (1 eq.) was added. The resulting reaction mixture was stirred at room temperature for 12-16 h. After completion, the reaction mixture was diluted with ice cold water and extracted with ethyl acetate. The organic layer was collected; washed with brine; dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford a crude compound. The crude compound was purified by either prep-HPLC or CombiFlash column chromatography to afford the desired compound.

Method C (AlMe3 mediated amidation): To a stirred solution of corresponding anilines (1.1 eq.) in DCM/Toluene (3 mL/mmol) at 0 °C under Argon atmosphere, A1Mb 3 (2M in toluene, 2.5 eq.) was added and the reaction mixture was stirred at 0 °C for 10 min and continued stirring at room temperature for 1 h. To this solution, corresponding ester compound (1 eq.) was added at 0 °C under Argon atmosphere and resulting reaction mixture was refluxed at 100 °C for 16 h. After completion, the reaction mixture was cooled to 0 °C; quenched with IN HC1 solution slowly and extracted with ethyl acetate. The combined organic layers were collected, dried over anhydrous sodium sulphate and concentrated in vacuo. The crude compound was purified by washing with methanol to afford the desired compound.

Method D (amide coupling using acid chloride/derivatives): To a stirred solution of amine compound (1 eq.) in DCM (1.01 mL/mmol) was added TEA (1.5-3 eq.) at 0 °C and stirred for 5 min. To this solution, corresponding acid chloride/carbamic chloride/chloroformate (1.1-1.5 eq.) was added slowly at 0 °C and the reaction mixture was allowed to stir at room temperature till completion. After completion, the reaction mixture was diluted with ice cold water and extracted with ethyl acetate/DCM. The organic layer was collected; washed with brine; dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford a crude compound. The crude compound was purified by either prep-HPLC or CombiFlash column chromatography to afford the desired compound.

General procedure for Grignard Reaction:

Method A (at lower temperature): To a stirred solution of keto compound (1 eq.) in dry THF (0.2 mL/mmol) in an inert atmosphere was added Grignard reagent (10 eq.) slowly via glass syringe at -78 °C and stirred the reaction mixture for 4 h at same temperature & then at room temperature for 2 h. After completion, the reaction mixture was diluted with sat. aq. solution of ammonium chloride and extracted with ethyl acetate/DCM. The organic layer was collected; washed with brine; dried over anhydrous sodium sulphate and concentrated on rota vapor to afford a crude compound. The crude compound was purified by either by CombiFlash column chromatography or prep-HPLC to afford the desired compound.

General method for Suzuki coupling:

Method A: To a mixture of halo compound (1 eq.) and corresponding boronic acid/boronate ester (1.2-1.5 eq.) in 1, 4-dioxane:water (4:1) (2.17 mL/mmol), Na 2 C0 3 (2-3 eq.) was added and purged with Argon for 15 min. To this solution, PdCh (dppf) (0.1 eq.) was added and purged with Argon for another 10 min. The resulting reaction mixture was stirred at 100 °C for 12-16 h. After completion of the reaction, the reaction mixture was filtered through Celite ® and evaporated to dryness. The residue was taken in ethyl acetate, washed with water, followed by brine, dried over anhydrous sodium sulphate and evaporated under reduced pressure. The crude product was purified by either CombiFlash column chromatography or prep-HPLC to afford the desired compound. General procedure for hydrogenation:

Method A: To a stirred solution of olefinic compound (1 eq.) in EtOAc (2.67 mL/mmol) under nitrogen atmosphere, 20% Pd/C (20% by w/w of olefinic compound) was added. The reaction mixture was stirred under hydrogen atmosphere (100 psi) at 40-50 °C for 4-7 h.

After completion, the reaction mixture was filtered through a pad of Celite and washed with EtOAc/methanol. The filtrate was concentrated under reduced pressure to compound which was purified by silica gel column chromatography or prep-HPLC to give the desired compound.

General procedure for Keto-reduction:

Method A: To a stirred solution of keto compound (1 eq.) in EtOH/MeOH (5 vol) (4.7 mL/mmol), at 0 °C under Argon atmosphere, NaBPE (1-2 eq.) was added and stirred at room temperature for 2-6 h. After completion, the reaction mixture was concentrated in vacuo, the residue obtained was diluted with water and extracted using ethyl acetate. The combined organic layers were collected, dried over anhydrous sodium sulphate, filtered, concentrated in vacuo and purified by silica gel column chromatography/prep. HPLC to give the desired compound. Note: THF was also (1 vol) added as a co-solvent for substrates which are having poor solubility in alcoholic solvents.

Intermediate 1

Methyl 3-bromo-l-methyl-lH-pyrrole-2-carboxylate. NaH (0.48 g, 11.7 mmol) was added to a stirred solution of methyl 3-bromo-lH-pyrrole-2-carboxylate (2.0 g, 9.8 mmol) in DMF (20 mL) at 0 °C and stirring continued at the same temperature for 1 h. Then CH3I (2.8 g, 19.6 mmol) was added and the mixture stirred at rt for 16 h. After completion, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulphate, filtered and concentrated under reduced pressure to afford methyl 3-bromo- 1-methyl- lH-pyrrole-2-carboxylate as a yellowish liquid. TLC: 30% EtOAc/hexanes (R/. 0.7); MS (m/z) Calcd. for CLHxBrNCF: 217.0, Found: 218.1 [M+l] + .

