Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HIGHLY ACTIVE COMPOUNDS AGAINST COVID-19
Document Type and Number:
WIPO Patent Application WO/2021/173713
Kind Code:
A1
Abstract:
The present invention is the use of purine nucleotide phosphoramidates or pharmaceutically acceptable salts thereof administered in an effective amount for the treatment or prevention of COVID-19, an infection caused by the SARS CoV-2 virus in a host, for example a human, in need thereof.

Inventors:
SOMMADOSSI JEAN-PIERRE (US)
MOUSSA ADEL (US)
Application Number:
PCT/US2021/019468
Publication Date:
September 02, 2021
Filing Date:
February 24, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ATEA PHARMACEUTICALS INC (US)
International Classes:
A61K31/7076; A61P31/14; C07H19/207
Domestic Patent References:
WO2016144918A12016-09-15
WO2018048937A12018-03-15
WO2019200005A12019-10-17
WO2020117966A12020-06-11
WO2018144640A12018-08-09
WO2013177219A12013-11-28
WO2013039920A12013-03-21
WO2014124430A12014-08-14
WO2010091386A22010-08-12
WO2014169278A12014-10-16
WO2014169280A22014-10-16
Foreign References:
US20190201433A12019-07-04
US20190255085A12019-08-22
US20110223659A12011-09-15
US10874687B12020-12-29
US20180016301W2018-01-31
US9828410B22017-11-28
US10000523B22018-06-19
US10005811B22018-06-26
US10239911B22019-03-26
US10815266B22020-10-27
US10870672B22020-12-22
US10870673B22020-12-22
US10875885B22020-12-29
US10874687B12020-12-29
US20190201433A12019-07-04
US20200222442A12020-07-16
US10519186B22019-12-31
US10906928B22021-02-02
US10894804B22021-01-19
US8933053B22015-01-13
US8759318B22014-06-24
US8658616B22014-02-25
US8263575B22012-09-11
US8119779B22012-02-21
US7951787B22011-05-31
US7115590B12006-10-03
US8895723B22014-11-25
US8871737B22014-10-28
US8772474B22014-07-08
US7517858B12009-04-14
US8889658B22014-11-18
US8846643B22014-09-30
US8710030B22014-04-29
US8309565B22012-11-13
US8008308B22011-08-30
US7790703B22010-09-07
US9173893B22015-11-03
US8609627B22013-12-17
Other References:
YOON ET AL.: "Design, Synthesis, and Anti-RNA Virus Activity of 6'-Fluorinated-Aristeromycin Analogues", JOURNAL OF MEDICINAL CHEMISTRY, vol. 62, 7 June 2019 (2019-06-07), pages 6346 - 6362, XP055829404, DOI: 10.1021/acs.jmedchem.9b00781
ZHOU ET AL., NATURE, vol. 579, 2020, pages 270
HUANG ET AL., LANCET, vol. 395, 2000, pages 497
LAU ET AL., PNAS, vol. 102, 2005, pages 14040 - 5
REST ET AL., INFECT GENET EVOL., vol. 3, 2003, pages 219 - 25
SCHOEMANFIELDING, VIROLOGY, vol. 16, 2019, pages 69
LUAN ET AL., BIOCHEM. BIOPHYS. RES. COMMUN, vol. 527, 2020, pages 165
HOFFMAN, M. ET AL., CELL, vol. 181, 2020, pages 271
YANG ET AL., INT. J. BIOL. SCI., vol. 16, 2020, pages 1724
AHN ET AL., ARCH VIROL, vol. 157, 2012, pages 2095
SUBISSI ET AL., PROC. NATL. ACAD. SCI., vol. 111, 2014, pages E3900
GAO ET AL., SCIENCE, vol. 368, 2020, pages 779 - 782
GOOD, S.S. ET AL., PLOS ONE, vol. 15, no. l, 2020, pages e0227104
BERLIBA ET AL., ANTIMICROB. AGENTS CHEMOTHER, vol. 63, no. 12, 2019, pages e01201 - 19
THEODORA W. GREEN: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
OGILVIE ET AL., CAN. J. CHEM., vol. 57, 1979, pages 2230
BERLIBA, E. ET AL., ANTIMICROB. AGENTS CHEMTHER., vol. 63, 2020, pages eO11201 - 19
MUNGAR, Q ET AL., EASL ABSTRACT, 2020
"Remington's Pharmaceutical Sciences", 1985, PUBLISHING COMPANY, pages: 1418
"Design, Synthesis, and Characterization of a Series of Cytochrome P(450) 3A-Activated Prodrugs (HepDirect Prodrugs) Useful for Targeting Phosph(on)ate-Based Drugs to the Liver", J. AM. CHEM. SOC., vol. 126, 2004, pages 5154 - 5163
A. RAYK. HOSTETLER: "Application of kinase bypass strategies to nucleoside antivirals", ANTIVIRAL RESEARCH, 2011, pages 277 - 291, XP028325608, DOI: 10.1016/j.antiviral.2011.08.015
M. SOFIA: "Nucleotide prodrugs for HCV therapy", ANTIVIRAL CHEMISTRY AND CHEMOTHERAPY, vol. 22, no. 23, 2011, pages 49
S. PEYROTTES ET AL.: "SATE Pronucleotide Approaches: An Overview", MINI REVIEWS IN MEDICINAL CHEMISTRY, vol. 4, 2004, pages 395, XP008088559
AIM ET AL.: "Geographical and temporal distribution of SARS-CoV-2 clades in the WHO European Region, January to June 2020", EURO SURVEILLANCE: BULLETIN EUROPEAN SUR LES MALADIES TRANSMISSIBLES = EUROPEAN COMMUNICABLE DISEASE BULLETIN, vol. 25, no. 32
GUAN ET AL.: "A genetic barcode of SARS-CoV-2 for monitoring global distribution of different clades during the COVID-19 pandemic.", INT J INFECT DIS., vol. 100, November 2020 (2020-11-01), pages 216 - 223, XP086345175, DOI: 10.1016/j.ijid.2020.08.052
RAMBAUT ET AL., PHYLOGENETIC ASSIGNMENT OF NAMED GLOBAL OUTBREAK LINEAGES, Retrieved from the Internet
RAMBAUT ET AL.: "A dynamic nomenclature proposal for SARS-CoV-2 lineages to assist genomic epidemiology", NAT MICROBIOL., vol. 5, no. 11, November 2020 (2020-11-01), pages 1403 - 1407, XP037277086, DOI: 10.1038/s41564-020-0770-5
RAMBAUT ET AL., SARS-COV-2 LINEAGES, Retrieved from the Internet
HERTEL ET AL., J. ORG. CHEM., vol. 53, 1988, pages 2406
PINHO ET AL., J. ORG. CHEM., vol. 27, 2017, pages 3468
REED, LJMUENCH, H. AM., J. HYGIENE, vol. 27, 1948, pages 493 - 497
PAUWELS, R ET AL., J. VIROL. METHODS, vol. 20, no. 4, 1988, pages 309 - 321
REED, LJMUENCH, H., AM. J. HYGIENE, vol. 27, 1948, pages 493 - 497
WANG, M. ET AL., CELL RESEARCH, vol. 30, 2020, pages 269
JOMSDOTTIR, H.R., VIROL. J., vol. 13, 2016, pages 24
JOMSDOTTIR, H.R., VIROL. J., vol. 13, no. 24, pages 2016
SHEAHAN, T.P. ET AL., SCI. TRANSL. MED., vol. 12, 2020, pages eabb5883
SUBISSI, L., PROC. NATL. ACAD. SCI. USA, vol. 111, no. 37, 2014, pages 3900 - 9
FERRON, F., PROC. NATL. ACAD. SCI. USA, vol. 115, no. 2, 2018, pages 162 - 171
CRAPO, J.D. ET AL., AM. REV. RESPIR. DIS., vol. 126, no. 2, 1982, pages 332 - 7
DEVAL, J. ET AL., PLOS PATHOG, vol. 11, no. 6, 2015, pages el004995
Attorney, Agent or Firm:
BELLOWS, Brent, R. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula I:

Formula I or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein:

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl; and

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl

2 A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula II: or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein: R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R4b are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl; and

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

3. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula IP or Formula IV:

Formula IV or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein:

X is selected from Ci-C3haloalkyl, C2-C4alkenyl, C2-C4alkynyl, and Ci- C3hydroxyalkyl;

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl; R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl; and

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

4. The method of claim 3, wherein the compound is selected from Formula Ilia, Formula Illb, Formula IIIc, Formula Hid, Formula Hie, and Formula Illf :

Formula IIIc or a pharmaceutically acceptable salt thereof.

5. The method of claim 3, wherein the compound is selected from Formula IVa, Formula IYb, Formula IVc, Formula IVd, Formula IVe, and Formula IVf:

Formula IVe

Formula IVf or a pharmaceutically acceptable salt thereof.

6. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula V:

Formula V or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein:

Y and Y’ are independently selected from F and Cl;

R1 is selected from Ci-C6alkyl, C3-C6cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl; and

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

7. The method of claim 6, wherein the compound is of the Formula: or a pharmaceutically acceptable salt thereof.

A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula VF

Formula VI or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R4b are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl;

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl;

R6 is selected from hydrogen, -C(0)R6A, -C(0)0R6A, Ci.6alkyl, -CH2-0-R6A;

R6A is selected from hydrogen, Ci-6alkyl, Ci-Cehaloalkyl, aryl, aryl(Ci-6alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R7is NH2, H, or - R¾9;

R8 and R9 are independently selected from hydrogen, Ci-6alkyl, -C(0)R6A, and -C(0)0R6A;

Y is selected from F and Cl; and

Z is selected from methyl, Ci-C3haloalkyl, C2-C4alkenyl, C2-C4alkynyl, Ci- C3hydroxyalkyl, and halogen. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula VIF

Formula V 11 or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein B is selected from

R1 is selected from Ci-C6alkyl, C3-C6cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl;

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl;

R6 is selected from hydrogen, -C(0)R6A, -C(0)0R6A, Ci^alkyl, -CH2-0-R6A, and -C(0)NR6BR6C;

R6A is selected from hydrogen, Ci^alkyl, Ci-C6haloalkyl, Ci-2oalkyl and C2- 2oalkenyl;

R6B and R6C are independently selected from hydrogen, Ci-2oalkyl, C2-2oalkenyl, aryl, arylalkyl, heteroaryl, and heteroarylalkyl;

R7 is NH2, H, or -NR8R9;

R8 and R9 are independently selected from hydrogen, Ci-6alkyl, -C(0)R6A, and -C(0)0R6A;

Y is selected from F and Cl;

Z is selected from methyl, Ci-C3haloalkyl, C2-C4alkenyl, C2-C4alkynyl, Ci- Cshydroxyalkyl, and halogen;

R40 is selected from H, Ci-3alkoxy, Ci-3alkyl, N3, CN, and halogen;

R41 is selected from H, Ci-3alkyl and halogen; and R42a and R42¾ are selected from Ci.3alkyl, NH2, H, -NR8R9, and -C(0)NR8R9.

10. The method of claim 9, wherein B is selected from

11. The method of any one of claims 1-10, wherein R2 is aryl.

12. The method of claim 11, wherein R2 is phenyl.

13. The method of any one of claims 1-12, wherein R3 is hydrogen.

14. The method of any one of claims 1-12, wherein R3 is methyl.

15. The method of any one of claims 1-13, wherein R4a is methyl and R4b is hydrogen.

16. The method of any one of claims 1-13, wherein R4a is methyl and R4b is methyl.

17. The method of any one of claims 1-16, wherein R5 is Ci-C6alkyl.

18. The method of claims 17, wherein R5 is selected from methyl, ethyl, and isopropyl.

19. The method of claim 18, wherein R5 is isopropyl.

20. The method of any one of claims 1-10, wherein:

R2 is phenyl;

R3 is hydrogen;

R4a is methyl;

R4b is hydrogen; and R5 is isopropyl.

21. The method of any one of claims 1-10, wherein:

R2 is phenyl;

R3 is hydrogen;

R4a is methyl;

R is hydrogen;

R5 is isopropyl; and the phosphoramidate is in the Sp-configuration.

22. The method of any one of claims 1-10, wherein:

R2 is phenyl;

R3 is hydrogen;

R4a is methyl;

R is hydrogen;

R5 is isopropyl; and the phosphoramidate is in the Rp-configuration.

23. The method of claim 1, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

24. The method of claim 23, wherein the compound is selected from the formula: or a pharmaceutically acceptable salt thereof.

25. The method of claim 23, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

26. The method of claim 23, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

27. The method of claim 23, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

28. The method of claim 23, wherein the compound is selected from the formula: or a pharmaceutically acceptable salt thereof.

29. The method of claim 23, wherein the compound is of the formula:

30. The method of claim 23, wherein the compound is selected from the formula:

31. The method of claim 23, wherein the compound is of the formula:

.CH3

HN

• 0.5 H2S04

32. The method of claim 23, wherein the compound is of the formula:

,CH

HN 3

N

'/ N

CH3 O <' - J 11 A

N NH2

Y°3r B' ^- CH Hrf F • 0.5 H2S04

33. The method of claim 23, wherein the compound is of the formula:

34. The method of claim 23, wherein the compound is selected from the formula:

.CH3

HN

35. The method of claim 4, wherein the compound is selected from:

or a pharmaceutically acceptable salt thereof.

36. The method of claim 7, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

37. The method of claim 36, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

38. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula VIII, Formula IX, or Formula X:

Formula X or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein

R10 is selected from

R10A is a stabilized phosphate prodrug that metabolizes in vivo to a monophosphate, diphosphate, or triphosphate;

R11 is selected from hydrogen and R1; and

R1 is selected from Ci-C6alkyl, C3-C6cycloalkyl, and -C(0)Ci-C6alkyl;

39. The method of claim 38, wherein the compound is selected from or a pharmaceutically acceptable salt thereof.

40. The method of claim 38, wherein compound is of the formula wherein

R10a is a stabilized phosphate prodrug that metabolizes in vivo to a monophosphate, diphosphate, or triphosphate.

41. The method of any one of claims 1-40, wherein the pharmaceutically acceptable carrier is in a dosage form suitable for oral administration.

42. The method of claim 41, wherein the dosage form is a solid dosage form.

43. The method of claim 42, wherein the solid dosage form is a tablet.

44. The method of claim 42, wherein the solid dosage form is a capsule.

45. The method of claim 41, wherein the dosage form is a liquid dosage form.

46. The method of claim 45, wherein the liquid dosage form is a solution or a suspension.

47. The method of any one of claims 1-40, wherein the pharmaceutically acceptable carrier is in a dosage form suitable for intravenous administration.

48. The method of any one of claims 1-40, wherein the pharmaceutically acceptable carrier is in a dosage form suitable for parenteral administration.

49. The method of any one of claims 1-48, wherein the compound is administered for at least one week.

50. The method of any one of claims 1-48, wherein the compound is administered for at least two weeks.

51. The method of any one of claims 1-48, wherein the compound is administered for at least one month.

52. The method of any one of claims 1-48, wherein the compound is administered for at least six weeks.

53. The method of any one of claims 1-48, wherein the compound is administered for at least two months.

54. The method of any one of claims 1-53, wherein the compound is administered once a day.

55. The method of any one of claims 1-53, wherein the compound is administered twice a day.

56. The method of any one of claims 1-53, wherein the compound is administered three times a day.

57. The method of any one of claims 1-56, wherein at least about 500 mg of the compound is administered.

58. The method of any one of claims 1-56, wherein at least about 600 mg of the compound is administered.

59. The method of any one of claims 1-56, wherein at least about 700 mg of the compound is administered.

60. The method of any one of claims 1-56, wherein at least about 800 mg of the compound is administered.

61. The method of any one of claims 1-56, wherein at least about 1000 mg of the compound is administered.

62. The method of any one of claims 1-56, wherein at least 1200 mg of the compound is administered.

63. The method of any one of claims 1-56, wherein at least about 1500 mg of the compound is administered.

64. The method of any one of claims 1-56, wherein at least about 550 mg of the compound is administered once a day.

65. The method of any one of claims 1-56, wherein at least about 550 mg of the compound is administered twice a day.

66. The method of any one of claims 1-56, wherein at least about 600 mg of the compound is administered once a day.

67. The method of any one of claims 1-56, wherein at least about 600 mg of the compound is administered twice a day.

68. The method of any one of claims 1-67, wherein the host is a human.

69. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a human in need thereof comprising administering a compound of the formula: or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically carrier, wherein the compound is administered in an oral dosage of about 550 mg twice a day.

70. A method for the treatment or prevention of COVID-19 caused by the SARS-CoV-2 virus in a human in need thereof comprising administering a compound of the formula: optionally in a pharmaceutically acceptable carrier, wherein the compound is administered in an oral dosage of about 600 mg twice a day.