Intermediate 2

3-Bromo-N-(3-chloro-4-fluorophenyl)-l-methyl-lH-pyrrole-2 -carboxamide. To a stirred solution of methyl 3-bromo- 1-methyl- lH-pyrrole-2-carboxylate (1.7 g, 7.8 mmol) and

4-fluoro-3-chloro aniline (1.34 g, 9.4 mmol) in THF (17 mL) at 0°C, LiHMDS (1M in THF, 39 mL, 39 mmol) was added and stirred at rt for 2h. After completion, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layer was dried over anhydrous sodium sulphate, filtered and concentrated under reduced pressure. The crude compound was triturated with ethyl acetate to afford 3-bromo-N-(3-chloro-4- fluorophenyl)- 1 -methyl- lH-pyrrole-2-carboxamide as an off-white solid. TLC: 10% EtOAc / hexanes (v/v) (R/. 0.5); MS (m/z) Calcd. for Ci 2 H 9 BrClFN 2 0: 330.0, Found: 331.2 [M + 1] + .

Intermediate 3

5-Oxo-l,3a,4,5,6,6a-hexahydropentalen-2-yl trifluoromethanesulfonate. To a solution of l,3,3a,4,6,6a-hexahydropentalene-2,5-dione (40.0g, 289.5 mmol) and pyridine (24.0 g, 304.0 mmol) in DCM (600 ml) was added Tf 2 0 (89.8 g, 318.5 mmol) dropwise at room temperature. The mixture was stirred at rt for 3 h. Brine (300 mL) was added and the aqueous layer extracted with DCM (200 mL x 3). The organic layer was separated, dried over Na 2 SC>4 and concentrated to give the crude product which was purified by silica gel column chromatography using 8:1 (v/v) petroleum ether/ethyl acetate to afford 5-oxo-l, 3a, 4, 5, 6, ha- hexahydropentalen-2-yl trifluoromethanesulfonate as a yellow oil. 1 H NMR (400 MHz, CDCb): d 5.63 (q, J= 1.92 Hz, 1 H ), 3.50 - 3.57 (m, 1 H), 3.00 -3.14 (m, 2 H), 2.58 - 2.67

(m, 1 H), 2.40 - 2.56 (m, 2 H), 2.26 - 2.34 (m, 1 H), 2.17 (ddd, 7=19.14, 7.34, 1.63 Hz, 1 H) ppm.

Intermediate 4

5-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)-3,3a,6,6a -tetrahydropentalen- 2(lH)-one. A mixture of 5-oxo-l,3a,4,5,6,6a-hexahydropentalen-2-yl trifluoromethanesulfonate (110.0 g, 407.0 mmol), 4,4,5,5-tetramethyl-2-(4, 4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-l,3,2-dioxaborolane (108.5 g, 427.4 mmol), Pd(dppf)Cl2 (8.9 g,

12.2 mmol) and potassium acetate (119.7 g, 1221.0 mmol) in dioxane (1000 mL) was stirred at 80 °C under an N2 atmosphere for 2 h. The reaction mixture was filtered through a pad of Celite ® 545 and the filter cake was washed with EtOAc (250 mL x 3). The filtrate was concentrated under vacuo and the residue was purified by silica gel column chromatography using 8:1 ( v/v ) petroleum ether/ethyl acetate to afford 5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-3,3a,6,6a-tetrahydropentalen-2(lH)-one as a yellow oil. 1 H NMR (400 MHz, CDCb): d 6.37 (q, J = 2.08 Hz, 1 H), 3.41 - 3.54 (m, 1 H), 2.93 -3.05 (m, 1 H), 2.79 (ddt, J = 16.48, 7.58, 2.64, 2.64 Hz, 1 H), 2.24 - 2.55 (m, 4 H), 1.95 - 2.07 (m, 1 H), 1.28 (s,

13 H) ppm.

Intermediate 5

N-(3-Chloro-4-fluorophenyl)-l-methyl-3-(5-oxo-l,3a,4,5,6, 6a- hexahydropentalen-2-yl)-lH-pyrrole-2-carboxamide. Na2C03 (1.45 g, 13.6 mmol) was added to a stirred mixture of 3-bromo-N-(3-chloro-4-fluorophenyl)- 1-methyl- lH-pyrrole-2- carboxamide (1.5 g, 4.5 mmol), 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,3a,4,6a- tetrahydropentalen-2(lH)-one (1.3 g, 5.40 mmol) in 1, 4-dioxane:water (4:1 ( v/v ), 15 mL) which was then purged with Argon for 15 min. To this was added Pd(dppf)Cl2 (0.033 g, 0.45 mmol) and the mixture purged with Argon for another 10 min. The resulting reaction was stirred at 100 °C for 16 h. After completion, the mixture was filtered through Celite ® 545 and evaporated to dryness. The residue was taken up in ethyl acetate, washed with water, followed by brine, dried over anhydrous sodium sulphate and evaporated under reduced pressure. The crude product was purified by column chromatography to afford compound N- (3 -chloro-4-fluorophenyl)- 1 -methyl-3 -(5 -oxo- 1 ,3 a, 4, 5 ,6, 6a-hexahydropentalen-2-yl)- 1H- pyrrole-2-carboxamide as a white solid. TLC: 20% EtOAc/hexanes (v/v) (Rf. 0.3); MS (m/z): Calcd. for C20H18CIFN2O2: 372.1, Found: 373.3 [M + 1] + .