71. The method of claim 69 or 70, wherein the oral dosage is a tablet.

72. The method of claim 69 or 70, wherein the oral dosage is a capsule.

73. The method of any one of claims 69-72, wherein the compound is administered for at least one week.

74. The method of any one of claims 69-72, wherein the compound is administered for at least two weeks.

75. The method of any one of claims 69-72, wherein the compound is administered for at least one month.

76. The method of any one of claims 1-75, wherein the SARS-CoV-2 virus has developed a natural or drug-induced mutation.

77. The method of claim 76, wherein the natural or drug-induced mutation is in a viral protein selected from an envelope (E) protein, a membrane (M) protein, and a spike (S) protein.

78. The method of claim 77, wherein the mutation is in a spike (S) protein.

79. The method of claim 76, wherein the natural or drug-induced mutation is in a viral protein selected from nspl, nsp2, nsp3, nsp4, nsp5, nsp 6, nsp7, nsp8, nsp9, nsplO, nspl2, nspl3, nspl4, nspl5, and nspl6.

80. The method of claim 76, wherein the natural or drug-induced mutation is in a viral protein selected from ORFlab, ORF3a, ORF6, ORF7a, ORF7b, ORF8, and ORFIO.

81. A compound of Formula I or Formula II:

Formula II or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, in an effective amount for use to treat or prevent the 2019 coronavirus disease (COVID-19) in a host in need thereof, wherein:

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-ealkyl, and C3-7cycloalkyl; and

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl

82. The compound of claim 81, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

83. The compound of claim 81, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

84. The compound of claim 82, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

85. The compound of claim 82, wherein the compound is of the formula:

86. The compound of claim 85, wherein the compound is of the formula:

87. The compound of claim 86, wherein the compound is of the formula:

88. A compound of Formula III or Formula IV:

Formula IV or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, in an effective amount for use to treat or prevent the 2019 coronavirus disease (COVID-19) in a host in need thereof, wherein:

X is selected from Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CH2F, CHF2, CF3, CH2CF3, CH2CHF2, CH2CH2F, CF2CH3, CF2CF3, and CH2C1), C2-C4alkenyl, C2-C4alkynyl, and Ci-C3hydroxyalkyl;

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R4b are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl; and

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

89. The compound of claim 88, wherein the compound is of Formula Ilia, Formula Illb, Formula IIIc, Formula Hid, or Formula Hie:

Formula Ilia

Formula Hid

Formula Illf or a pharmaceutically acceptable salt thereof.

90. The compound of claim 88, wherein the compound is of Formula IVa, Formula IVb, Formula IVc, Formula IVd, or Formula Vie: Formula IVf or a pharmaceutically acceptable salt thereof.

91. A compound of Formula V, Formula VI, or Formula VII :

Formula V 11 or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, wherein

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl;

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl;

R6 is selected from hydrogen, -C(0)R6A, -C(0)0R6A, Ci^alkyl, -CH2-0-R6A;

R6A is selected from hydrogen, Ci-6alkyl, Ci-Cehaloalkyl, aryl, aryl(Ci-6alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R7is NH2, H, or -NR8R9;

R8 and R9 are independently selected from hydrogen, Ci-6alkyl, -C(0)R6A, and -C(0)0R6A;

Y and Y’ are independently selected from F and Cl;

Z is selected from methyl, Ci-C3haloalkyl, C2-C4alkenyl, C2-C4alkynyl, Ci- Cshydroxyalkyl, and halogen;

B is selected from

R40 is selected from H, Ci-3alkoxy, Ci^alkyl, N3, CN, and halogen;

R41 is selected from H, Ci-3alkyl and halogen; and

R42a and R42b are selected from Ci.3alkyl, NH2, H, -NR¾9, and -C(0) R¾9.

92. The compound of any one of claims 81-91, wherein the pharmaceutically acceptable carrier is in a dosage form suitable for oral administration.

93. The compound of claim 92, wherein the dosage form is a solid dosage form.

94. The compound of claim 93, wherein the solid dosage form is a tablet.

95. The compound of claim 93, wherein the solid dosage form is a capsule.

96. The compound of claim 92, wherein the dosage form is a liquid dosage form.

97. The compound of claim 96, wherein the liquid dosage form is a solution or a suspension.

98. The use of a compound of Formula I, Formula II, Formula III, or Formula IV:

Formula I

or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, in the manufacture of a medicament for the treatment or prevention of the 2019 coronavirus disease (COVED- 19) in a host in need thereof wherein

X is selected from Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CHiF, CHF2, CF3, CH2CF3, CH CHF2, CH2CH2F, CF2CH3, CF2CF3, and CH2C1), C2-C4alkenyl, C2-C4alkynyl, and Ci-C3hydroxyalkyl;

R1 is selected from Ci-C6alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7Cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl;

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl

99. The use of claim 98, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

100. The use of claim 99, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

101. The use of claim 100, wherein the compound is of the formula: or a pharmaceutically acceptable salt thereof.

102. The use of claim 99, wherein the compound is of the formula:

• 0.5 H2S04

103. The use of claim 102, wherein the compound is of the formula:

104. The use of claim 103, wherein the compound is of the formula:

105. The use of a compound of Formula V, Formula VI, or Formula VII:

Formula V 11 or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, in the manufacture of a medicament for the treatment or prevention of the 2019 coronavirus disease (COVED- 19) in a host in need thereof wherein

R1 is selected from Ci-Cealkyl, C3-C6Cycloalkyl, and -C(0)Ci-C6alkyl;

R2 is hydrogen, Ci-6alkyl, C3-7cycloalkyl, aryl, aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R3 is hydrogen or Ci-6alkyl;

R4a and R are independently selected from hydrogen, Ci-6alkyl, and C3-7cycloalkyl;

R5 is hydrogen, Ci-6alkyl, Ci-6haloalkyl, C3-7cycloalkyl, aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl;

R6 is selected from hydrogen, -C(0)R6A, -C(0)0R6A, Ci^alkyl, -CH2-0-R6A;

R6A is selected from hydrogen, Ci-ealkyl, Ci-G,haloalkyl, aryl, aryl(Ci-6alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R7 is NH2, H, or -NR8R9;

R8 and R9 are independently selected from hydrogen, Ci-6alkyl, -C(0)R6A, and -C(0)0R6A;

Y is selected from F and Cl; Z is selected from methyl, Ci-C3haloalkyl, C2-C4alkenyl, C2-C4alkynyl, Ci- C3hydroxyalkyl, and halogen;

B is selected from

R40 is selected from H, Ci-3alkoxy, Ci^alkyl, N3, CN, and halogen;

R41 is selected from H, Ci-3alkyl and halogen; and

R42a and R42b are selected from Ci.3alkyl, NH2, H, -NR¾9, and -C(0) R8R9.

106. The use of any one of claims 98-105, wherein the pharmaceutically acceptable carrier is in a dosage form suitable for oral administration.

107. The use of claim 106, wherein the dosage form is a solid dosage form.

108. The use of claim 107, wherein the solid dosage form is a tablet.

109. The use of claim 107, wherein the solid dosage form is a capsule.

110. The use of claim 106, wherein the dosage form is a liquid dosage form.

111. The use of claim 110, wherein the liquid dosage form is a solution or a suspension.

Description:
HIGHLY ACTIVE COMPOUNDS AGAINST COVID-19

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 62/982,670, filed February 27, 2020; U.S. Provisional Application No. 62/994,206, filed March 24, 2020; U.S. Provisional Application No. 63/032,247, filed May 29, 2020; U.S. Provisional Application No. 63/039,352, filed June 15, 2020; U.S. Provisional Application No. 63/040,985, filed June 18, 2020; U.S. Provisional Application No. 63/054,680, filed July 21, 2020; U.S. Provisional 63/073,328 filed September 1, 2020; and, U.S. Provisional 63/146,456 filed February 5, 2021. These applications are incorporated by reference in their entireties for all purposes.

FIELD OF THE INVENTION

The present invention is directed to the use of selected purine nucleotides and their pharmaceutically acceptable salts that have advantageous activity and dosage convenience for the treatment or prevention of the SARS-CoV-2 vims that causes COVID-19 in a host, typically a human, in need thereof.

BACKGROUND OF THE INVENTION

In December 2019, a number of patients in Wuhan, China were diagnosed with pneumonia. These patients exhibited symptoms similar to the SARS (severe acute respiratory syndrome) outbreak in 2002-2003. In January 2020, the infectious cause was identified as a novel coronavirus that was named severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and the resulting disease called coronavirus disease 2019 (COVID-19). This potentially severe and sometimes lethal disease quickly spread throughout the world. On March 11, 2020, the World Health Organization declared COVID-19 a global pandemic.

The majority of patients infected with the SARS-CoV-2 virus exhibit mild, cold-like symptoms, including fever, cough, fatigue, shortness of breath, muscle aches, and loss of taste and/or smell. These symptoms usually resolve with minimal medical care in a few weeks. However, occasionally, symptoms persist for months. The vims may cause long-term damage to the lungs, heart, and brain. Furthermore, in some patients, especially older adults, immunocompromised individuals, or those with underlying conditions, the vims can cause severe symptoms that result in hospitalization, ventilation, and/or death.

SARS-CoV-2 is a coronavims (CoV), which is in the order Nidovirales , family Coronaviridae, subfamily Coronavirinae, which are enveloped vimses with a single-strand, positive-sense RNA genome. SARS-CoV-2 is approximately 30 kilobases in size, which is among the largest known RNA genomes. Related coronaviruses include severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). However, SARS-CoV-2 only shares 79.5% of its genome with SARS-CoV, and is therefore considered a new human-infecting betacoronavirus (Zhou et al. Nature 2020, 579, 270) Compared to SARS-CoV and MERS-CoV, SARS-CoV-2 exhibits a faster human-to-human transmission rate (Huang et al., Lancet 2000, 395, 497), making it particularly challenging to contain and dangerous.

CoVs often originate as enzootic infections that cross the animal-human species barrier and progress to establish zoonotic diseases in humans (Lau et al., PNAS 2005, 102, 14040-5; Rest et al., Infect Genet Evol 2003, 3, 219-25). Cross-species barrier jumps allowed CoVs such as the SARS CoV and the Middle Eastern respiratory syndrome CoV (MERS) to manifest as virulent human viruses (Schoeman and Fielding, Virology 2019, 16, 69). Similarly, genome sequencing has revealed that SARS-CoV-2 is 96% identical at the whole-genome level to a bat coronavirus (Zhou et al. Nature 2020, 579, 270) and therefore most likely originated in bats.

SARS-CoV-2 enters human cells by binding to angiotensin converting enzyme 2 (hACE2) receptors. Spike glycoproteins on the surface of the virus envelope bind to the ACE2 receptor and then the human transmembrane protease serine 2 cleaves and activates the spike protein, which allows SARS-CoV-2 to enter the cell through endocytosis or direct fusion with the host membrane (Luan et al. Biochem. Biophys. Res. Commun. 2020: 527, 165; Hoffman, M. et al. Cell, 2020, 181, 271; Yang et al. Int. J. Biol. Sci. 2020, 16, 1724).

Once inside the cell, SARS-CoV-2 transcription and replication is mediated by a multi subunit polymerase complex. The catalytic subunit of the complex is the RNA-dependent RNA polymerase (RdRp) known as nspl2. While the isolated nspl2 subunit is capable of conducting the polymerase reaction by itself, the presence of cofactors nsp7 and nsp8 significantly increases the efficiency of the polymerase reaction (Ahn et al. Arch Virol. 2012, 157, 2095; Subissi et al. Proc. Natl. Acad. Sci., 2014, 111, E3900).

In April 2020, a crystal structure of the SARS-CoV-2 nsp-12, in complex with nsp-7 and nsp-8, was resolved (Gao et al. Science 2020, 368:779-782). The structure of nspl2 contains a polymerase C-terminal RdRp domain that is connected to an N-terminal extension domain referred to as the nidovirus RdRp-associated nucleotidyltransferase (NiRAN) domain. This NiRAN domain, which is conserved in all nidoviruses that are able to conduct nucleotidylation activity, is characterized by an a and b fold composed of eight a helices and a five stranded b-sheet (Gao et al. Science 2020, 368:779-782). The C-terminal domain has been characterized as a “cupped right hand” domain with finger, thumb, and palm subdomains.

As the SARS-CoV-2 virus has spread throughout the world, it has exhibited a high rate of mutation and a number of mutated forms of the virus are circulating globally. These mutations have the potential to affect the vims’ s ability to cause infections and the rate of transmission. For example, the United Kingdom identified the B.l.1.7 variant, which was named the Variant of Concern 202012/01 by Public Flealth England in the fall of 2020. This variant has eight mutations in the spike region. There is evidence that this variant spreads more quickly and easily and may be associated with increased risk of death. In South Africa, other variants, B.1.351 and 501 Y.V2, have been identified Both variants share some mutations with the B.l 1.7 variant, including the N501Y mutation. Brazil has also identified a variant known as P.1 that contains mutations that may affect the virus’s ability to be recognized by antibodies. For this reason, it is important to develop therapies that are able to treat mutated forms of the vims, especially those with mutations on the spike protein.

The history of creating therapeutics for human coronavims diseases illustrates the complexity and challenges of the problem. There were no commercial vaccines or dmgs approved MERS-CoV and SARS-CoV despite the fact that the vimses were discovered in 2012 and 2003, respectively.

The lack of approved treatment, in combination with its high mortality rate and its ease and speed of transmission, highlights the need for the development of an effective COVET- 19 antiviral medication.

It is therefore an object of the present invention to provide compounds, compositions, and methods for the treatment and prevention of the SARS-CoV-2 vims that causes COVID-19.

SUMMARY OF THE INVENTION

The present invention provides a treatment for the SARS-CoV-2 vims in a host in need thereof comprising administering an effective amount of a selected purine nucleotide compound as further described herein for the advantageous treatment, prevention, or prophylaxis of the SARS- CoV-2 vims that causes COVID-19. These purine nucleotides exhibit focused activity against the vims.

Further, and importantly, these compounds can be administered to hosts, such as humans, in need thereof, using a simple solid oral dosage form that can be conveniently taken at home or generally outside of a medical facility without requiring parenteral administration or hospitalization. If desired or appropriate, the active compounds described herein can alternatively be administered parenterally or orally in a medical facility. The therapy can be used to treat mild, moderate or severe disease. In one embodiment of the present invention, a compound of Formula I or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered in an effective amount to a host, typically a human, in need thereof with COVID- 19 or a host at risk of infection or reinfection with the SARS-CoV-2 virus, i.e., as a prophylactic (and wherein the term prophylactic means total prevention or minimization of acquired infection relative to disease without such prophylactic treatment), wherein:

R 1 is selected from Ci-C 6 alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C 6 alkyl;

R 2 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), C3-7cycloalkyl, or aryl (including phenyl and napthyl) and in an alternative embodiment, R 2 is aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R 3 is hydrogen or Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl);

R 4a and R 4b are independently selected from hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), and C3-7cycloalkyl; and

R 5 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), Ci- 6 haloalkyl, or C3-7cycloalkyl and in an alternative embodiment, R 5 is aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

Non-limiting examples of Ci-Csalkyl include methyl, ethyl, propyl, isopropyl, butyl, t- butyl, sec-butyl, isobutyl, -C bC^CIE -CH(CH2CH3)2, and -CFhCF^C hCFF^. Non-limiting examples of C3-C6Cycloalkyl include cyclopropyl, CFh-cyclopropyl, cyclobutyl, and CFh- cyclobutyl.

A non-limiting example of aryl(Ci-C4alkyl)- is benzyl A non-limiting example aryl is phenyl.

In one embodiment, the SARS-CoV-2 virus is wild-type. In another embodiment, the SARS-CoV-2 virus has developed a natural or drug-induced mutation, for example, but not limited to, a mutation in a viral protein selected from an envelope (E) protein, membrane (M) protein, spike (S) protein, nspl, nsp2, nsp3, nsp4, nsp5, nsp 6, nsp7, nsp8, nsp9, nsplO, nspl2, nspl3, nspl4, nspl5, nsp 16, ORFlab, ORF3a, ORF6, ORF7a, ORF7b, ORF8, and ORFIO. A non-limiting example of a compound of Formula I is Compound 1 or a pharmaceutically acceptable salt thereof. In one embodiment, Compound 1 or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered to a host in need thereof, such as a human, infected with SARS-CoV-2, or to a host at risk of infection with the SARS-CoV- 2 vims, i.e., as a prophylactic.

Compound 1

Compound 1 is depicted above without regard to stereochemistry at the phosphorus atom, which is chiral. Compound 1 can be used either without regard to stereochemistry at the phosphorus, or a phosphoro-racemic form, or with any desired ratio of phosphorus R- and S-enantiomers of the compound, including enantiomerically enriched (i.e., up to at least 90%, 95%, 98%, 99%, or even 100% free of the opposite enantiomer, which are in fact diastereomers because there are multiple chiral carbons in the molecule). Compound 1A is the S-enantiomer and Compound IB is the R- enantiomer.

Compound IB Compound 1, including Compound 1A and Compound IB are potent inhibitors against COVED- 19 caused by the SARS-CoV-2 virus. As described in Example 5 and Example 6, Compound 1A exhibits an EC90 value of 0.64 mM against SARS-Cov-2 in HAE cells (human airway epithelial cells). The assay using HAE cells is an in vitro model of the lung and is a representative system for SARS-CoV-2 replication. It has also surprisingly been discovered that the active triphosphate metabolite of Compound 1A is robustly formed when exposed to normal primary bronchial and nasal epithelial cells. As described in Example 7, when Compound 1A was incubated in human nasal and bronchial epithelial cells, the half-life of the active triphosphate species is greater than 1.5 days in both bronchial and nasal epithelial cells. This could not have been predicted in advance and is especially important in treating patients with early stages of the infection when the virus is heavily concentrated in the nasal and bronchial cells.

The data herein presented shows that the compound concentrates in the lung over the liver, and the previously reported data confirms that the compounds also preferentially concentrate in the liver over the heart (see for example, Example 19 of PCT Application PCT/US2018/016301). Taken together, this data confirms that the compound concentrates in the target organ, the lung, over the liver or heart, reducing toxicity

In one embodiment, Compound 1, such as Compound 1A or Compound IB or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered in an effective amount to a host, for example, a human, in need thereof infected with the SARS-CoV-2 virus, or a host at risk of infection with the SARS-CoV-2 virus, i.e., as a prophylactic. In one embodiment, the pharmaceutically acceptable salt is a hemi-sulfate salt, shown below as Compound 2, Compound 2A, and Compound 2B:

Compound 2

Compound 2B

As described in Example 8, Compound 2A was administered to non-human primates and the intracellular concentration of the active triphosphate species was measured in lung, kidney, and liver cells. Surprisingly, the active triphosphate metabolite concentrates in the lung over the liver (Table 8) and the half-life of the active metabolite in the lung is 9.4 hours (FIG. 7A). This is important because COVID-19 typically presents as a respiratory illness. In fact, the active triphosphate species concentration is 1.6 times higher in the lung than the liver after twice a day oral administration of 30 mg/kg of Compound 2A. Furthermore, the compounds of the present invention may inhibit SARS-CoV-2 infection via a unique mechanism of action that accounts for high selectivity. CoV viral replication is achieved at the RNA-dependent RNA polymerase (RdRp) nspl2 subunit, which is activated by co factors nsp7 and nsp8. As discussed in Example 6, there is a 30-fold difference in Compound 1A activity against SARS-CoV-2 and MERS-CoV, even though MERS-CoY and other CoVs, such as SARS-CoV-1 and SARS-CoV-2, do not exhibit significant structural differences at the RdRp active site. This suggests that polymerase inhibition is not the sole basis for differential activity against these viruses.

COVID-19 is an acute viral infection for which antiviral therapeutics may be effective within the first stage of the infection when viral load is at its maximum and there is rapid viral replication initially in nasal, throat and pulmonary cells. The availability of a potent, safe, oral antiviral administered to individuals infected with SARS-CoV-2 in the early stages of the disease has the potential to avert clinical illness, minimize long term damage, and mitigate the COVED- 19 pandemic. As described above, it has been shown that selected compounds of the present invention are able to concentrate in the lungs over the liver. This is therapeutically beneficial when treating patients in the first stages of infection with the SARS-CoV-2 pathogen when it is desirable to prevent or lessen late-stage viral damage. High concentrations in the lung over the heart and liver is also therapeutically beneficial for treating patients in later stages of the infection.

The present invention also includes the use of a compound of Formula II in an effective amount to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof as described herein: or a pharmaceutically acceptable salt thereof, wherein:

R 1 is selected from Ci-C 6 alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C 6 alkyl;

R 2 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), C3-7cycloalkyl, or aryl (including phenyl and napthyl) and in an alternative embodiment, R 2 is aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R 3 is hydrogen or Cnsalkyl (including methyl, ethyl, propyl, and isopropyl);

R 4a and R 4b are independently selected from hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), and C3-7cycloalkyl; and

R 5 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), Ci- 6 haloalkyl, or C3-7cycloalkyl and in an alternative embodiment, R 5 is aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

In one embodiment, a compound of Formula II or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered in an effective amount to a host in need thereof infected with the SARS-CoV-2 virus, or to a host at risk of infection or reinfection with the SARS-CoV-2 vims, i.e., as a prophylactic.

A non-limiting example of a compound of Formula II is Compound 3 or a pharmaceutically acceptable salt thereof.

In one embodiment, Compound 3 or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered in an effective amount to a host in need thereof infected with the SARS-CoV-2 virus, or to a host at risk of infection with the SARS-CoV- 2 virus, i.e., as a prophylactic. Compound 3 can be used in a phosphoro-racemic form, or with any desired ratio of phosphorus R- and S-enantiomers of the compound, including enantiomerically enriched material up to pure enantiomers. Compound 3A is the S-enantiomer and Compound 3B is the R-enantiomer.

Compound 3B

An additional non-limiting example of Formula II includes Compound 4 Alternative configurations of Compound 4 include Compound 4A and Compound 4B. In one embodiment, Compound 4, optionally in a pharmaceutically acceptable carrier, is administered in an effective amount to a host in need thereof infected with the SARS-CoV-2 virus or to a host at risk of infection, i.e., as a prophylactic.

Compound 4B

The present invention also includes the use of a compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof as described herein:

Formula III or a pharmaceutically acceptable salt thereof, wherein:

X is selected from Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH 3 , CF 2 CFS, and CH C1), C 2 -C alkenyl, C 2 -C 4 alkynyl, and Ci-C3hydroxyalkyl; and R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease is a compound or a pharmaceutically acceptable salt thereof of Formula Ilia, Formula Illb, Formula IIIc, Formula Hid, Formula Hie, or Formula Illf:

Formula IIIc

Formula Illf

The present invention also includes the use of a compound of Formula IV in an effective amount to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof as described herein:

Formula IV or a pharmaceutically acceptable salt thereof, wherein:

X is selected from Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH , CF 2 CF 3 , and CH 2 C1), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, and Ci-C3hydroxyalkyl; and

R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease is a compound or a pharmaceutically acceptable salt thereof of Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula IVe, or Formula IVf:

The present invention also includes the use of a compound of Formula V in an effective amount to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof as described herein: or a pharmaceutically acceptable salt thereof, wherein: Y and Y’ are independently selected from Cl and F; and R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

Non-limiting examples of a compound of Formula V include

The present invention also includes the use of a compound of Formula VI to treat or prevent COVED- 19 in a host in need thereof as described herein: wherein

R 6 is selected from hydrogen, -C(0)R 6A , -C(0)0R 6A , Ci- 6 alkyl, and -CH 2 -0-R 6A and in an alternative embodiment, -C(0)NR 6B R 6C ;

R 6A is selected from hydrogen, Ci^alkyl, Ci-G,haloalkyl (for example, -CHCb, -CCb, -CH2CI, -CF3, -CHF2, -CH2F), aryl, and aryl(Ci- 6 alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl, and in an alternative embodiment, R 6A is selected from Ci-2oalkyl and C2-2oalkenyl;

R 6B and R 6C are independently selected from hydrogen, Ci-2oalkyl, C2-2oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl wherein the Ci-2oalkyl, C2-2oalkenyl, aryl, aryl(Ci- ealkyl)-, heteroaryl, and heteroarylalkyl can optionally be substituted with at least one substituent selected from alkoxy (including but not limited to methoxy and ethoxy), hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R 7 is NFh, H, or -NR¾ 9 ; R 8 and R 9 are independently selected from hydrogen, Ci- 6 alkyl, -C(0)R 6A , and -C(0)0R 6A ; Y is selected from F and Cl;

Z is selected from methyl, Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH 3 , CF 2 CF , and CH 2 C1), C 2 - C4alkenyl, C 2 -C4alkynyl, Ci-C3hydroxyalkyl, and halogen (including Cl and F), and in an alternative embodiment, Z is Ciualkyl; and

R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

Non-limiting examples of R 6 include Additional non-limiting examples of R 6 include: Additional non -limiting examples of R 6 include:

Additional non -limiting examples of R 6 include:

Non-limiting examples of a compound of Formula VI include

5

The present invention also includes the use of a compound of Formula VII to treat or prevent

COVID-19 in a host in need thereof as described herein:

Formula VII wherein

B is selected from

R 6A is selected from hydrogen, Ci^alkyl, Ci-C 6 haloalkyl (for example, -CHCh, -CCI 3 , -CH 2 CI, -CF 3 , -CHF 2 , -CH 2 F), aryl, and aryl(Ci- 6 alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl and in an alternative embodiment, R 6A is selected from Ci- 2 oalkyl and C 2-2 oalkenyl;

R 6B and R 6C are independently selected from hydrogen, Ci- 2 oalkyl, C 2-2 oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl wherein the Ci- 2 oalkyl, C 2-2 oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl can optionally be substituted with at least one substituent selected from alkoxy (including but not limited to methoxy and ethoxy), hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R 7 is NH 2 , H, or -NR 8 R 9 ;

R 8 and R 9 are independently selected from hydrogen, Ci- 6 alkyl, -C(0)R 6A , and -C(0)0R 6A ;

Y is selected from F and Cl;

Z is selected from methyl, Ci-C 3 haloalkyl (including Ci- 3 fluoroalkyl and Ci- 3 chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH 3 , CF 2 CF 3 , and CH 2 C1), C 2 - C4alkenyl, C2-C4alkynyl, Ci-C3hydroxyalkyl, and halogen (including Cl and F), and in an alternative embodiment Z is Ci^alkyl;

R 40 is selected from H, Ci^alkoxy, Ci^alkyl, N 3 , CN, and halogen (including Cl and F);

R 41 is selected from H, Ci- 3 alkyl (including methyl) and halogen (including Cl, F, and Br);

R 42a and R 42¾ are independently selected from Ci alkyl (including methyl), NH 2 , H, - NR 8 R 9 , and -C(0)NR 8 R 9 ; and

R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein. In one embodiment, the invention also includes a compound of Formula Vila, Formula Vllb, Formula Vile, and Formula Vlld:

Non-limiting examples of B include:

The present invention also includes the use of a compound of Formula VIII, Formula IX, or Formula X wherein R 10 is a monophosphate, a diphosphate, a triphosphate, or R 10A wherein R 10A is a stabilized phosphate prodrug that metabolizes in vivo to a monophosphate, diphosphate, or triphosphate to treat or prevent COVID-19 disease in a host in need thereof as described herein:

Formula X wherein

R 10 is selected from

R 10A is a stabilized phosphate prodrug that metabolizes in vivo to a monophosphate, diphosphate, or triphosphate; R 11 is selected from hydrogen and R 1 ; and

R 1 is selected from Ci-C 6 alkyl, C3-C6cycloalkyl, and -C(0)Ci-C 6 alkyl.

Non-limiting examples of compounds of Formula VIII, Formula IX, or Formula X include:

Additional non-limiting examples of compounds of Formula VIII, Formula IX, or Formula

X include:

The phosphorus in any of the Formulas described herein may be chiral and thus can be provided as an R or S enantiomer or mixture thereof, including a racemic mixture. The compound is typically at least 90% free of the opposite enantiomer, and can be at least 95%, 96%, 97%, 98%, 99% or even 100% free of the opposite enantiomer. Unless described otherwise, the compound is at least 90% free of the opposite enantiomer. For example, Compound 1 is depicted without regard to stereochemistry at the phosphorus atom, which is chiral. Compound 1 can be used in a racemic form, or with any desired ratio of phosphorus R p - and S p -enantiomers of the compound, including enantiomerically enriched material up to pure enantiomers. Compound 1A has S-stereochemistry at the phosphorus and Compound IB has R-stereochemistry at the phosphorus. In some embodiments, Compound 1 is used in a form at least 90% free of the opposite enantiomer, and can be at least 98%, 99% or even 100% free of the opposite enantiomer. For example, Compound 1A can be at least 90%, 95%, 98%, 99%, or even 100% free of the opposite R p - enantiomer. Alternatively, Compound IB can be at least 90%, 95%, 98%, 99%, or even 100% free of the opposite S p -enantiomer.

Similarly, Compound 2 is depicted without regard to stereochemistry at the phosphorus atom, which is chiral. Compound 2 can be used in a racemic form, or with any desired ratio of phosphorus R- and S-enantiomers of the compound, including enantiomerically enriched material up to pure enantiomers. Compound 2A has S-stereochemistry at the phosphorus and Compound 2B has R-stereochemistry at the phosphorus. In some embodiments, Compound 2 is used in a form at least 90% free of the opposite enantiomer, and can be at least 98%, 99% or even 100% free of the opposite enantiomer. In one embodiment, Compound 2A can be at least 90%, 95%, 98%, 99%, or even 100% free of the opposite R p -enantiomer. In one embodiment, Compound 2B can be at least 90%, 95%, 98%, 99%, or even 100% free of the opposite S p -enantiomer.

Unless described otherwise, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII drawn with regard to stereochemistry at the phosphorus atom is at least 90% free of the opposite enantiomer.

Compounds, compositions, dosage forms, and methods are provided for the treatment of COVED- 19 caused by the SARS-CoV-2 virus in a host in need thereof via administration of an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof can also be used in an effective amount prophylactically to prevent or restrict the progression of COVID-19 in a host in need thereof who has been exposed to the virus or who is at risk of infection or reinfection.

The weight of active compound in the dosage form described herein is with respect to either the free form or the salt form of the compound unless otherwise specifically indicated. For example, approximately 600 mg of Compound 2 is the equivalent of approximately 550 mg of Compound 1. In one non-limiting embodiment, a loading dose is 1100 mg/day (free base) (i.e., 1200 mg/day hemisulfate salt of Compound 1), and a maintenance dose is 550 mg/day (free base) (i.e, 600 mg/day of hemisulfate salt)). In one embodiment, the loading dose is administered once and the maintenance dose is administered twice a day for at least 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 days.

In certain embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of at least about 100, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, or 1700 mg. In certain embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered at a dose of at least 250 mg, 500 mg, at least 600 mg, at least 700 mg, at least 800 mg, at least 900 mg, at least 1000 mg, at least 1100 mg, at least 1200 mg, at least 1300 mg, at least 1400 mg or at least 1500 mg.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, including Compound 1 or a pharmaceutically acceptable salt thereof, for example Compound 2, is administered in a dosage form of about 550 mg once day.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, including Compound 1 or a pharmaceutically acceptable salt thereof, for example Compound 2, is administered in a dosage form of about 600 mg once a day.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 550 mg twice a day.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 600 mg twice a day.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 550 mg twice a day for at least five days, optionally with the standard of care. In one embodiment, Compound 1 or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 550 mg twice a day for at least five days, optionally with the standard of care. In one embodiment, Compound 1 is Compound 1A. In one embodiment, Compound 1 is Compound IB.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 600 mg twice a day for at least of days, optionally with the standard of care. In one embodiment, Compound 2 or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 600 mg twice a day for at least five days, optionally with the standard of care. In one embodiment, Compound 2 is Compound 2A. In one embodiment, Compound 2 is Compound 2B.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 550 mg twice a day for at least about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or more days, optionally with the standard of care. In one embodiment, Compound 1 or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 550 mg twice a day for at least about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or more days, optionally with the standard of care.

In certain embodiments, a compound of Formula I, Formula II, Formula IP, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 600 mg twice a day for at least about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or more days, optionally with the standard of care. In one embodiment, Compound 2 or a pharmaceutically acceptable salt thereof is administered in a dosage form of about 600 mg twice a day for at least about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or more days, optionally with the standard of care.

In certain embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, for example Compound 1 or Compound 3 or a pharmaceutically acceptable salt thereof, including Compound 2 or Compound 4, is administered at an initial dose (or loading dose) followed by a maintenance dose, wherein the loading dose is at the discretion of the physician based on the severity of the presented disease and the size of the patient. In certain embodiments, the loading dose is about or at least 1.5 times greater, about or at least 2 times greater, about or at least 2.5 times greater, or about or at least 3 times greater than the maintenance dose. In one embodiment, the loading dose is administered once, twice, three, four, or more times before the first maintenance dose, and may be given once, twice, three, or four times a day as instructed by the physician.

In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, for example Compound 1 or Compound 3 or a pharmaceutically acceptable salt thereof, including Compound 2 or Compound 4, is administered at a daily loading dose (which can be provided in one or several dosages throughout the day) of at least about 800 mg, at least about 900 mg, at least about 1000 mg, at least about 1100 mg, at least about 1200 mg, at least about 1300 mg, or at least about 1400 mg followed by a maintenance dose of at least about 300 mg, at least about 350 mg, at least about 400 mg, at least about 450 mg, at least about 500 mg, at least about 550 mg, at least about 600 mg, at least about 650 mg, at least about 700 mg, or at least about 750 mg and the maintenance dose is taken once, twice, or three times a day. In one embodiment, the maintenance dose is taken twice a day, and optionally over 1, 2, 3, or 4 days. In one embodiment, the maintenance dose is thereafter administered 1, 2 or 3 times a day for at least about 4 days, at least about 5 days, at least about 6 days, at least about 7 days, at least about 8 days, at least about 9 days, at least about 10 days, at least about 15 days, at least about 20 days, at least about 25 days, or more.