Example 1

N-(3-Chloro-4-fhiorophenyl)-l-methyl-3-(5-oxooctahydropen talen-2-yl)-lH- pyrrole-2-carboxamide. In an autoclave, 20% Pd/C (30mg, 15% by w/w) was added to a stirred solution of N-(3-chloro-4-fluorophenyl)-l-methyl-3-(5-oxo-l,3a,4,5,6,6a- hexahydropentalen-2-yl)-lH-pyrrole-2-carboxamide (0.2 g, 0.5 mmol) in EtOAc (2 mL) under a nitrogen atmosphere. The reaction mixture was stirred at 50 °C under a hydrogen atmosphere (100 psi) for 16 h. After completion, the mixture was filtered through a pad of Celite ® 545 and washed with methanol. The filtrate was concentrated under reduced pressure and the crude compound purified by silica gel column chromatography to afford N-(3-chloro- 4-fluorophenyl)-l-methyl-3-(5-oxooctahydropentalen-2-yl)-lH- pyrrole-2-carboxamide as an off white solid. TLC: 20% EtOAc/hexanes (v/v) (Rf. 0.5). MS (m/z): Calcd for C20H20CIFN2O2 : 374.12, Found: 374.93 [M + 1] + . l H NMR (400 MHz, DMSO-ifc): d 10.12 (s, 1H), 7.97 (dd, 7= 6.4, 2.0 Hz, 1H), 7.60 - 7.57 (m, 1H), 7.38 (t, 7 = 9.2 Hz, 1H), 6.82 (s, 1H), 6.04 (d, 7= 2.0 Hz, 1H), 3.48 (s, 3H), 3.29 - 3.27 (m, 1H), 2.67 - 2.65 (m, 2H), 2.42 - 2.40 (m, 2H), 2.29 - 2.23 (m, 2H), 207 - 1.99 (m, 2H), 1.37 - 1.28 (m, 2H) ppm.

Example 19

N-(3-Chloro-4-fluorophenyl)-3-(5-hydroxyoctahydropentalen -2-yl)-l-methyl-lH- pyrrole-2-carboxamide. NaBSE (18 mg, 0.48 mmol) was added to a stirred solution of N-(3- chloro-4-fluorophenyl)-l-methyl-3-(5-oxooctahydropentalen-2- yl)-lH-pyrrole-2- carboxamide (0.15 g, 0.4 mmol) in MeOH (1.5 mL) at 0 °C and stirring continued at rt for 1 h. After completion, the reaction mixture was concentrated in vacuo and the residue diluted with water and extracted using ethyl acetate. The combined organic layer was collected, dried over anhydrous sodium sulphate, filtered, concentrated under reduced pressure. The crude compound was triturated with CH3CN to afford N-(3-chloro-4-fluorophenyl)-3-(5- hydroxyoctahydropentalen-2-yl)-l -methyl- lH-pyrrole-2-carboxamide as an off-white solid. MS ( m/z ): Calcd. for C20H22CIFN2O2 : 376.1, Found; 377.0 [M + 1] + ; Ή NMR (400 MHz, DMSO- e): d 10.07 (s, 1H), 7.97 (dd, 7 = 7.0, 2.4 Hz, 1H), 7.60-7.52 (m, 1H), 7.38 (t, 7 = 9.6 Hz, 1H), 6.81 (d, 7 = 2.8 Hz, 1H), 5.98 (d, 7 = 2.8 Hz, 1H), 4.47 (s, 1H), 4.06 (t, 7 = 6.0 Hz, 1H), 3.64 (s, 3H), 3.28 - 3.10 (m, 1H), 2.38 - 2.28 (m, 2H), 2.20 - 2.06 (m, 2H), 1.92 - 1.80 (m, 2H), 1.54 - 1.25 (m, 4H) ppm.

Ethyl 3-bromo-l-methyl-lH-indole-2-carboxylate. To a solution of ethyl 3-bromo- lH-indole-2-carboxylate (1.0 g, 3.73 mmol) in dry DMF (20 mL) was added NaH (60%, 1.0 eq, 3.73 mmol) and the mixture stirred for 20 min at rt. Mel (1.0 eq, 3.73 mmol) was then added and the reaction was stirred for 1 h at room temperature. The mixture was added to an aqueous LiCl solution (40 mL) and extracted with EtOAc (20 mL x 2). The combined organic layers were dried over anhydrous Na2SC>4 and concentrated in vacuo to provide ethyl 3- bromo-1 -methyl- lH-indole-2-carboxylate was a light brown oil which was used to the next step without further purification. MS (m/z): Calcd.: 281.0, Found: 282.0 [M + 1] + . Intermediate 7