In certain embodiments, Compound 1 or Compound 3 or a pharmaceutically acceptable salt thereof, including Compound 2 or Compound 4, is administered at a dose of at least about 300 mg, at least about 350 mg, at least about 400 mg, at least about 450 mg, at least about 500 mg, at least about 550 mg, at least about 650, or at least about 750 and the dose is taken once, twice, or three times a day. In one embodiment, Compound 1 or a pharmaceutically acceptable salt thereof, for example Compound 2, is administered at a dose of at least about 500 mg, at least about 550 mg, or at least 600 mg and the dose is taken twice daily. In one embodiment, Compound 1 or a pharmaceutically acceptable salt thereof, for example Compound 2, is administered at a loading dose of at least about 1000 mg, at least about 1100 mg, or at least about 1200 mg followed by a maintenance dose of at least about 500 mg, at least about 550 mg, or at least 600 mg twice daily. In one embodiment, the maintenance dose is administered for at least about 4, 5, 6, 7, 8, 9, 10, or more days. In one embodiment, Compound 1 is Compound 1A. In one embodiment, Compound 1 is Compound IB. In one embodiment, Compound 2 is Compound 2A. In one embodiment, Compound 2 is Compound 2B

In one embodiment, Compound 1 is administered at a dose of at about 550 mg and the dose is taken twice daily. In one embodiment, Compound 1 is Compound 1A. In one embodiment, Compound 1 is Compound IB.

In one embodiment, Compound 2 is administered at a dose of at about 600 mg and the dose is taken twice daily. In one embodiment, Compound 2 is Compound 2A. In one embodiment, Compound 2 is Compound 2B.

In certain embodiments, the method of the present invention includes administering a compound as described herein, such as Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, for example Compound 1 or Compound 2, once, twice, three, or four or more times a day as necessary to treat the infection. In one embodiment, a compound of Formula I, Formula II, Formula PI, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, for example Compound 1 or Compound 2, is administered for at least about 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, or more days, or for a length of time at the discretion of a healthcare provider. Alternatively, the compound can be administered for a time period that is appropriate to avoid infection or reduce the severity of an infection of a human or other animal at risk of becoming infected with the virus.

In one embodiment, the compound of the present invention is administered indefinitely until the risk of infection or reinfection no longer exits. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered for at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or at least 6 months or more. In certain embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered once, twice, three, or four or more times a day.

In another alternative embodiment, a method to prevent transmission is provided that includes administering an effective amount of one of the compounds described herein to a human in need thereof for a sufficient length of time prior to exposure to a high risk situation, including during travel or public events or meetings, or if the host is in a high risk group, including for example, up to 3, 5, 7, 10, 12, 14 or more days prior to a communicable situation, and then during and optionally after the potential exposure. Alternatively, the selected compound as described herein can be administered for an indefinite period in a maintenance dosage to protect a person in a high-risk environment.

The present invention also includes compounds of Formula XI and Formula XII:

Formula XI Formula XII or a pharmaceutically acceptable salt thereof wherein

R 12a and R 12b are oxygen protecting groups and at least one of R 12a and R 12b is -C(0)0Ci- ealkyl, for example -C(0)0/Bu, or -C(0)0-benzyl wherein the alkyl and benzyl group can be optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl.

In one embodiment, R 12a is -C(0)0Ci- 6 alkyl or -C(0)0-benzyl and R 12b is an oxygen protecting group which when attached to the oxygen is an ester, ether, or silyl ether moiety. In an alternative embodiment, R 12b is -C(0)0Ci- 6 alkyl or -C(0)0-benzyl and R 12a is an oxygen protecting group which when attached to the oxygen is an ester, ether, or silyl ether moiety. In one embodiment, R 12a and R 12b are both -C(0)0Ci- 6 alkyl, for example -C(0)0/Bu. In one embodiment, R 12a and R 12b are both -C(0)0-benzyl.

In one embodiment, a compound of Formula XII is Formula XIIA:

Formula XIIA In one embodiment, a compound of Formula XII is Formula XIIB:

Formula CPB

The present invention thus includes the following features:

(a) A method for the treatment of COVID-19 caused by the SARS-CoV-2 virus in a host in need thereof comprising administering an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula

VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically carrier;

(b) A method for the prevention or minimization of COVID-19 in a host in need thereof comprising administering an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula

IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically carrier;

(c) The method of (b) for the prevention or minimization of reinfection by the S ARS-CoV- 2 virus or contracting COVID-19 in a host in need thereof;

(d) The method of (a) - (c) wherein the compound is Compound 1;

(e) The method of (a) - (c) wherein the compound is Compound 1A;

(f) The method of (a) - (c) wherein the compound is Compound IB;

(g) The method of (a) - (c) wherein the compound is Compound 2;

(h) The method of (a) - (c) wherein the compound is Compound 2A;

(i) The method of (a) - (c) wherein the compound is Compound 2B;

(j) The method of (a) - (c) wherein the compound is Compound 3;

(k) The method of (a) - (c) wherein the compound is Compound 3A;

(l) The method of (a) - (c) wherein the compound is Compound 3B;

(m)The method of (a) - (c) wherein the compound is Compound 4;

(n) The method of (a) - (c) wherein the compound is Compound 4A;

(o) The method of (a) - (c) wherein the compound is Compound 4B;

(p) The method of (a) - (c) wherein the compound is of Formula Ilia;

(q) The method of (a) - (c) wherein the compound is of Formula Illb;

(r) The method of (a) - (c) wherein the compound is of Formula IIIc;

(s) The method of (a) - (c) wherein the compound is of Formula Hid; (t) The method of (a) - (c) wherein the compound is of Formula Hie;

(u) The method of (a) - (c) wherein the compound is of Formula Illf;

(v) The method of (a) - (c) wherein the compound is of Formula IVa;

(w) The method of (a) - (c) wherein the compound is of Formula IVb;

(x) The method of (a) - (c) wherein the compound is of Formula IVc;

(y) The method of (a) - (c) wherein the compound is of Formula IVd,

(z) The method of (a) - (c) wherein the compound is of Formula IVe;

(aa) The method of (a) - (c) wherein the compound is of Formula IVf;

(bb) The method of (a) - (c) wherein the compound is of Formula V;

(cc) The method of (a) - (c) wherein the compound is of Formula VI;

(dd) The method of (a) - (c) wherein the compound is of Formula VII;

(ee) The method of (a) - (c) wherein the compound is of Formula VIII, Formula IX, or Formula X;

(ff) A compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof for use to treat a COVID-19 infection caused by the SARS-CoV-2 virus in a host in need thereof, optionally in a pharmaceutically acceptable carrier;

(gg) A compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof for use to prevent or minimize (relative to without treatment) an infection caused by the SARS-CoV-2 virus in a host in need thereof, optionally in a pharmaceutically acceptable carrier;

(hh) The compound of (gg) to prevent a reinfection caused by the SARS-CoV-2 virus in a host in need thereof;

(ii) The use of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, in the manufacture of a medicament for the treatment of the 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus;

(jj) The use of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, in the manufacture of a medicament for the prevention of COVID- 19 caused by the SARS-CoV-2 virus in a host in need thereof; (kk) The use of (jj) to prevent a reinfection caused by the SARS-CoV-2 virus in a host in need thereof;

(11) Any of the above embodiments, wherein the pharmaceutically acceptable carrier is in a dosage form suitable for oral administration;

(mm) The dosage form of (11) wherein the dosage form is a solid dosage form;

(nn) The dosage form of (mm) in the form of a tablet;

(oo) The dosage form of (mm) in the form of a capsule;

(pp) The dosage form of (11) wherein the dosage form is a liquid dosage form;

(qq) The dosage form of (pp) in the form of a solution or a suspension;

(rr) Any of embodiments (a)-(kk), wherein the pharmaceutically acceptable carrier is in a dosage form suitable for intravenous administration;

(ss) Any of embodiments (a)-(kk), wherein the pharmaceutically acceptable carrier is in a dosage form suitable for parenteral administration;

(tt) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered once a day;

(uu) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered twice a day;

(vv) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered three times a day;

(ww) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered for at least one week, ten days, two weeks, three weeks, one month, at least two months, at least three months, at least four months, at least five months, or at least six months or more.

(xx) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered at least once, at least twice, or at least three times a day indefinitely until the risk of infection no longer exists;

(yy) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered at a dose of at least about 400 mg;

(zz) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered at a dose of at least about 500 mg;

(aaa) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered at a dose of at least about 550 mg;

(bbb) Any of the above embodiments, wherein a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered at a dose of at least about 600 mg;

(ccc) A method for the treatment of COVID-19 in a host in need thereof comprising administering Compound 1, wherein Compound 1 is administered at a dose of at least about 550 mg and the dose is administered twice a day;

(ddd) A method for the treatment of COVID- 19 virus in a host in need thereof comprising administering Compound 1, wherein Compound 1 is administered at a loading dose of at least about 1100 mg followed by a maintenance dose of at least about 550 mg twice a day;

(eee) Embodiment (ccc or ddd) wherein Compound 1 is Compound 1A;

(fff) Embodiment (ccc or ddd) wherein Compound 1 is Compound IB;

(ggg) A method for the treatment of COVID-19 in a host in need thereof comprising administering Compound 2, wherein Compound 2 is administered at a dose of at least about 600 mg and the dose is administered twice a day;

(hhh) A method for the treatment of COVID-19 in a host in need thereof comprising administering Compound 2, wherein Compound 2 is administered at a loading dose of at least about 1200 mg followed by a maintenance dose of at least about 600 mg twice a day;

(iii) Embodiment (ggg or hhh) wherein Compound 2 is Compound 2A; (jjj) Embodiment (ggg or hhh) wherein Compound 2 is Compound 2B;

(kkk) A compound of Formula II or a pharmaceutically acceptable salt thereof;

(111) Compound 4 or a pharmaceutically acceptable salt as described herein,

(mmm) Compound 4A and Compound 4B as described herein;

(nnn) A pharmaceutical formulation comprising an effective amount of a compound of Formula II, optionally in a pharmaceutically acceptable carrier.

(ooo) A compound of Formula XI or Formula XII;

(ppp) A compound of Formula XIIA;

(qqq) A compound of Formula XIIB;

(rrr) A compound of Formula VII;

(sss) A compound of Formula Vila;

(ttt) A compound of Formula Vllb;

(uuu) A compound of Formula Vile;

(vvv) A compound of Formula Vlld; and

(www) A pharmaceutical composition comprising an effective amount of a compound of Formula Vila, Formula Vllb, Formula Vile, or Formula Vlld.

BRIEF DESCRIPTION OF FIGURES

FIG. 1 is a graph of the concentration of triphosphate Compound 1-6 in human bronchial and nasal epithelial cells after exposure to 10 mM of Compound 1A as described in Example 7. The half-life of Compound 1-6 in bronchial cells and nasal cells was 39 hours and 38 hours, respectively. The x-axis is the time post-washout measured in hours and the y-axis is the concentration of Compound 1-6 in pmol/million cells.

FIG. 2 is a graph comparing the triphosphate Compound 1-6 levels in human bronchial epithelial cells following exposure to Compound 1A (1A), ALS-8112 (ALS), and the 4’-Me substituted prodrug isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)- 4-fluoro-3-hydroxy-2,4-dimethyltetrahydrofuran-2-yl)methoxy) (phenoxy)phosphoryl)-L-alaninate (4’-Me) as described in Example 7. The x-axis is the time post-washout measured in hours and the y-axis is the concentration of Compound 1-6 in pmol/million cells.

FIG. 3 is a graph comparing the triphosphate Compound 1-6 levels in human nasal epithelial cells following exposure to Compound 1A (1A), ALS-8112 (ALS), and 4’-Me substituted prodrug isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4-fluoro-3 -hydroxy - 2,4-dimethyltetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl )-L-alaninate (4’-Me) as described in Example 7. The x-axis is the time post-washout measured in hours and the y-axis is the concentration of Compound 1-6 in pmol/million cells. FIG. 4 is a graph of the mean plasma profile of Compound 1A in monkeys administered 30 mg/kg oral doses of Compound 2A twice a day (BID) for 3 days as described in Example 8. The x- axis is the time post-dose measured in hours and the y-axis is the plasma concentration of Compound 1A measured in ng/mL.

FIG. 5 is a graph of the mean plasma profile of metabolite Compound 1-2 in monkeys administered 30 mg/kg oral doses of Compound 2A twice a day (BID) for 3 days as described in Example 8. The x-axis is the time post-dose measured in hours and the y-axis is the plasma concentration of Compound 1-2 measured in ng/mL.

FIG. 6 is a graph of the mean plasma profile of triphosphate surrogate metabolite Compound 1-7 in monkeys administered 30 mg/kg oral doses of Compound 2A twice a day (BID) for 3 days as described in Example 8. The x-axis is the time post-dose measured in hours and the y-axis is the plasma concentration of Compound 1-7 measured in ng/mL.

FIG. 7A is a graph of the triphosphate Compound 1-6 concentration in lung, kidney, and liver tissue in monkeys following administration of 30 mg/kg oral doses of Compound 2A twice a day (BID) for 3 days as described in Example 8. The half-life in the lung, kidney, and liver was 9.4 hours, 8.0 hours, and 4.3 hours, respectively. The x-axis is the time post-dose measured in hours and the y-axis is the tissue concentration of Compound 1-6 measured in ng/g.

FIG. 7B is a graph of triphosphate Compound 1-6 concentration in lung, kidney, and liver tissue in monkeys following administration of 30 mg/kg oral doses of Compound 2A twice a day (BID) for 3 days as described in Example 8. The tissue concentration of Compound 1-6 is shown 2 hours, 12 hours, 24 hours, and 48 hours post last dose. The x-axis is time post the last dose measured in hours and the y-axis is tissue concentration measured in mM.

FIG. 8 is a graph of levels of triphosphate Compound 1-6 in hepatocytes incubated with Compound 2A as described in Example 9 and previously described in from Good, S.S. et al. 2020 PLoS ONE 15(l):e0227104. The concentration was 7 times higher in human hepatocytes than in monkeys. The x-axis is the incubation time measured in hours and the y-axis is the Compound 1-6 concentration measured in pmol/10 6 cells.

FIG. 9 is the simulation of intracellular concentrations of Compound 1-6 in human lung tissue as described in Example 10. The predicted lung concentration is based on predicted trough (Co ) steady-state plasma Compound 1-7, a plasma surrogate for intracellular triphosphate Compound 1-6, (Berliba, e. et al. 2019 Antimicrob. Agents Chemother. 63(12):e01201-19) in humans multiplied by a ratio of 1.6 (the triphosphate concentration in the lung is 1.6 times greater than in the liver at steady-state trough levels as described in Example 8). The x-axis is time measured in days and the y-axis is simulated lung Compound 1-6 concentration measured in mUI. FIG. 10 is a stimulation of intracellular concentrations of Compound 1-6 in human lung tissue using the two approaches as described in Example 10. The solid curve represents predicted lung concentrations of the active triphosphate Compound 1-6 metabolite after correcting for the Compound 1-6 lung-to-liver concentration ratio of 1.6. The dotted curve represents predicted lung concentrations of the active triphosphate Compound 1-6 metabolite after correcting for the Compound 1-6 lung-to-Compound 1-7 plasma ratio of 1.2. The horizontal line represents the EC90 of Compound 1A against SARS-CoV-2 in HAE cells in vitro (0.47 pM). The x-axis is time measured in days and the y-axis is simulated lung Compound 1-6 concentration measured in m\1 FIG. 11 is an illustration of a compound of Formula I, which can also be administered as a pharmaceutically acceptable salt.

DETAILED DESCRIPTION OF THE INVENTION

The invention disclosed herein is a method for the treatment or prevention of the 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus in a host, for example a human, in need thereof comprising administering an effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof: wherein

R 1 is selected from Ci-C 6 alkyl, C3-C6cycloalkyl, and -C(0)Ci-C 6 alkyl;

R 2 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), C3-7cycloalkyl, or aryl (including phenyl and napthyl) and in an alternative embodiment, R 2 is aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R 3 is hydrogen or Ci-ealkyl (including methyl, ethyl, propyl, and isopropyl);

R 4a and R 4b are independently selected from hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), and C3-7cycloalkyl; and

R 5 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), Ci- 6 haloalkyl, or C3-7cycloalkyl and in an alternative embodiment, R 5 is aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl. Non-limiting examples of a compound of Formula I include Compound 1 and Compound 2. In one embodiment, the compounds are administered as the S-enantiomer, such as Compound 1A In one embodiment, the compounds are administered as the R-enantiomer, such as Compound IB. In one embodiment, a compound of Formula I is Compound 2, Compound 2A, or Compound

2B.

CH 3 O A U HQ f_ · 0.5 H

'F 2 SO 4

Compound 2B

Alternative configurations of Compound 1 or a pharmaceutically acceptable salt thereof that can be used include:

Additional alternative configurations of Compound 1 or a pharmaceutically acceptable salt thereof, for example, Compound 2 that can be used include:

Non-limiting examples of a compound of Formula I include:

or a pharmaceutically acceptable salt thereof.

Additional non-limiting examples of a compound of Formula I include:

or a pharmaceutically acceptable salt thereof.

5 Additional non-limiting examples of compounds of Formula I include: or a pharmaceutically acceptable salt thereof.

The present invention also includes the use of an effective amount of a compound of Formula II to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof:

Formula II or a pharmaceutically acceptable salt thereof, wherein:

R 1 is selected from Ci-C 6 alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C 6 alkyl;

R 2 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), C3-7cycloalkyl, or aryl (including phenyl and napthyl) and in an alternative embodiment, R 2 is aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R 3 is hydrogen or Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl);

R 4a and R 4b are independently selected from hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), and C3-7cycloalkyl; and

R 5 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), Ci- 6 haloalkyl, or C3-7cycloalkyl and in an alternative embodiment, R 5 is aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl.

In one embodiment, a compound of Formula II or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered in an effective amount to a host in need thereof with COVID-19, or at risk of infection with the SARS-CoV-2 vims, i.e., as a prophylactic.

Non-limiting examples of a compound of Formula II include Compound 3 and Compound 4. In one embodiment, the compounds are administered as the S-enantiomer, such as Compound 3A and Compound 4A. In one embodiment, the compounds are administered as the R-enantiomer, such as Compound 3B or Compound 4B.

Compound 3A Compound 4A

,CH

HN 3

Compound 4B

Alternative configurations of Compound 3 or a pharmaceutically acceptable salt thereof include:

Additional alternative configurations of Compound 4 include:

Additional alternative configurations of Compound 3 or a pharmaceutically acceptable salt thereof, for example, Compound 4 that can be used include:

Non-limiting examples of a compound of Formula II include: or a pharmaceutically acceptable salt thereof.

Additional non-limiting examples of a compound of Formula II include:

or a pharmaceutically acceptable salt thereof.

Additional non-limiting examples of compounds of Formula II include:

or a pharmaceutically acceptable salt thereof. The present invention also includes the use of an effective amount of a compound of

Formula III to treat or prevent COVED- 19 disease caused by the SARS-CoV-2 virus in a host in need thereof:

Formula III or a pharmaceutically acceptable salt thereof, wherein:

R 1 is selected from Ci-C 6 alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C 6 alkyl;

R 2 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), C3-7cycloalkyl, or aryl (including phenyl and napthyl) and in an alternative embodiment, R 2 is aryl(Ci-C4alkyl)-, heteroaryl, or heteroalkyl;

R 3 is hydrogen or Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl);

R 4a and R 4b are independently selected from hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), and C3-7cycloalkyl; and R 5 is hydrogen, Ci- 6 alkyl (including methyl, ethyl, propyl, and isopropyl), Ci- 6 haloalkyl, or C3-7cycloalkyl and in an alternative embodiment, R 5 is aryl(Ci-C4alkyl)-, aryl, heteroaryl, or heteroalkyl; and

X is selected from Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH 3 , CF 2 CF , and CH 2 C1), C 2 -C alkenyl, C 2 -C4alkynyl, and Ci-C3hydroxy alkyl.

In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof is a compound of Formula Ilia: or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula Ilia, R 1 is methyl.

In one embodiment of Formula Ilia, R 1 is cyclopropyl.

In one embodiment of Formula Ilia, R 2 is phenyl.

In one embodiment of Formula Ilia, R 2 is napthyl.

In one embodiment of Formula Ilia, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula Ilia, R 5 is isopropyl.

In one embodiment of Formula Ilia, the compound is the S p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Ilia, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Ilia, the pharmaceutically acceptable salt is the hemi- sulfate salt. Non-limiting examples of a compound of Formula Ilia include:

Additional non-limiting examples of a compound of Formula Ilia include: In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof is a compound of Formula Illb:

Formula Illb or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula Illb, R 1 is methyl.

In one embodiment of Formula Illb, R 1 is cyclopropyl.

In one embodiment of Formula Illb, R 2 is phenyl.

In one embodiment of Formula Illb, R 2 is napthyl. In one embodiment of Formula Illb, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula Illb, R 5 is isopropyl.

In one embodiment of Formula Illb, the compound is the S p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Illb, the compound is the Rp-isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Illb, the pharmaceutically acceptable salt is the hemi- sulfate salt.

Non-limiting examples of a compound of Formula Illb include: W ° 2021/173713

PCT/ US2021/019468

Additional non-limiting examples of a compound of Formula Illb include:

In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof is a compound of Formula IIIc:

Formula IIIc or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IIIc, R 1 is methyl.

In one embodiment of Formula IIIc, R 1 is cyclopropyl.

In one embodiment of Formula IIIc, R 2 is phenyl.

In one embodiment of Formula IIIc, R 2 is napthyl.

In one embodiment of Formula IIIc, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula IIIc, R 5 is isopropyl.

In one embodiment of Formula IIIc, the compound is the Sp-isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IIIc, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IIIc, the pharmaceutically acceptable salt is the hemi- sulfate salt. Non-limiting examples of a compound of Formula IIIc include:

HN HN

Additional non-limiting examples of a compound of Formula IIIc include: In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof is a compound of Formula Hid:

,R 1

HN

Formula Hid or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula Hid, R 1 is methyl.

In one embodiment of Formula Hid, R 1 is cyclopropyl.

In one embodiment of Formula Hid, R 2 is phenyl.

In one embodiment of Formula Hid, R 2 is napthyl.

In one embodiment of Formula Hid, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula Hid, R 5 is isopropyl.

In one embodiment of Formula Hid, the compound is the S p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Hid, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Illd, the pharmaceutically acceptable salt is the hemi- sulfate salt.

Non-limiting examples of a compound of Formula Hid include:

5 Additional non-limiting examples of a compound of Formula Hid include:

HIM HN

In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof is a compound of Formula Hie:

Formula Hie or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula Hie, R 1 is methyl.

In one embodiment of Formula Hie, R 1 is cyclopropyl. In one embodiment of Formula Hie, R 2 is phenyl.

In one embodiment of Formula Hie, R 2 is napthyl. In one embodiment of Formula Hie, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula Hie, R 5 is isopropyl.

In one embodiment of Formula Hie, the compound is the S p -isomer and the phosphoramidate is in the L-configuration. In one embodiment of Formula Hie, the compound is the Rp-isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula Hie, the pharmaceutically acceptable salt is the hemi- sulfate salt. Non-limiting examples of a compound of Formula Hie include:

Additional non-limiting examples of a compound of Formula Hie include:

In one embodiment, the compound of Formula III to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof is a compound of Formula Illf:

Formula Illf or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula Illf, R 1 is methyl.

In one embodiment of Formula TTTf R 1 is cyclopropyl.

In one embodiment of Formula Illf, R 2 is phenyl. In one embodiment of Formula Illf, R 2 is napthyl.

In one embodiment of Formula TTTf R 4a is hydrogen and R 4b is methyl In one embodiment of Formula Illf, R 5 is isopropyl.

In one embodiment of Formula Illf, the compound is the Sp-isomer and the phosphoramidate is in the L-configuration. In one embodiment of Formula Illf, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula inf, the pharmaceutically acceptable salt is the hemi- sulfate salt. Non-limiting examples of a compound of Formula Illf include:

Additional non-limiting examples of a compound of Formula Hie include:

5 Non-limiting examples of a compound of Formula III include:

The present invention also includes the use of a compound of Formula IV to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus in a host in need thereof as described herein:

Formula IV or a pharmaceutically acceptable salt thereof, wherein:

X is selected from Ci-C3haloalkyl (including Ci-3fluoroalkyl and Ci-3chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH , CF 2 CF 3 , and CH 2 C1), C -C 4 alkenyl, C 2 -C 4 alkynyl, and Ci-C3hydroxyalkyl; and

R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease is a compound or a pharmaceutically acceptable salt thereof of Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula IVe, or Formula IVf:

Formula IVb

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease caused by the SARS-CoV-2 vims is a compound of Formula IVa: or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IVa, R 1 is methyl.

In one embodiment of Formula IVa, R 1 is cyclopropyl.

In one embodiment of Formula IVa, R 2 is phenyl.

In one embodiment of Formula IVa, R 2 is napthyl.

In one embodiment of Formula IVa, R 4a is hydrogen and R 4h is methyl.

In one embodiment of Formula IVa, R 5 is isopropyl.

In one embodiment of Formula IVa, the compound is the Sp-isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVa, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

Non-limiting examples of a compound of Formula IVa include:

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus is a compound of Formula IVb: or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IVb, R 1 is methyl.

In one embodiment of Formula IVb, R 1 is cyclopropyl.

In one embodiment of Formula IVb, R 2 is phenyl.

In one embodiment of Formula IVb, R 2 is napthyl.

In one embodiment of Formula IVb, R 4a is hydrogen and R is methyl.

In one embodiment of Formula IVb, R 5 is isopropyl.

In one embodiment of Formula IVb, the compound is the S p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVb, the compound is the Rp-isomer and the phosphoramidate is in the L-configuration. Non-limiting examples of a compound of Formula IVb include:

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus is a compound of Formula IVc:

.R 1

HN

Formula IVc or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IVc, R 1 is methyl.

In one embodiment of Formula IVc, R 1 is cyclopropyl. In one embodiment of Formula IVc, R 2 is phenyl.

In one embodiment of Formula IVc, R 2 is napthyl.

In one embodiment of Formula IVc, R 4a is hydrogen and R is methyl.

In one embodiment of Formula IVc, R 5 is isopropyl.

In one embodiment of Formula IVc, the compound is the Sp-isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVc, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVc, the pharmaceutically acceptable salt is the hemi- sulfate salt. Non-limiting examples of a compound of Formula IVc include:

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease is a compound of Formula IVd:

Formula IVd or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IVd, R 1 is methyl.

In one embodiment of Formula IVd, R 1 is cyclopropyl.

In one embodiment of Formula IVd, R 2 is phenyl.

In one embodiment of Formula IVd, R 2 is napthyl.

In one embodiment of Formula IVd, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula IVd, R 5 is isopropyl.

In one embodiment of Formula IVd, the compound is the S p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVd, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVd, the pharmaceutically acceptable salt is the hemi- sulfate salt.

Non-limiting examples of a compound of Formula IVd include:

In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease is a compound of Formula IVe: R

Formula IVe or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IVe, R 1 is methyl.

In one embodiment of Formula IVe, R 1 is cyclopropyl.

In one embodiment of Formula IVe, R 2 is phenyl.

In one embodiment of Formula IVe, R 2 is napthyl.

In one embodiment of Formula IVe, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula IVe, R 5 is isopropyl.

In one embodiment of Formula IVe, the compound is the Sp-isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVe, the compound is the Rp-isomer and the phosphoramidate is in the L-configuration. In one embodiment of Formula IVe, the pharmaceutically acceptable salt is the hemi- sulfate salt.

Non-limiting examples of a compound of Formula IVe include: In one embodiment, the compound of Formula IV to treat or prevent COVID-19 disease caused by the SARS-CoV-2 virus is a compound of Formula IVf: Formula IVf or a pharmaceutically acceptable salt thereof.

In one embodiment of Formula IVf, R 1 is methyl.

In one embodiment of Formula IVf, R 1 is cyclopropyl.

In one embodiment of Formula IVf, R 2 is phenyl. In one embodiment of Formula IVf, R 2 is napthyl.

In one embodiment of Formula IVf, R 4a is hydrogen and R 4b is methyl.

In one embodiment of Formula IVf, R 5 is isopropyl.

In one embodiment of Formula IVf, the compound is the Sp-isomer and the phosphoramidate is in the L-configuration. In one embodiment of Formula IVf, the compound is the R p -isomer and the phosphoramidate is in the L-configuration.

In one embodiment of Formula IVf, the pharmaceutically acceptable salt is the hemi- sulfate salt.

The present invention also includes the use of a compound of Formula V to treat or prevent COVET- 19 disease caused by the SARS-CoV-2 vims as described herein:

Formula V or a pharmaceutically acceptable salt thereof, wherein:

Y and Y’ are independently selected from Cl and F; and

R 1 , R 2 , R 3 , R 4a , R 4b , and R 5 are as defined herein.

In one embodiment of Formula IV, Y’ is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is F, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula IV, Y’ is F, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-G,alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is Cl, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl. In one embodiment of Formula IY, Y’ is F, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula IV, Y’ is F, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl. In one embodiment of Formula IV, Y’ is F, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula IV, Y’ is F, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl. Non-limiting examples of a compound of Formula V include

Additional non -limiting examples of Formula V include

The present invention also includes the use of a compound of Formula VI to treat or prevent COVED- 19 in a host in need thereof as described herein: wherein

R 6 is selected from hydrogen, -C(0)R 6A , -C(0)0R 6A , Ci- 6 alkyl, and -CH 2 -0-R 6A and in an alternative embodiment, -C(0)NR 6B R 6C ;

R 6A is selected from hydrogen, Ci^alkyl, Ci-G, haloalkyl (for example, -CHC , -CCI3, -CH2CI, -CF3, -CHF2, -CH2F), aryl, and aryl(Ci- 6 alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl and in an alternative embodiment, R 6A is selected from Ci-2oalkyl and C2-2oalkenyl;

R 6B and R 6C are independently selected from hydrogen, Ci-2oalkyl, C2-2oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl wherein the Ci-2oalkyl, C2-2oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl can optionally be substituted with at least one substituent selected from alkoxy (including but not limited to methoxy and ethoxy), hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R 7 is NH 2 , H, or -NR 8 R 9 ;

R 8 and R 9 are independently selected from hydrogen, Ci- 6 alkyl, -C(0)R 6A , and -C(0)0R 6A ; Y is selected from F and Cl;

Z is selected from methyl, Ci-C 3 haloalkyl (including Ci- 3 fluoroalkyl and Ci- 3 chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH2CF3, CH2CHF2, CH 2 CH 2 F, CF2CH3, CF2CF3, and CH2CI), C - C 4 alkenyl, C 2 -C 4 alkynyl, Ci-C 3 hydroxyalkyl, and halogen (including Cl and F), and in an alternative embodiment Z is Ci- 4 alkyl; and R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

Non-limiting examples of R 6 include In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NFb

In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A

In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A . In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl. In one embodiment of Formula VI, Z is CFb, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is CFh, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C6alkyl, and R 7 is NFh

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C6alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CF3, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is HC(0)R 6A

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, and R 5 is Ci-C6alkyl.

In one embodiment of Formula VI, Z is CF 3 , Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is Fh.

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is R 8 R 9 .

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is HC(0)R 6A .

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl. In one embodiment of Formula VI, Z is Cl, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-Cr,alkyl.

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is M

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is HC(0)0R 6A .

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CFhF, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is Mh.

In one embodiment of Formula VI, Z is CFhCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CFhCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CFhCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A .

In one embodiment of Formula VI, Z is CFhCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-G,alkyl.

In one embodiment of Formula VI, Z is CHCH2, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CHCH2, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl. In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cealkyl, and R 7 is NFh.

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A .

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CCH, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NΉ2.

In one embodiment of Formula VI, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A .

In one embodiment of Formula VI, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A

In one embodiment of Formula VI, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is F, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is F, Y is F, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is C¾, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NH2. In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cealkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A

In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CFb, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CF 3 , Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C6alkyl, and R 7 is NFb.

In one embodiment of Formula VI, Z is CF 3 , Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C6alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CF 3 , Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C6alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CF 3 , Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, R 5 is Ci-C6alkyl, and R 7 is NHC(0)R 6A

In one embodiment of Formula VI, Z is CF3, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CF3, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CF3, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C 4 alkyl, and R 5 is Ci-C6alkyl.

In one embodiment of Formula VI, Z is CF 3 , Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-Cbalkyl.

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NFb.

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H. In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Galkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is Cl, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is C¾F, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is Mh.

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A .

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is G-Galkyl.

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CFhF, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CHCFh, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is Nth.

In one embodiment of Formula VI, Z is CFhCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CFhCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Galkyl, and R 7 is NR 8 R 9 . In one embodiment of Formula VI, Z is CFhCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cealkyl, and R 7 is NHC(0)R 6A .

In one embodiment of Formula VI, Z is CFhCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CHCH2, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CHCH2, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is CHCH2, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NH2.

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is H.

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NR 8 R 9 .

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)R 6A

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, and R 7 is NHC(0)0R 6A .

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, and R 5 is Ci-Cealkyl.