3-Bromo-N-(3-chloro-4-fluorophenyl)-l-methyl-lH-indole-2- carboxamide. To a solution of ethyl 3 -bromo-1 -methyl- lH-indole-2-carboxylate (550 mg, 1.95 mmol) in dry toluene (10 mL) was added AlMe 3 (2.0 M, 4.0 eq, 7.80 mmol) and the mixture stirred for 0.5 h at rt. 3-Chloro-4-fluoroaniline (2.0 eq, 3.90 mmol) was then added and stirring continued overnight at 90 °C under an Ar atmosphere. The mixture was quenched with MeOH and filtered. To the filtrate was added water (20 mL) and the mixture extracted with EtOAc (15 mL x 2). The combined organic layers were dried over anhydrous Na2SC>4 and concentrated in vacuo. The residue was purified by recrystallization to afford 3-bromo-N-(3-chloro-4- fluorophenyl)-! -methyl- lH-indole-2-carboxamide as an off-white solid. MS (m/z)· Calcd.: 379.9, Found: 381.0 [M + 1] + .

Intermediate 8

N-(3-Chloro-4-fluorophenyl)-l-methyl-3-(5-oxo-l,3a,4,5,6, 6a- hexahydropentalen-2-yl)-lH-indole-2-carboxamide. 5-(4,4,5,5-Tetramethyl-l,3,2- dioxaborolan-2-yl)-3,3a,6,6a-tetrahydropentalen-2(lH)-one (1.5 eq, 1.76 mmol), Na2CC>3 (2.5 eq, 2.94 mmol) and Pd(dppf)Cl2 (0.1 eq, 0.12 mmol) were added to a solution of 3-bromo-N- (3-chloro-4-fluorophenyl)-l-methyl-lH-indole-2-carboxamide (447 mg, 1.18 mmol) in a mixture solution of dioxane/water (5/1 ( v/v ), 12 mL). The reaction mixture was stirred 3 h at 80 °C under an Ar atmosphere. The mixture was filtered and to the filtrate was added EtOAc (30 mE) which was washed with water (20 mL x 2). The organic layer was dried over anhydrous Na2SC>4 and concentrated in vacuo. The residue was purified by prep-TLC to afford N-(3-chloro-4-fluorophenyl)-l-methyl-3-(5-oxo-l, 3a, 4,5,6, 6a-hexahydropentalen-2- yl)-lH-indole-2-carboxamide as a light- yellow oil. MS (m/z)· Calcd.: 422.1, Found: 423.2 [M

+ ir.

Example 43

N-(3-Chloro-4-fluorophenyl)-l-methyl-3-(5-oxooctahydropen talen-2-yl)-lH- indole-2-carboxamide. To a solution of N-(3-chloro-4-fluorophenyl)-l-methyl-3-(5-oxo- l,3a,4,5,6,6a-hexahydropentalen-2-yl)-lH-indole-2-carboxamid e (349 mg, 0.83 mmol) in THF (30 mL) was added MeOH (3 drops) and Pd/C (60 mg). The reaction mixture was stirred for 2 h at rt under an ¾ atmosphere (balloon). The mixture was filtered, and the filtrate was concentrated in vacuo. The residue was purified by reverse phase column chromatography to afford N-(3-chloro-4-fluorophenyl)-l-methyl-3-(5-oxooctahydropental en- 2-yl)-lH-indole-2-carboxamide as a white solid. MS (m/z): Calcd.: 424.1, Found: 425.1 [M + 1] + . l H NMR (400 MHz, CDCF): d 7.86 (d, J = 3.6 Hz, 1H), 7.67 (d, J = 8.0 Hz, 1H), 7.54 (s, 1H), 7.46 - 7.38 (m, 1H), 7.38 - 7.30 (m, 2H), 7.20 - 7.10 (m, 2H), 3.81 (s, 3H), 3.68 (m, 1H), 2.96 - 2.82 (m, 2H), 2.64 (dd, J = 19.2, 9.6 Hz, 2H), 2.44 - 2.32 (m, 2H), 2.27 (dd, J = 19.2, 2.8 Hz, 2H), 2.12 - 2.00 (m, 2H) ppm. Example 44

N-(3-Chloro-4-fluorophenyl)-3-(5-hydroxyoctahydropentalen -2-yl)-l-methyl-lH- indole-2-carboxamide. To a solution of N-(3-chloro-4-fluorophenyl)-l-methyl-3-(5- oxooctahydropentalen-2-yl)-lH-indole-2-carboxamide (120 mg, 0.28 mmol) in MeOH (3 mL) was added NaB¾ (2 eq, 0.57 mmol) and the reaction mixture stirred at rt for 1 h. The mixture was quenched with water and concentrated in vacuo. The residue was purified by prep-TLC and reserved column chromatography to afford a white solid N-(3-chloro-4- fluorophenyl)-3-(5-hydroxyoctahydropentalen-2-yl)- 1-methyl- lH-indole-2-carboxamide. MS (m/z): Calcd.: 426.1, Found: 427.1 [M + 1] + ; ^-NMR (400 MHz, CDCh): d 8.00 (d, /= 8.0 Hz, 1H), 7.86 (s, 1H), 7.52 (s, 1H), 7.44 - 7.36 (m, 1H), 7.36 - 7.28 (m, 2H), 7.20 - 7.06 (m, 2H), 4.52 - 4.42 (m, 1H), 3.80 (s, 3H), 3.54 - 3.38 (m, 1H), 2.72 - 2.56 (m, 2H), 2.44 - 2.28 (m, 2H), 2.28 - 2.14 (m, 2H), 2.12 - 2.00 (m, 2H), 1.76 - 1.64 (m, 2H), 1.54 (s, 1H) ppm.

Intermediate 9

Ethyl 3-bromo-lH-pyrrolo[3,2-b]pyridine-2-carboxylate. To a solution of ethyl lH-pyrrolo[3,2-b]pyridine-2-carboxylate (1.1 g, 5.79 mmol) in DCM (20 mL) was added NBS (1.1 eq, 6.37 mmol) and the reaction stirred for 2 h at rt. The mixture was washed with water (20 mL x 2) and the organic layer was dried over anhydrous Na2SC>4 then concentrated in vacuo. The residue was purified by recrystallization (1:1 ( v/v ) petroleum ether/EtOAc) to afford ethyl 3-bromo-lH-pyrrolo[3,2-b]pyridine-2-carboxylate as an off-white solid. MS (m/z): Calcd.: 267.9, Found: 269.1 [M + 2] + .

Intermediate 10

Ethyl 3-bromo-l-methyl-lH-pyrrolo[3,2-b]pyridine-2-carboxylate. To a solution of ethyl 3-bromo-lH-pyrrolo[3,2-b]pyridine-2-carboxylate (1.34 g, 4.99 mmol) in dry DMF (20 mL) was added NaH (60%, 1.1 eq, 5.48 mmol). The mixture was stirred for 20 min at rt. Mel (1.1 eq, 5.48 mmol) was then added and the reaction stirred for 2 h at room temperature. The reaction mixture was added to an aqueous LiCl solution (40 mL) and extracted with EtOAc (20 mL x 2). The combined organic layers were dried over anhydrous Na 2 S0 4 and concentrated in vacuo. The crude product was purified by recrystallization (3 : 1 ( v/v ) petroleum ether/EtOAc) to afford ethyl 3-bromo-l-methyl-lH-pyrrolo[3,2-b]pyridine-2- carboxylate as an off-white solid. MS (m/z): Calcd.: 283.0, Found: 285.1 [M + 2] + .

Intermediate 11

3-Bromo-N-(3-chloro-4-fluorophenyl)-l-methyl-lH-pyrrolo[3 ,2-b]pyridine-2- carboxamide. To a solution of ethyl 3-bromo-l-methyl-lH-pyrrolo[3,2-b]pyridine-2- carboxylate (700 mg, 2.47 mmol) in dry toluene (10 mL) was added AlMe 3 (2.0 M, 4.0 eq, 9.89 mmol) at rt and the mixture stirred for 0.5 h. 3-Chloro-4-fluoroaniline (2.0 eq, 4.95 mmol) was then added and stirring continued overnight at 90 °C under an Ar atmosphere. The mixture was quenched with MeOH and filtered. To the filtrate was added water (20 mL) and the mixture extracted with EtOAc (10 mL x 2). The combined organic layers were dried over anhydrous Na2SC>4 and concentrated in vacuo. The residue was purified by prep-TLC to afford an off-white solid 3-bromo-N-(3-chloro-4-fluorophenyl)-l-methyl-lH-pyrrolo[3,2- b]pyridine-2-carboxamide. MS ( m/z ): Calcd.: 379.9, Lound: 382.0 [M + 2] + .

Intermediate 12

N-(3-Chloro-4-fluorophenyl)-l-methyl-3-(5-oxo-l,3a,4,5,6, 6a- hexahydropentalen-2-yl)-lH-pyrrolo[3,2-b]pyridine-2-carboxam ide. To a solution of 3- bromo-N-(3-chloro-4-fluorophenyl)-l-methyl-lH-pyrrolo[3,2-b] pyridine-2-carboxamide (175 mg, 0.46 mmol) in a solution of dioxane/water (5/1 (v/v), 5 mL) was added 5-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-3,3a,6,6a-tetrahydropen talen-2(lH)-one (1.5 eq, 0.69 mmol), Na2CC>3 (2.5 eq, 1.15 mmol) and Pd(dppf)Cl2 (0.1 eq, 0.05 mmol). The reaction was stirred 3 h at 70 °C under an Ar atmosphere. After cooling to rt, the mixture was filtered and EtOAc (20 mL) added to the filtrate. This solution washed with water (20 mL x 2), dried over anhydrous Na2SC>4 then concentrated in vacuo. The residue was purified by prep-TLC to afford a yellow oil N-(3-chloro-4-fluorophenyl)-l-methyl-3-(5-oxo-l,3a,4,5,6,6a- hexahydropentalen-2-yl)-lH-pyrrolo[3,2-b]pyridine-2-carboxam ide. MS (m/z.y. Calcd.: 423.1, Lound: 424.1 [M + 1] + .