In one embodiment of Formula VI, Z is CCH, Y is Cl, R 1 is cyclopropyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is methyl, and R 5 is Ci-C 6 alkyl.

Non-limiting examples of a compound of Formula VI include Additional non-limiting examples of a compound of Formula VI include:

The present invention also includes the use of a compound of Formula VII to treat or prevent COVID-19 in a host in need thereof as described herein: wherein

B is selected from

R 6 is selected from hydrogen, -C(0)R 6A , -C(0)0R 6A , Ci^alkyl, and -CH 2 -0-R 6A and in an alternative embodiment, -C(0)NR 6B R 6C ;

R 6A is selected from hydrogen, Ci^alkyl, Ci-C 6 haloalkyl (for example, -CHCh, -CCI3, -CH2CI, -CF3, -CHF2, -CH2F), aryl, and aryl(Ci- 6 alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl and in an alternative embodiment, R 6A is selected from Ci-2oalkyl and C2-2oalkenyl; R 6B and R 6C are independently selected from hydrogen, Ci- 2 oalkyl, C 2-2 oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl wherein the Ci- 2 oalkyl, C 2-2 oalkenyl, aryl, aryl(Ci- 6 alkyl)-, heteroaryl, and heteroarylalkyl can optionally be substituted with at least one substituent selected from alkoxy (including but not limited to methoxy and ethoxy), hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R 7 is NH 2 , H, or -NR 8 R 9 ,

R 8 and R 9 are independently selected from hydrogen, Ci- 6 alkyl, -C(0)R 6A , and -C(0)0R 6A ;

Y is selected from F and Cl;

Z is selected from methyl, Ci-C 3 haloalkyl (including Ci- 3 fluoroalkyl and Ci- 3 chloroalkyl, such as CH 2 F, CHF 2 , CF 3 , CH 2 CF 3 , CH 2 CHF 2 , CH 2 CH 2 F, CF 2 CH 3 , CF 2 CF 3 , and CH 2 C1), C - C 4 alkenyl, C 2 -C 4 alkynyl, Ci-C 3 hydroxyalkyl, and halogen (including Cl and F), and in an alternative embodiment Z is Ciualkyl;

R 40 is selected from H, Ci- 3 alkoxy, Ci alkyl, N 3 , CN, and halogen (including Cl and F);

R 41 is selected from H, Ci- 3 alkyl (including methyl) and halogen (including Cl, F, and Br); R 42a and R 42b are selected from Ci. 3 alkyl (including methyl), NFh, H, -NR 8 R 9 , and -

C(0)NR 8 R 9 ; and

R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

Non-limiting examples of B include: In one embodiment, the invention also includes a compound of Formula Vila, Formula Vllb, Formula Vile, and Formula Vlld: In one embodiment of Formula VII, Z is CH 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl. R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NH2.

In one embodiment of Formula Vila, Z is CH 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is Ci-6alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NFh. In a further embodiment, R 41 is methyl and R 42a is NFh.

In one embodiment of Formula Vllb, Z is CFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci-ealkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci-6alkyl or H. In a further embodiment, R 41 is Ci-oalkyl or H, R 42a is -NHCH3, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFh, and R 42b is NH 2 .

In one embodiment of Formula Vile, Z is CFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is NFh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCFh, and R 42b is H. In a further embodiment, R 41 is Ci-6alkyl or H, R 42a is NH2, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Nth.

In one embodiment of Formula Vlld, Z is Cth, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH3. In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCth.

In one embodiment of Formula VII, Z is CH 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cealkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is Nth. In a further embodiment, R 1 is methyl and R 7 is NH 2 .

In one embodiment of Formula Vila, Z is CH3, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NFh. In a further embodiment, R 41 is methyl and R 42a is Nth. In one embodiment of Formula Vllb, Z is Cth, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci-ealkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is Nth, and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci-ealkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFh, and R 42b is NH 2.

In one embodiment of Formula Vile, Z is CH 3 , Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is C 1 -C h alky 1, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is NFh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NllCIIi, and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vlld, Z is CFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3. In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3 .

In one embodiment of Formula VII, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NFh. In one embodiment of Formula Vila, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl. R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci-6alkyl and R 42a is Mh. In a further embodiment, R 41 is methyl and R 42a is Fh.

In one embodiment of Formula Vllb, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen,

R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Coalkyl, R 6 is hydrogen, R 40 is hydrogen; and B is In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment,

R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci-6alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFh, and R 42b is B. In one embodiment of Formula Vile, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen,

R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is H, R 42a is NH 2 , and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCH 3 , and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is Mh, and R 42b is Ci-6alkyl or H. In a further embodiment, R 41 is Ci-6alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci. 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 . In one embodiment of Formula Vlld, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl. R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is

-C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCFb.

In one embodiment of Formula VII, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NFh.

In one embodiment of Formula Vila, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NFh. In a further embodiment, R 41 is methyl and R 42a is NFh.

In one embodiment of Formula Vllb, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vile, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 41 is H, R 42a is Fh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is - HCH 3 , and R 42¾ is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH 2 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is C 1 -r,al kyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci. 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vlld, Z is Cl, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 41 is halogen and R 42a is -C(0) R 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is

-C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3.

In one embodiment of Formula VII, Z is CH2F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NH2.

In one embodiment of Formula Vila, Z is CH2F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NFh. In a further embodiment, R 41 is methyl and R 42a is NFh.

In one embodiment of Formula Vllb, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cr, alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH 2 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci. 6 alkyl or H, R 42a is -NHCFb, and R 42b is NH 2 .

In one embodiment of Formula Vile, Z is CH2F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is NFh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCH 3 , and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci. 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vlld, Z is CFhF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCFh. In one embodiment of Formula VII, Z is CFBF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Gal kyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NH2. In a further embodiment, R 1 is methyl and R 7 is NH2.

In one embodiment of Formula Vila, Z is CH2F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is G-Galkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is Ci-6alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is MB. In a further embodiment, R 41 is methyl and R 42a is MB.

In one embodiment of Formula Vllb, Z is CFBF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci-ealkyl. In a further embodiment,

In one embodiment of Formula Vile, Z is CFBF, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is G-Galkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is d2, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -MICH3, and R 42b is H. In a further embodiment, R 41 is Ci-6alkyl or H, R 42a is Mir, and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH3, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH3, and R 42b is NH2.

In one embodiment of Formula Vlld, Z is CH2F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH3. In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0) HCH 3 . In one embodiment of Formula VII, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NH2. In one embodiment of Formula Vila, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NH2. In a further embodiment, R 41 is methyl and R 42a is NH2.

In one embodiment of Formula Vllb, Z is CHCH2, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cr, alkyl, R 6 is hydrogen, R 40 is hydrogen; and In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH 2 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vile, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is Fh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is - HCH 3 , and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH 2 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci^alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vlld, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is halogen and R 42a is -C(0) R 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3.

In one embodiment of Formula VII, Z is CHCH2, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NH2. In a further embodiment, R 1 is methyl and R 7 is NH2. In one embodiment of Formula Vila, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is C i -C h alky 1, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci-6alkyl and R 42a is MB. In a further embodiment, R 41 is methyl and R 42a is IB.

In one embodiment of Formula Vllb, Z is CHCFh, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cr, alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment,

In one embodiment of Formula Vile, Z is CHCH2, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is MB, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCH 3 , and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is MB, and R 42b is Ci-6alkyl or H. In a further embodiment, R 41 is Ci-6alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci. 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 . In one embodiment of Formula Vlld, Z is CHCH2, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is C 1 -C h alky 1, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 4i is halogen and R 42a is -C(0)NR 8 R y . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3 .

In one embodiment of Formula VII, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is MB. In a further embodiment, R 1 is methyl and R 7 is NFh.

In one embodiment of Formula Vila, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NH2. In a further embodiment, R 41 is methyl and R 42a is NH2.

In one embodiment of Formula Vllb, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cr, alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH2, and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci-ealkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is - HCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vile, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is H, R 42a is N¾, and R 42b is H. In a further embodiment, R 41 is H, R 42a is - HCH 3 , and R 42¾ is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH 2 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is C 1 -r,al kyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci. 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is NH 2 .

In one embodiment of Formula Vlld, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and

In a further embodiment, R 41 is halogen and R 42a is -C(0) R 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3.

In one embodiment of Formula VII, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coal kyl, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NH2. In a further embodiment, R 1 is methyl and R 7 is NH2.

In one embodiment of Formula Vila, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coal kyl, R 6 is hydrogen, R 40 is cyano or nitro; In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NH 2 . In a further embodiment, R 41 is methyl and R 42a is NH 2.

In one embodiment of Formula Vllb, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is C 1 -Chalky 1, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH2, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is

-NHCH 3 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH3, and R 42b is NH 2

In one embodiment of Formula Vile, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is C 1 -Chalky 1, R 6 is hydrogen, R 40 is cyano or nitro;

In a further embodiment, R 41 is H, R 42a is NH 2 , and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCH 3 , and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH3, and R 42b is NH 2.

In one embodiment of Formula Vlld, Z is CCH, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is C 1 -Chalky 1, R 6 is hydrogen, R 40 is cyano or nitro; In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3.

In one embodiment of Formula VII, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl. R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NFh.

In one embodiment of Formula Vila, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is Ci-6alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NFh. In a further embodiment, R 41 is methyl and R 42a is NFh.

In one embodiment of Formula Vllb, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen,

R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci-ealkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is

-NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCth, and R 42b is NFh. In one embodiment of Formula Vile, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci -Coalkyl. R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is H, R 42a is NFh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCFh, and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42¾ is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFh, and R 42b is Ci-6alkyl or H. In a further embodiment, R 41 is Ci-6alkyl or H, R 42a is -NHCH3, and R 42b is NH 2 . In one embodiment of Formula Vlld, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen,

R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Coalkyl, R 6 is hydrogen, R 40 is hydrogen; and B is

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 In a further embodiment, R 41 is halogen and R 42a is -0(0)NHO¾. In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -C(0)NHCH 3 .

In one embodiment of Formula VII, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 1 is Ci- 6 alkyl and R 7 is NFh. In a further embodiment, R 1 is methyl and R 7 is NFh.

In one embodiment of Formula Vila, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C ,alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is H. In a further embodiment, R 41 is methyl and R 42a is H. In a further embodiment, R 41 is Ci- 6 alkyl and R 42a is NH 2 . In a further embodiment, R 41 is methyl and R 42a is NFh.

In one embodiment of Formula Vllb, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C ,alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 41 is H, R 42a is H, and R 42b is Ci- 6 alkyl. In a further embodiment, R 41 is H, R 42a is H, and R 42b is methyl. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NH2, and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH 3 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH3, and R 42b is NH 2

In one embodiment of Formula YIIc, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-C 6 alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 41 is H, R 42a is NFh, and R 42b is H. In a further embodiment, R 41 is H, R 42a is -NHCFb, and R 42b is H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is NFh, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NR 8 R 9 , and R 42b is Ci. 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCFb, and R 42b is Ci- 6 alkyl or H. In a further embodiment, R 41 is Ci- 6 alkyl or H, R 42a is -NHCH3, and R 42b is NH 2

In one embodiment of Formula Vlld, Z is F, Y is F, R 1 is methyl, R 2 is aryl, R 3 is hydrogen, R 4a is hydrogen, R 4a is Ci-C4alkyl, R 5 is Ci-Cr, alkyl, R 6 is hydrogen, R 40 is cyano or nitro; and B is

In a further embodiment, R 41 is halogen and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is halogen and R 42a is -C(0)NHCH 3 . In a further embodiment, R 41 is Br and R 42a is -C(0)NR 8 R 9 . In a further embodiment, R 41 is Br and R 42a is -0(0)NHO¾.

Non-limiting examples of compounds of Formula VII include:

The present invention also includes the use of a compound of Formula VIII, Formula IX, or Formula X wherein R 10 is a monophosphate, a diphosphate, a triphosphate, or R 10A wherein R 10A is a stabilized phosphate prodrug that metabolizes in vivo to a monophosphate, diphosphate, or triphosphate to treat or prevent COVID-19 disease in a host in need thereof as described herein:

Formula X wherein R 10 is selected from

R 10A is a stabilized phosphate prodrug that metabolizes in vivo to a monophosphate, diphosphate, or triphosphate;

R 11 is selected from hydrogen and R 1 ; and

R 1 is selected from Ci-C 6 alkyl, C3-C6Cycloalkyl, and -C(0)Ci-C 6 alkyl;

The present invention also includes compounds of Formula XI and Formula CP:

Formula XI Formula XII or a pharmaceutically acceptable salt thereof; wherein

R 12a and R 12b are oxygen protecting groups and at least one of R 12a and R 12b is -C(0)0Ci- 6 alkyl, for example -C(0)0/Bu, or -C(0)0-benzyl wherein the alkyl and benzyl group can be optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl.

In one embodiment, R 12a is -C(0)OCi- 6 alkyl or -C(0)0-benzyl and R 12¾ is an oxygen protecting group which when attached to the oxygen is an ester, ether, or silyl ether moiety. In an alternative embodiment, R 12b is -C(0)OCi- 6 alkyl or -C(0)0-benzyl and R 12a is an oxygen protecting group which when attached to the oxygen is an ester, ether, or silyl ether moiety. In one embodiment, R 12a and R 12¾ are both -C(0)OCi- 6 alkyl, for example -C(0)0/Bu. In one embodiment, R 12a and R 12b are both -C(0)0-benzyl.

In one embodiment, a compound of Formula XII is Formula XIIA:

Formula XII A

In one embodiment, a compound of Formula XII is Formula XIIB: Formula XIIB

For example, the protecting group that when attached to the oxygen can be an ester moiety, for example benzoate acetate. In one embodiment, the oxygen protecting group that when attached to the oxygen is a silyl ether moiety (for example (trimethyl silyl (TMS), triisopropyl silyl (TIPS), tert-butyldimethylsilyl (TBDMS or TBS) or tert-butyldiphenylsilyl (TBDPS). In one embodiment, the oxygen protecting group that when attached to the oxygen is an ether moiety, for example methyl ether, methoxymethyl ether, or benzyl ether. These protecting groups can be installed according to one of the procedures described in Theodora W. Green, Protective Groups in Organic Synthesis , Third Edition, John Wiley & Sons (1999), which is incorporated by reference, for the protection of hydroxyls. For example, when the oxygen protecting group which when attached to the oxygen is an ester moiety, the compound of Formula XI or Formula XII can be prepared according to the conditions described in the text on page 149-178 and when the oxygen protecting group is a silyl ether moiety when attached to the oxygen, the compound of Formula XI or Formula XII can be prepared according to the conditions described in the text on page 113-147. In one embodiment, the protecting group is a tert-butyldimethylsilyl (TBS) group. The TBS group is selectively installed on the primary alcohol over the secondary alcohol using the conditions described in the text on page 128 and in Ogilvie et al. Can. J Chem. 1979, 57, 2230. These conditions include the use of TBSC1, DMAP, and M¾ in DMF at 25 °C.

Non-limiting examples of additional protecting groups which when attached to the oxygen also include bromobenzoate, /?-methoxybenzyloxymethyl ether (MPBM), o-nitrobenzyloxymethyl ether (NBOM), / nitrobenzyloxymethyl ether, /-butoxymethyl ether, 2,2,2-trichloroethoxymethyl ether, 3-bromotetrahydropyranyl ether, tetrahydropyranyl ether, tetrahydrothiopyranyl ether, 1- m ethoxy cyclohexyl ether, l,4-dioxan-2-yl ether, tetrahydrofuranyl ether, tetrahydrothiofuranyl ether, a substituted phenyl ether, 2-picolyl ether, 4-picolyl ether, l,3-benzodithiolan-2-yl ether, /;- chlorophenoxyacetate ester, 3-phenylpropionate ester, /;-pheny1 benzoate ester, alkyl /7-riitrophenyl carbonyl, alkyl benzyl carbonyl, alkyl / methoxybenzyl carbonyl, alkyl o-nitrobenzyl carbonyl, and alkyl / nitrobenzyl carbonyl.

Non-limiting examples of R 12a and R 12b include: Non-limiting examples of a compound of Formula XI and XII include:

In some embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula IX, Formula X, or Formula XIII, for example Compound 1A, Compound IB, Compound 2A, Compound 2B, Compound 3A, Compound 3B, Compound 4A, or Compound 4B is used in a form at least 90% free of the opposite phosphorus enantiomer, and can be at least 98%, 99% or even 100% free of the opposite phosphorus enantiomer. Compound 1 (Isopropyl((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H -purin-9- yl)-4-fluoro-3 -hydroxy -4-methyltetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L - alaninate) was previously described in U S. Patent Nos. 9,828,410; 10,000,523; 10,005,811; 10,239,911; 10,815,266; 10,870,672; 10,870,673; 10,875,885; and, 10,874,687; U.S. applications US 2019-0201433 and US 2020-0222442; and, PCT Applications WO 2016/144918; WO 2018/048937; WO 2019/200005; and, WO 2020/117966 assigned to Atea Pharmaceuticals. The synthesis of Compound 1 is described in Example 1 below.