Example 45

N-(3-Chloro-4-fluorophenyl)-l-methyl-3-(5-oxooctahydropen talen-2-yl)-lH- pyrrolo[3,2-b]pyridine-2-carboxamide. To a solution of N-(3-chloro-4-fluorophenyl)-l- methyl-3-(5-oxo-l,3a,4,5,6,6a-hexahydropentalen-2-yl)-lH-pyr rolo[3,2-b]pyridine-2- carboxamide (266 mg, 0.63 mmol) in THF (10 mL) was added MeOH (3 drops) and Pd/C (30 mg). The reaction mixture was stirred for 4 h at rt under a ¾ atmosphere (balloon). The mixture was filtered, and the filtrate was concentrated in vacuo. The residue was purified by reversed column chromatography to afford a white solid N-(3-chloro-4-fluorophenyl)-l- methyl-3-(5-oxooctahydropentalen-2-yl)-lH-pyrrolo[3,2-b]pyri dine-2-carboxamide. MS (m/z): Calcd.: 425.0, Found: 426.2 [M + 1] + ; ¾ NMR (400 MHz, CDCb): d 8.46 (dd, / =

4.6, 1.4 Hz, 1H), 7.88 (d, / = 4.4 Hz, 1H), 7.66 - 7.56 (m, 2H), 7.48 - 7.40 (m, 1H), 7.23 - 7.14 (m, 2H), 3.85 (s, 3H), 3.70 - 3.58 (m, 1H), 2.92 - 2.78 (m, 2H), 2.64 (dd, /= 19.2, 4.0 Hz, 2H), 2.58 - 2.46 (m, 4H), 2.34 - 2.24 (m, 2H) ppm.

Example 47

N-(3-Chloro-4-fluorophenyl)-3-(5-hydroxyoctahydropentalen -2-yl)-l-methyl-lH- pyrrolo[3,2-b]pyridine-2-carboxamide. To a solution of N-(3-chloro-4-fluorophenyl)-l- methyl-3-(5-oxooctahydropentalen-2-yl)-lH-pyrrolo[3,2-b]pyri dine-2-carboxamide (130 mg, 0.31 mmol) in MeOH (3 ml) was added NaBtLi (2 eq, 0.61 mmol) and the reaction stirred at rt for 1 h. The reaction was quenched with water and concentrated in vacuo. The residue was purified by prep-TLC and reversed column chromatography to afford a white solid N-(3- chloro-4-fluorophenyl)-3-(5-hydroxyoctahydropentalen-2-yl)-l -methyl-lH-pyrrolo[3,2- b]pyridine-2-carboxamide. MS (m/z) Calcd.: 427.1, Found: 428.2 [M + 1] + ; ¾ NMR (400 MHz, CDCh): d 8.42 (dd, 7 = 4.8, 1.2 Hz, 1H), 7.90 - 7.87 (m, 1H), 7.69 (s, 1H), 7.64 (dd, 7 = 8.4, 1.2 Hz, 1H), 7.46 - 7.43 (m, 1H), 7.23 - 7.15 (m, 2H), 4.85 (brs, 1H), 4.39 - 4.35 (m, 1H), 3.84 (s, 3H), 3.58 - 3.48 (m, 1H), 2.79 - 2.62 (m, 4H), 2.38 - 2.30 (m, 2H), 2.03 - 1.95 (m, 2H), 1.89 - 1.81 (m, 2H) ppm. VI. Biological Data

Assay Measuring Activity of Test Compounds on Viral Production from HepAD38 Cells

HepAD38 cells grown in a T-150 flask (Corning, cat#: 430825) with Growth Medium (DMEM/F12 (1:1) (Hyclone, cat#: SH30023.02), IX Pen/Strep (Invitrogen, cat#: 15140- 122), 10% FBS (Tissue Culture Biologies, cat#: 101), 250 pg/mF G418 (Alfa Aesar, cat#: J62671), lpg/mF Tetracycline (Teknova, cat#: T3320)) were detached with 0.25% trypsin- EDTA (Invitrogen, cat#: 25200-056). Tetracycline-free treatment medium (15 ml, DMEM/F12 (1:1) T lx Pen/step, with 2% FBS, Tet-system approved (Clontech, cat#: 631106) were then added to mix, transferred into a 50 mF conical tube (Falcon, cat#: 21008-918,) and spun at 1300 rpm for 5 min. Pelleted cells were then re-suspended/washed with 50 mF of IX DPBS (Invitrogen, cat#: 14190-136) 2 times and 50 mF treatment medium twice. HepAD38 cells were then re-suspended with 10 mL of treatment medium, syringed and counted. Wells of 96-well clear bottom TC plate (Coming, cat#: 3904,) were seeded at 50,000 cells/well in 180 pF of treatment medium, and 20 pF of either 10% DMSO (Sigma, cat#: D4540) as controls or a 10X solution of test compounds in 10% DMSO in treatment media was added for a final compound concentration starting at 10 pM, and plates were incubated in 5% CO2 incubator at 37°C for 5 days.