Compound 2 was previously disclosed in U.S. Patent No. 10,519,186; 10,906,928; 10,894,804; and, 10,874,687 and PCT Applications WO 2018/144640; WO 2019/200005, and, WO 2020/117966 assigned to Atea Pharmaceuticals. Compound 2A has demonstrated potent in vitro activity against clinical isolates of hepatitis C virus (HCV) by inhibiting the RNA-dependent RNA polymerase (RdRp) (Good, S. S. et al. PLoS ONE 15(1), e0227104 (2020)). Compound 2A has been evaluated in a Phase lb study (Berliba, E. et al. Antimicrob. Agents Chemther. 63, eO 11201-19 (2020)) and a Phase 2 clinical trial (Mungar, Q. et al. EASL abstract (2020)). In the latter study, Compound 2A was safe and well-tolerated for up to 12 weeks in HCV-infected subjects and achieved a high rate of efficacy.

The synthesis of Compound 2 (the hemi-sulfate salt of isopropyl((S)-(((2R,3R,4R,5R)-5-(2- amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-me thyltetrahydrofuran-2- yl)methoxy)(phenoxy)phosphoryl)-L-alaninate) is described in Example 1 below.

In one embodiment Compound 2 is provided in a pharmaceutically acceptable composition or solid dosage form thereof.

A non-limiting illustrative process for the preparation of Compound 2 includes

(i) a first step of dissolving Compound 1 in an organic solvent, for example, acetone, ethyl acetate, methanol, acetonitrile, or ether, or the like, in a flask or container;

(ii) charging a second flask or container with a second organic solvent, which may be the same as or different from the organic solvent in step (i), optionally cooling the second solvent to 0-10 degrees C, and adding dropwise H2SO4 to the second organic solvent to create a TkSCri/organic solvent mixture; and wherein the solvent for example may be methanol;

(iii) adding dropwise the fhSCE/solvent mixture at a molar ratio of 0.5/1.0 from step (ii) to the solution of Compound 1 of step (i) at ambient or slightly increased or decreased temperature (for example 23-35 degrees C);

(iv) stirring the reaction of step (iii) until precipitate of Compound 2 is formed, for example at ambient or slightly increased or decreased temperature;

(v) optionally filtering the resulting precipitate from step (iv) and washing with an organic solvent; and (vi) optionally drying the resulting Compound 2 in a vacuum, optionally at elevated a temperature, for example, 55, 56, 57, 58, 59, or 60 °C.

In one embodiment, the solvent of step (iii) is an alcohol, for example methanol, ethanol, or isopropyl alcohol. In one embodiment, the solvent of step (iii) is an alkyl ester, for example ethyl acetate.

Scheme 1 provides the metabolic pathway of a compound of Formula I, which involves the initial de-esterification of the phosphoramidate (Compound 1) to form metabolite 1-1, which spontaneously decomposes to metabolite 1-2. Metabolite 1-2 is next converted to the N 6 -methyl- 2, 6-diaminopurine-5’ -monophosphate derivative (metabolite 1-3), which is in turn metabolized to the free 5’-hydroxyl-N 6 -methyl-2,6-diaminopurine nucleoside (metabolite 1-8) and ((2//,3//,4//,5//)-5-(2-amino-6-oxo-l ,6-dihydro-9H-purin-9-yl)-4-fluoro-3-hydroxy-4- methyltetrahydrofuran-2-yl)methyl dihydrogen phosphate as the 5’ -monophosphate (metabolite 1- 4). Metabolite 1-4 is anabolized to the corresponding diphosphate (metabolite 1-5) and then the active triphosphate derivative (metabolite 1-6). The 5’ -triphosphate can be further metabolized to generate 2-amino-9-((2i?,3i?,4f?,5i?)-3-fluoro-4-hydroxy-5-(hydroxyme thyl)-3- methyltetrahydrofuran-2-yl)-l,9-dihydro-6H-purin-6-one (1-7). Metabolite 1-7 is measurable in plasma and is therefore a surrogate for the active triphosphate (1-6), which is not measurable in plasma.

Definitions

A “patient” or “host” or “subject” is a human or non-human animal in need of treatment or prevention of COVED- 19 caused by the SARS-CoV-2 virus. Typically, the host is a human. A “patient” or “host” or “subject” also refers to, for example, a mammal, primate (e.g., human), cow, sheep, goat, horse, dog, cat, rabbit, rat, mice, bird, bat and the like.

The term “prophylactic” or “preventative” when used refers to the administration of an active compound to prevent, reduce the likelihood of an occurrence or a reoccurrence of COVID- 19, or to minimize a new infection relative to infection that would occur without such treatment.

The present invention includes both treatment and prophylactic or preventative therapies. In one embodiment, the active compound is administered to a host who has been exposed to and is thus at risk of contracting COVID-19. In another alternative embodiment, a method to prevent transmission is provided that includes administering an effective amount of one of the compounds described herein to humans for a sufficient length of time prior to exposure to crowds that can be infected, including during travel or public events or meetings, including for example, up to 3, 5, 7, 10, 12, 14 or more days prior to a communicable situation.

The terms "coadminister,” "coadministration," or “in combination” are used to describe the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, according to the present invention in combination with at least one other antiviral active agent. The timing of the coadministration is best determined by the medical specialist treating the patient. It is sometimes desired that the agents be administered at the same time. Alternatively, the drugs selected for combination therapy may be administered at different times to the patient. Of course, when more than one viral or other infection or other condition is present, the present compounds may be combined with other agents to treat that other infection or condition as required.

A “pharmaceutically acceptable salt” is a derivative of the disclosed compound in which the parent compound is modified to an inorganic and organic, acid or base addition salt thereof without undue toxicity. The salts of the present compounds can be synthesized from the parent compound with a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are typical, where practicable. Salts of the present compounds may optionally be provided in the form of a solvate.

Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional salts and the quaternary ammonium salts of the parent compound formed, for example, from inorganic or organic acids that are not unduly toxic. For example, acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC-(CH2)n-COOH where n is 0-4, and the like, or using a different acid that produces the same counterion. Lists of additional suitable salts may be found, e.g., in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., p. 1418 (1985).

The compound can be delivered in any molar ratio of salt that delivers the desired result. For example, the compound can be provided with less than a molar equivalent of a counter ion, such as in the form of a hemi-sulfate salt. Alternatively, the compound can be provided with more than a molar equivalent of counter ion, such as in the form of a di-sulfate salt. Non-limiting examples of molar ratios of the compound to the counter ion include 1 : 0.25, 1 : 0.5, 1 : 1, and 1 : 2

“Alkyl” is a straight chain or branched saturated aliphatic hydrocarbon group. In certain embodiments, the alkyl is C1-C2, C1-C3, C1-C4, C1-C5, or C1-C6 (i.e., the alkyl chain can be 1, 2, 3, 4, 5, or 6 carbons in length) . The specified ranges as used herein indicate an alkyl group with length of each member of the range described as an independent species. For example, C1-C6 alkyl as used herein indicates an alkyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species and Ci-C4alkyl as used herein indicates an alkyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, e/V-pentyl, neopentyl, n-hexyl, 2-methylpentane, 3-methylpentane, 2,2-dimethylbutane and 2,3-dimethylbutane.

“Cycloalkyl” is a saturated mono-cycle hydrocarbon ring system. Non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.

"Alkenyl" refers to a non-aromatic hydrocarbon group which contains at least one double bond between adjacent carbon atoms and a similar structure to an alkyl group as otherwise described herein. For example, an alkenyl group can have to 4 carbon atoms (i.e., C 2 -C 4 alkenyl). Examples of suitable alkenyl groups include, but are not limited to, ethenyl or vinyl (-CH=CH 2 ), allyl (- CH 2 CH=CH 2 ), 1 -butenyl (-C=CH-CH 2 CH 3 ) and 2-butenyl (-CH 2 CH=CHCH 2 ).

The term "alkynyl" refers to a non-aromatic hydrocarbon group containing at least one triple bond between adjacent carbon atoms and a similar structure to an alkyl group as otherwise described herein. For example, an alkynyl group can have 2 to 4 carbon atoms (i.e., C2-C4 alkynyl). Examples of alkynyl groups include, but are not limited to ethynyl and propargyl.

"Aryl" indicates aromatic groups containing only carbon in the aromatic ring or rings. In one embodiment, the aryl groups contain 1 to 3 separate or fused rings and is 6 to about 14 or 18 ring atoms, without heteroatoms as ring members. Aryl groups include, for example, phenyl and naphthyl, including 1 -naphthyl and 2-naphthyl. In one embodiment, aryl groups are pendant. An example of a pendant ring is a phenyl group substituted with a phenyl group. In one embodiment, the aryl group is optionally substituted as described above. In one embodiment, aryl groups include, for example, dihydroindole, dihydrobenzofuran, isoindoline-l-one and indolin-2-one.

“Aryl(alkyl)-” is an alkyl group as described herein substituted with an aryl group as described herein. For example, aryl(CH2)- is benzyl. Examples of aryl(alkyl)- include benzyl, 2- phenyl (alkyl), 3-phenyl(alkyl), and napthyl(alkyl). “Heteroaryl” refers to a stable monocyclic, bicyclic, or multicyclic aromatic ring which contains from 1 to 3, or in some embodiments from 1, 2, or 3 heteroatoms selected from N, O, S, B, and P (and typically selected from N, O, and S) with remaining ring atoms being carbon, or a stable bicyclic or tricyclic system containing at least one 5, 6, or 7 membered aromatic ring which contains from 1 to 3, or in some embodiments from 1 to 2, heteroatoms selected from N, O, S, B or P with remaining ring atoms being carbon. In one embodiment, the only heteroatom is nitrogen. In one embodiment, the only heteroatom is oxygen. In one embodiment, the only heteroatom is sulfur. Monocyclic heteroaryl groups typically have from 5 or 6 ring atoms. When the total number of S and O atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to one another. Examples of heteroaryl groups include, but are not limited to, pyridinyl (including, for example, 2- hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4- hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, furyl, thienyl, isoxazolyl, thiazolyl, oxadiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, tetrahydrofuranyl, and furopyridinyl.

The term "heteroalkyl" refers to an alkyl, alkenyl, alkynyl, or haloalkyl moiety as defined herein wherein a CEE group is either replaced by a heteroatom or a carbon atom is substituted with a heteroatom for example, an amine, carbonyl, carboxy, oxo, thio, phosphate, phosphonate, nitrogen, phosphorus, silicon, or boron. In one embodiment, the only heteroatom is nitrogen. In one embodiment, the only heteroatom is oxygen. In one embodiment, the only heteroatom is sulfur. In one embodiment, "heteroalkyl" is used to indicate a heteroaliphatic group (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-6 carbon atoms.

The term phosphoramidate is used throughout the specification to describe a moiety at the 5’ position of the furanose ring of the nucleoside and forms a prodrug form of the nucleoside compound, wherein the phosphorus atom is linked through a 5’-0- bond and wherein the phosphorus is also covalently bound to at least one nitrogen, forming a P-N bond. In some embodiments, the phosphorus is covalently linked to the amino moiety of a natural or synthetic amino acid (which may be in the form of an ester). Phosphoramidate groups for use in the present invention include, for example, those of the structures:

Other phosphorami dates for use in the present invention include those of the structure: wherein:

R P1 is an optionally substituted linear, branched, or cyclic alkyl group, or an optionally substituted aryl, heteroaryl or heterocyclic group or a linked combination thereof; and R P2 is a -NR N1 R N2 group or a B’ group; wherein: R N1 and R N2 are each independently H, Ci-salkyl, (C 3 -C 7C ycloalkyl)Co-C 4 alkyl-, (aryl)Co- C 4 alkyl-, (C 3 -Csheterocyclo)Co-C 4 alkyl-, or (heteroaryl)Co-C 4 alky-; which may be optionally substituted; or

R N1 and R N2 along with the nitrogen atom to which that are attached, join to form a 3 to 7 membered heterocyclic ring; group; wherein:

R 13 is hydrogen, (Ci-Cg)alkyl, (C 2 -Cs)alkenyl, (C 2 -Cs)alkynyl, (C 3 -C 8 cycloalkyl)Co-

C 4 alkyl-, (aryl)Co-C 4 alkyl-, (C 3 -C 6 heterocyclo)Co-C 4 alkyl-, (heteroaryl)Co-C 4 alky-, or the sidechain of an amino acid, for example a sidechain of an amino acid (as otherwise described herein) often selected from the group consisting of alanine, b-alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine (often R 13 is hydrogen, methyl, isopropyl, or isobutyl);

R 14 is hydrogen, (Ci-Cs)alkyl, (C 2 -Cs)alkenyl, (C 2 -Cs)alkynyl, (C 3 -C 8C ydoalkyl)Co- C 4 alkyl-, (aryl)Co-C 4 alkyl-, (C 3 -C 6 heterocyclo)Co-C 4 alkyl-, (heteroaryl)Co-C 4 alky-, or the sidechain of an amino acid, for example a sidechain of an amino acid (as otherwise described herein) often selected from the group consisting of alanine, b-alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine (often R 14 is hydrogen, methyl, isopropyl, or isobutyl);

R 15 is hydrogen or Ci-C 3 alkyl; or

R 13 and R 14 can form a (C 3 -C7)cycloalkyl or (C3-C7)heterocyclic group; or

R 13 and R 14 or R 16 can form (C 3 -C 6 )heterocyclic group; and

R 16 is hydrogen, (Ci-Cs)alkyl, (C 3 -C 6 )alkenyl, (C 3 -C 6 )alkynyl, (C 3 -C 8 cycloalkyl)Co-C 4 alkyl, (aryl)Co-C 4 alkyl-, (C 3 -C 6 heterocyclo)Co-C 4 alkyl-, (heteroaryl)Co-C 4 alky-.

Preferred R P1 groups include optionally substituted phenyl, naphthyl, and monocyclic heteroaryl groups, especially those groups (particularly lipophilic groups) which enhance bioavailability of the compounds in the cells of the patient and which exhibit reduced toxicity, enhanced therapeutic index and enhanced pharmacokinetics (the compounds are metabolized and excreted more slowly). Stabilized Phosphate Prodrugs

Stabilized phosphate prodrugs are moieties that can deliver a mono, di, or triphosphate in vivo. For example, McGuigan has disclosed phosphoramidates in US Patent Nos : 8,933,053; 8,759,318; 8,658,616; 8,263,575; 8,119,779; 7,951,787 and 7,115,590. Alios has disclosed thiophosphoramidates in US 8,895,723 and 8,871,737 incorporated by reference herein. Alios has also disclosed cyclic nucleotides in US Patent No. 8,772,474 incorporated by reference herein. Idenix has disclosed cyclic phosphoramidates and phosphoramidate/SATE derivatives in WO 2013/177219 incorporated by reference herein. Idenix has also disclosed substituted carbonyloxymethylphosphoramidate compounds in WO 2013/039920 incorporated by reference herein. Hostetler has disclosed lipid phosphate prodrugs, see, for example, US 7,517,858. Hostetler has also disclosed lipid conjugates of phosphonate prodrugs, see, for example, US 8,889,658; 8,846,643; 8,710,030; 8,309,565; 8,008,308; and 7,790,703. Emory University has disclosed nucleotide sphingoid and lipid derivatives in WO 2014/124430. RFS Pharma has disclosed purine nucleoside monophosphate prodrugs in WO 2010/091386. Cocrystal Pharma Inc. has also disclosed purine nucleoside monophosphate prodrugs in US Patent No.: 9,173,893 incorporated by reference herein. HepDirect™ technology is disclosed in the article "Design, Synthesis, and Characterization of a Series of Cytochrome P(450) 3A-Activated Prodrugs (HepDirect Prodrugs) Useful for Targeting Phosph(on)ate-Based Drugs to the Liver," (J. Am. Chem. Soc. 126, 5154-5163 (2004). Additional phosphate prodrugs include, but are not limited to phosphate esters, 3’,5’-cyclic phosphates including CycloSAL, SATE derivatives (S-acyl-2thioesters) and DTE (dithiodiethyl) prodrugs. For literature reviews that disclose non-limiting examples see: A. Ray and K. Hostetler, “Application of kinase bypass strategies to nucleoside antivirals,” Antiviral Research (2011) 277- 2 1; M. Sofia, “Nucleotide prodrugs for HCV therapy,” Antiviral Chemistry and Chemotherapy 2011; 22-23-49; and S. Peyrottes et al., “SATE Pronucleotide Approaches: An Overview,” Mini Reviews in Medicinal Chemistry 2004, 4, 395. In one embodiment, a 5 ’-prodrug described in any of these patent filings or literature can be used in the R 10A position of the presented compounds.