Subsequently viral load production was assayed by quantitative PCR (qPCR) of the HBV core sequence. PCR reaction mixture containing forward primers HBV-f 5'- CTGTGCCTTGGGTGGCTTT-3’ (IDT DNA), Reverse primers HBV-r 5’- A AGG AA AG A AGTC AG A AGGC AA A A- 3 ' (IDT DNA), Fluorescent TaqMan tm Probes HB V-probe 5 '-FAM/AGCTCCAAA/ZEN/TTCTTTATAAGGGTCGATGTC/3IABkFQ -3 ' (IDT DNA), 10 pF/well of PerfeCTa ® qPCR ToughMix® (Quanta Biosciences, Cat#: 95114- 05K), and 6 pF/well of DEPC water (Alfa Aesar, cat#: J62087) was prepared. Four pF of supernatant was added to 16 pF of the reaction mixture in a qPCR plate (Applied Biosytems, Cat#: 4309849), sealed with a film (Applied Biosystems, Cat#: 4311971), centrifuged for a few seconds, and subsequently run on an Applied Biosystems VIIA7. The PCR mixture was incubated at 45°C for 5 min, then 95 °C for 10 min, followed by 40 cycles of 10 seconds at 95 °C and 20 seconds at 60 °C. Viral load was quantified against known HBV DNA standards by using ViiA™ 7 Software. Viral load in the supernatant from wells with treated cells were compared against viral load in supernatant from DMSO control wells (> 3 per plate). Cell viability assay was performed with CellTiter-Glo Luminescent Cell Viability Assay (Promega, cat#: G7573) with modification. Mixed appropriate amount of CellTiter- Glo (CTG) IX DPBS in a 1: 1 ratio, added 100 uL of the mixture to each well followed completely removal of all supernatant in each well without touching cell surface. Incubated the plate at room temperature for 10 min on an orbital shaker, and then read the plate with a plate reader (TECAN M1000 or Envision). ECso or CC50 values were calculated through curve-fitting of the four-parameter nonlinear-logistic -regression model (GraphPad Prism or Dotmatics). CC50 values were all >10 mM.

Table 1- gives the viral load lowering EC50 values for exemplified compounds of the invention grouped in the following ranges: A indicates EC50 < 0.1 mM; B indicates EC50 of >0.1 to <1.0 mM; C indicates EC50 of >1.0 to <10 mM.

Table 1. Viral load lowering for exemplified compounds of the invention

VII. Stereochemistry of Examples

AIA-225

5-Amino-N-(3-chloro-4-fluorophenyl)-3-(5-hydroxy-5-(methy lthiomethyl)- octahydropentalen-2-yl)-l-methyl-lH-pyrazole-4-carboxamide. To a solution of 5-amino- N-(3-chloro-4-fluorophenyl)-3-(hexahydro-rH-spiro[oxirane-2, 2'-pentalene]-5'-yl)-l-methyl- lH-pyrazole-4-carboxamide (200 mg, 0.495 mmol) in THF/H2O (6 mL/2 mL) was added NaSMe (138.6 mg, 1.98 mmol). The mixture was stirred at rt overnight. The solvent was removed and the crude product purified by silica gel column chromatography using 3 : 1 (v/v) petroleum ether/ethyl acetate to afford 5-amino-N-(3-chloro-4-fluorophenyl)-3-(5-hydroxy-5- (methylthiomethyl)octahydropentalen-2-yl)-l -methyl- lH-pyrazole-4-carboxamide (100 mg, 44.7%) as a yellow solid. MS ( m/z ): Calcd.: 452.1, Found: 452.2 [M+l] + . AIA-227-1, AIA-227-2

5-Amino-N-(3-chloro-4-fluorophenyl)-3-((2r,5r)-5-hydroxy- 5- (methylsulfonylmethyl)octahydropentalen-2-yl)-l-methyl-lH-py razole-4-carboxamide (AIA-227-1) and 5-Amino-N-(3-chloro-4-fluorophenyl)-3-((2s,5s)-5-hydroxy-5- (methylsulfonylmethyl)octahydropentalen-2-yl)-l-methyl-lH-py razole-4-carboxamide (AIA-227-2). To a solution of 5-amino-N-(3-chloro-4-fluorophenyl)-3-(5-hydroxy-5- (methylthiomethyl)octahydropentalen-2-yl)- 1 -methyl- 1 H-pyrazole-4-carboxamide (100 mg, 0.22 mmol) in DCM (5 mL) was added m-CPBA (114.8 mg, 0.66 mmol). The mixture was stirred at rt overnight. The solvent was removed, and the crude material purified by silica gel column chromatography using 3:1 (v/v) DCM/MeOH to afford AIA-227 (40 mg, 37.3%) as a white solid. MS ( m/z ): Calcd.: 484.1, Found: 484.3 [M+l] + . AIA-227 was separated by SFC to give AIA-227-1 (4 mg) as a white solid and AIA-227-2 (4 mg) as a white solid. AIA-227 - 1: l H NMR (400 MHz, DMSO-rfc): d 8.95 (s, 1H), 7.91 (dd, / = 6.8, 2.4 Hz, 1H), 7.54 - 7.50 (m, 1H), 7.35 (t, / = 9.2 Hz, 1H), 5.97 (s, 2H), 4.79 (s, 1H), 3.59 - 3.53 (m, 1H), 3.49 (s, 3H),

3.35 (s, 2H), 2.97 (s, 3H), 2.67-2.60 (m, 2H), 2.18 - 2.12 (m, 2H), 2.07 - 2.02 (m, 2H), 1.45 -

1.36 (m, 4H) ppm. AIA-227-2: l H NMR (400 MHz, DMSO-d6): d 8.94 (s, 1H), 7.91 (dd, / = 2.8, 2.4 Hz, 1H), 7.53 - 7.49 (m, 1H), 7.34 (t, / = 9.2 Hz, 1H), 5.97 (s, 2H), 4.87 (s, 1H), 3.49 (s, 3H), 3.43 - 3.35 (m, 1H), 3.25 (s, 2H), 2.97 (s, 3H), 2.49 (s, 2H), 2.15 - 2.09 (m, 2H), 2.02 - 1.97 (m, 2H), 1.73 - 1.60 (m, 4H) ppm. AIA-227-2

Alternative synthesis of 5-amino-N-(3-chloro-4-fluorophenyl)-3-((2s,5s)-5- hydroxy-5-(methylsulfonylmethyl)octahydropentalen-2-yl)-l-me thyl-lH-pyrazole-4- carboxamide. To a solution of dimethylsulfone (77.0 g, 818.7 mmol) in THF (800 mL) was added n-BuLi (327.5 mL, 818.7 mmol, 2.5M) dropwise at -78 °C. The resulting solution was allowed to warm to -20 °C and stirred for 1 hr. The reaction was cooled to -78 °C, and a solution of AIA-002 (40.0 g, 102.3 mmol) in anhydrous tetrahydrofuran (1200 mL) was added over 2 hr. The mixture was warmed to rt and stirred for an additional 4 hr. The reaction mixture was quenched with saturated aqueous ammonium chloride solution (200 mL). The solvent was removed, followed by dilution with water, extraction with ethyl acetate (3 x 200 mL), drying over Na2SC>4, filtration, and concentration to give the crude product.

The crude product was purified by column chromatography using 0-5% (v/v) methanol in DCM and basic prep-HPLC to afford 5-amino-N-(3-chloro-4-fluorophenyl)-3-((2s,5s)-5- hydroxy-5-(methylsulfonylmethyl)octahydropentalen-2-yl)-l-me thyl-lH-pyrazole-4- carboxamide (26.0 g, 52.4%) as a white solid. MS (m/z): Calcd.: 484.1, Found: 485.2 [M + 1] + ; ¾ NMR (400 MHz, DMSO-ifc): d 8.96 (s, 1H), 7.92 (dd, /= 6.8, 2.8 Hz, 1H), 7.54 - 7.50 (m, 1H), 7.35 (t, /= 8.8 Hz, 1H), 5.98 (s, 2H), 4.88 (s, 1H), 3.49 (s, 3H), 3.42 - 3.37 (m, 1H), 3.25 (s, 2H), 2.97 (s, 3H), 2.15 - 2.10 (m, 2H), 2.03 - 1.97 (m, 2H), 1.73 - 1.60 (m, 4H) ppm.

A crystal with size of 0.08 x 0.10 x 0.20mm of compound AIA-227-2 was obtained from EtOH after 20 days of volatilization and was used for X-ray diffraction data collection. The data were collected on a Bruker SMART CCD area-detector diffractometer at room temperature using CuKa radiation by w/f scan mode. 10846 reflections were collected, of which 3754 reflections were unique (Rint = 0.0507).

The crystal belongs to monoclinic crystal system, with a space group P2i/c. The unit cell parameters were as follows: a= 6.6143(3), £>=14.0381(8), c=23.6870(14)A, a=y=90.0°, ^97.702(3) °, V= 2179.5(2)A 3 , Z=4.

The structure was solved by direct methods and all of the non-H atoms were refined against F 2 by full-matrix least-squares methods using the SHELXTL program. All H atoms were placed in geometrically idealized positions and constrained to ride on their parent atoms. Multi-scans absorption correction method was used, and the maximum and minimum transmission parameters were 0.7531 and 0.6017, respectively. The final R, wRi, GOF are 0.0457, 0.1293 and 1.024, respectively.

There is one C 21 H 26 FCIN 4 O 4 S molecule in the asymmetric unit and hydrogen bonds can be found between them, which play an important role for the stable packing of the crystal structure.

The ORTEP plot for compound AIA-227-2 is present in Fig. 1. The relative stereochemistry scheme of compound AIA-227-2 is shown in Fig. 2. The depictions of stereochemistry in the chemical structures of related examples are based on this assignment.

INCORPORATION BY REFERENCE

All publications and patents mentioned herein, including those items listed below, are hereby incorporated by reference in their entirety for all purposes as if each individual publication or patent was specifically and individually incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.

EQUIVALENTS

While specific embodiments of the subject disclosure have been discussed, the above specification is illustrative and not restrictive. Many variations of the disclosure will become apparent to those skilled in the art upon review of this specification. The full scope of the disclosure should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.

Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present disclosure.