In one alternative embodiment, the stabilized phosphate prodrugs, include, but are not limited to those described inU.S. Patent No. 9,173,893 and U S. Patent No. 8,609,627, incorporated by reference herein, including for processes of preparation. For example, 5 ’-prodrugs can be represented by the group: wherein Z is O or S; R 17 and R 18 , when administered in vivo, are capable of providing the nucleoside monophosphate, diphosphate, or triphosphate. Representative R 12 and R 13 are independently selected from:

(a) OR 19 where R 19 is selected from H, Li, Na, K, phenyl and pyridinyl and wherein phenyl and pyridinyl are optionally substituted with one to three substituents independently selected from the group consisting of (CH2)o-6C02R 20 and (CH2)o-6CON(R 20 )2;

R 20 is independently H, Ci- 20 alkyl, the carbon chain derived from a fatty alcohol (such as oleyl alcohol, octacosanol, triacontanol, linoleyl alcohol, and etc) or Ci- 20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C 3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 alkyl, or C 1-5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C 3-10 cycloalkyl, or cycloalkyl;

(c) the ester of a D-amino acid or L-amino acid: wherein

R 21 is restricted to those sidechains occurring in natural L-amino acids, and R 22 is H, Ci- 20 alkyl, the carbon chain derived from a fatty alcohol (such as oleyl alcohol, octacosanol, triacontanol, linoleyl alcohol, and etc) or C 1.20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C 3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 alkyl, or C 1.5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)- amino, fluoro, C 3-10 cycloalkyl, or cycloalkyl;

(d) R 17 and R 18 can come together to form a ring: wherein

R 23 is H, Ci - 2 o alkyl, Ci- 20 alkenyl, the carbon chain derived from a fatty alcohol (such as oleyl alcohol, octacosanol, triacontanol, linoleyl alcohol, etc) or C 1-20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C 3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C1-5 alkyl, or C1-5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C 3-10 cycloalkyl, or cycloalkyl;

(e) R 17 and R 18 can come together to form a ring selected from wherein

R 24 is selected from H, Ci- 20 alkyl, Ci- 20 alkenyl, the carbon chain derived from a fatty acid (such as oleic acid, linoleic acid, and the like), and Ci- 20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C 3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 alkyl, or C 1-5 alkyl substituted with a lower alkyl, alkoxy, di (lower alkyl)- amino, fluoro, C3-10 cycloalkyl, or cycloalkyl; and

R 25 is O or NH. In an alternate embodiment, 3 ’,5 ’-prodrugs can be represented by:

Formula X’ wherein: when chirality exists at the phosphorous center it may be wholly or partially R p or S p or any mixture thereof, and can be enantiomertically enriched;

R 26 is selected from fatty alcohol derived (for example but not limited to linoleyl-O- and oleyl-O-; R 11 is selected from R 1 and hydrogen; and R 1 , R 21 , R 22 , and R 19 are as defined herein.

Isotopic Substitution The present invention includes the use of an effective amount of a compound of Formula I

(including for example Compound 1, 1A or IB), Formula II (including for example, Compound 3, 3A, or 3B), Formula III (including for example, a compound of Formula Ilia, Formula Mb, Formula IIIc, Formula Hid, Formula e, or Formula Illf), Formula IV (including for example, a compound of Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula IVe, or Formula IVf), Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X or a pharmaceutically acceptable salt thereof, wherein the compounds have a desired isotopic substitutions of atoms at amounts above the natural abundance of the isotope, i.e., enriched. Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons. By way of general example and without limitation, isotopes of hydrogen, for example, deuterium ( 2 H) and tritium (¾) may be used anywhere in described structures. Alternatively or in addition, isotopes of carbon, e.g., 13 C and 14 C, may be used. An example of an isotopic substitution is deuterium for hydrogen at one or more locations on the molecule to improve the performance of the drug. The deuterium can be bound in a location of bond breakage during metabolism (an a-deuterium kinetic isotope effect) or next to or near the site of bond breakage (a b-deuterium kinetic isotope effect). Achillion Pharmaceuticals, Inc. (WO/2014/ 169278 and WO/2014/169280) describes deuteration of nucleotides to improve their pharmacokinetic or pharmacodynamic, including at the 5-position of the molecule.

Substitution with isotopes such as deuterium can afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Substitution of deuterium for hydrogen at a site of metabolic break down can reduce the rate of or eliminate the metabolism at that bond. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including protium ( 1 H), deuterium ( 2 H) and tritium ( 3 H). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.

The term "isotopically-labeled" analog refers to an analog that is a "deuterated analog", a " 13 C-labeled analog," or a "deuterated/ 13 C-labeled analog." The term "deuterated analog" means a compound described herein, whereby a H-isotope, i.e., hydrogen/protium ( 1 H), is substituted by a H-isotope, i.e., deuterium ( 2 H). Deuterium substitution can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted by at least one deuterium.

In certain embodiments, the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest. In some embodiments it is deuterium that is 90, 95 or 99% enriched at a desired location. Unless indicated to the contrary, the deuteration is at least 80% at the selected location. Deuteration of the nucleoside can occur at any replaceable hydrogen that provides the desired results.

In one embodiment, a compound of Formula XIII or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically carrier, is used to treat or prevent COVID-19 disease caused by SARS-CoV-2 in a host in need thereof as described herein:

Formula XIII wherein

R 6 is selected from hydrogen, -C(0)R 6A , -C(0)0R 6A , Ci-ealkyl, and -CH 2 -0-R 6A ;

R 6 is selected from hydrogen, -C(0)R 6A , -C(0)0R 6A , Ci- 6 alkyl, and -CH 2 -0-R 6A and in an alternative embodiment, -C(0)NR 6B R 6C ;

R 6A is selected from hydrogen, Ci^alkyl, Ci-G, haloalkyl (for example, -CHCb, -CCI 3 , -CH 2 CI, -CF 3 , -CHF 2 , -CH 2 F), aryl, and aryl(Ci- 6 alkyl)- wherein the aryl group is optionally substituted with a substituent selected from alkoxy, hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl and in an alternative embodiment, R 6A is selected from Ci- 2 oalkyl and C 2-2 oalkenyl;

R 6B and R 6C are independently selected from hydrogen, Ci- 2 oalkyl, C 2-2 oalkenyl, aryl, aryl alkyl, heteroaryl, and heteroarylalkyl wherein the Ci- 2 oalkyl, C 2-2 oalkenyl, aryl, aryl alkyl, heteroaryl, and heteroarylalkyl can optionally be substituted with at least one substituent selected from alkoxy (including but not limited to methoxy and ethoxy), hydroxy, nitro, bromo, chloro, fluoro, azido, and haloalkyl;

R 30 is CH 3 , CD 3 , CHD 2 , or CH 2 D;

R 31 is NH 2 or D or in an alternative embodiment, CD 3 ;

R 32 is CH 3 , CD 3 , CHD 2 , or CH 2 D; and

Y is selected from F and Cl; and

R 1 , R 2 , R 3 , R 4a , R , and R 5 are as defined herein.

For example, non-limiting examples of compounds of Formula XIII include: ,CH

HN 3

Additional examples of compound of Formula XIII include: ,CD

HN 3

CH 3 0 L ¾ / H0 ' ' -p · 0.5 H 2 S0 4

\J

^CD

HN 3

N N

CH 3 O A NH 2

H3C rvW AA n'

0.5 H 2 S0 4 C H3 0 H " F ·

Methods of Treatment or Prophylaxis

Treatment, as used herein, refers to the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, in an effective amount to a host, for example a human, that is or may become infected with the SARS-CoV-2 virus. In one embodiment the method of treatment comprises administration of an effective amount of Compound 1A or Compound 3A or a pharmaceutically acceptable salt thereof, for example Compound 2A or Compound 4A. In one embodiment the method of treatment comprises administration of an effective amount of Compound IB or Compound 3B or a pharmaceutically acceptable salt thereof, for example Compound 2B or Compound 4B.

The present invention also includes prophylactic or preventative therapies. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered to a host who has been exposed to and thus is at risk of infection or at risk of reinfection with the SARS-CoV-2 virus. Prophylactic treatment may be administered, for example, to a subject not yet exposed to or infected with SARS-CoV-2, but who is susceptible to, or otherwise at risk of exposure or infection with COVID-19. In one embodiment, a host at risk for infection or reinfection is administered a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof indefinitely until the risk of exposure no longer exists.

In another alternative embodiment, a method to prevent transmission is provided that includes administering an effective amount of one of the compounds described herein to humans for a sufficient length of time prior to exposure to crowds that can be infected, including during travel or public events or meetings, including for example, up to 3, 5, 7, 10, 12, 14 or more days prior to a communicable situation, either because the human is infected or to prevent infection from an infected person in the communicable situation.

In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered in an effective amount for at least two weeks, three weeks, one month, two months, three months, four months, five months, or six months or more after infection.

The invention is directed to a method of treatment of COVID-19, including drug resistant and multidrug resistant forms of the virus and related disease states, conditions, or complications of the viral infection, including pneumonia, such as 2019 novel coronavirus-infected pneumonia (NCIP), acute lung injury (ALI), and acute respiratory distress syndrome (ARDS). Additional non limiting complications include hypoxemic respiratory failure, acute respiratory failure (ARF), acute liver injury, acute cardiac injury, acute kidney injury, septic shock, disseminated intravascular coagulation, blood clots, multisystem inflammatory syndrome, chronic fatigue, rhabdomyolysis, and cytokine storm.

The method also comprises administering to a host in need thereof, typically a human, an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, optionally in combination with at least one additional bioactive agent, for example, an additional anti-viral agent, further optionally in combination with a pharmaceutically acceptable carrier additive and/or excipient.

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof to a patient in need thereof results in a reduction in the incidence of progressive respiratory insufficiency (PRI) as measured by greater than or equal to a 1-tier or even a 2-tier or more increase in respiratory support methods required to maintain satisfactory oxygenation (SpC > 93%) using the 6-tier hierarchical levels of respiratory support methods described below.

The scale of increasing respiratory support levels includes:

Level 1: Normal oxygenation on room air (SpCh >_93%), no need for supplemental O2

Level 2: Persistent hypoxemia on room air (SpCh > 93) with requirement for low-level supplemental O2 by nasal cannular or mask (up to 2L/min) to maintain SpCL > 93

Level 3: Requirement for higher levels of passive supplemental O2 by nasal cannular or mask (up to 2L/min) to maintain SpCh > 93

Level 4: Requirement for oxygenation by positive-pressure devices, e.g., Continuous Positive Airway Pressure (CPAP) or Bi-level Positive Airway Pressure (BiPAP) or other non- invasive positive-pressure respiratory support methods to main satisfactory oxygenation and/or ventilation

Level 5 : Requires invasive respiratory support (intubated mechanical ventilation or ECMO)

Level 6: Death

In one embodiment, the reduction in PRI is an increase from level 5 to level 3, level 5 to level 2, or level 5 to level 1. In one embodiment, the reduction in PRI is an increase from level 4 to level 2 or level 4 to level 1. In one embodiment, the reduction in PRI is an increase from level 3 to level 1.

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof reduces the median time to Clinical Recovery (status 6, 7, or 8 in the NIAID Clinical Status scale using an adapted National Institute of Allergy and Infectious Diseases (NIAID) ordinal scale of Clinical Status) by at least 3, 4, 5, or more days. In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof results in an improvement as measured by the adapted ordinal scale of Clinical Status.

From most severe disease to progressively less severe disease, the stages of the adapted ordinal scale of overall Clinical Status are defined as follows:

1. Death

2. Hospitalized, on invasive mechanical ventilation or ECMO

3. Hospitalized, on non-invasive ventilation or high flow oxygen devices

4. Hospitalized, requiring supplemental oxygen 5. Hospitalized, not requiring supplemental oxygen - requiring ongoing medical care (COVED- 19 related or otherwise)

6. Hospitalized, not requiring supplemental oxygen; no longer requires close medical care for COVED- 19

7. Not hospitalized, but with limitation on activities and needing close outpatient care for COVED- 19 manifestations

8. Not hospitalized, no limitations on activities, no need for continued close medical care

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof reduces the median time to Clinical Recovery (status 6, 7, or 8 in the NIAID Clinical Status scale using an adapted National Institute of Allergy and Infectious Diseases (NIAED) ordinal scale of Clinical Status) by at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, or at least 10 days.

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof reduces the duration of hospitalization for a patient infected with the SARS-CoV-2 virus.

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof reduces the time to sustained non- detectable SARS-CoV-2 virus in the nose and/or throat in a patient infected with the SARS-CoV-2 virus.

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof reduces respiratory failure or death.

In one embodiment, the administration of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof reduces the proportion of patients in a hospital population who are SARS-CoV-2 positive after at least about 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 days of treatment.

In another embodiment, a method of treating or preventing a SARS-CoV-2 infection in a host, typically a human, in need thereof is provided by administering to the host an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, wherein the SARS-CoV-2 infection is caused by a viral variant that has developed a natural or drug-induced mutation over the wild-type.

In some embodiments, the SARS-CoV-2 variant has a natural mutation or drug-induced mutation in a viral protein selected from an envelope (E) protein, membrane (M) protein, spike (S) protein, nspl, nsp2, nsp3, nsp4, nsp5, nsp 6, nsp7, nsp8, nsp9, nsplO, nspl2, nspl3, nspl4, nspl5, nspl6, ORFlab, ORF3a, ORF6, ORF7a, ORF7b, ORF8, and ORFIO. In some embodiments, the SARS-CoV-2 variant has a mutation which results in the acquired resistance to one or more anti viral drugs.

In some embodiments, the SARS-CoV-2 variant has a deletion of the spike protein amino acids H69 and V70.

In some embodiments, the SARS-CoV-2 variant has a deletion of the spike protein amino acids D614G.

In some embodiments, the SARS-CoV-2 variant has a deletion of the spike protein amino acid Y 144.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution N501Y.

In some embodiments, the SARS-CoV-2 variant which has a spike protein amino acid substitution A570D.

In some embodiments, the SAR.S-CoV-2 variant has a spike protein amino acid substitution

P681H.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

T716I.

In some embodiments, the SAR.S-CoV-2 variant has a spike protein amino acid substitution

S982A.

In some embodiments, the SAR.S-CoV-2 variant has a spike protein amino acid substitution D1118H.

In some embodiments, the SARS-CoV-2 variant has a premature stop codon mutation Q27stop in the protein product of ORF8.

In some embodiments, the SAR.S-CoV-2 variant has a spike protein amino acid substitution K417N.

In some embodiments, the SAR.S-CoV-2 variant has a spike protein amino acid substitution

E484K.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

K417N. In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution D215G.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution A701V.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

L18F.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

R246I.

In some embodiments, the SARS-CoV-2 variant has a spike protein deletion at amino acids 242-244

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

Y453F.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

I692V.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution M1229I.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution N439K.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution A222V.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

S477N.

In some embodiments, the SARS-CoV-2 variant has a spike protein amino acid substitution

A376T.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution

P323L.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution

Y455I.

In some embodiments, the SARS-CoV-2 variant has a Orf8 protein amino acid substitution

R52I.

In some embodiments, the SARS-CoV-2 variant has an ORF8 protein amino acid substitution Y73C.

In some embodiments, the SARS-CoV-2 variant has a nucleoside (N) protein amino acid substitution D3L. In some embodiments, the SARS-CoV-2 variant has a nucleoside (N) protein amino acid substitution S235F.

In some embodiments, the SARS-CoV-2 variant has a ORFlab protein amino acid substitution T 100 II.

In some embodiments, the SARS-CoV-2 variant has a ORFlab protein amino acid substitution A1708D

In some embodiments, the SARS-CoV-2 variant has a ORFlab protein amino acid substitution I2230T.

In some embodiments, the SARS-CoV-2 variant has a ORFlab protein amino acid SGF 3675-3677 deletion.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution S861X, wherein X is any amino acid.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution

F480V.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution

V557L.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution D484Y.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution F480X, wherein X=any amino acid.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution V557X, wherein X=any amino acid.

In some embodiments, the SARS-CoV-2 variant has a nspl2 protein amino acid substitution D484X, wherein X=any amino acid.

In some embodiments, the SARS-CoV-2 variant includes a deletion of the spike protein amino acids 69-70, deletion of the spike protein amino acid Y144, the spike protein amino acid substitution N501 Y, the spike protein amino acid substitution A570D, the spike protein amino acid substitution D614G, the spike protein amino acid substitution P681H, the spike protein amino acid substitution T716I, the spike protein amino acid substitution S982A, the spike protein amino acid substitution D1118H, and a premature stop codon mutation (Q27stop) in the protein product of ORF8.

In some embodiments, the SARS-CoV-2 variant includes amino acid substitutions in the spike protein of N501 Y, K417N, E484K, D80A, D215G, L18F, and R246I in the spike protein, and amino acid deletion at amino acids 242-244 of the spike protein. In some embodiments, the SARS-CoV-2 variant is selected from SARS-CoV-2 clade O, S, L, V, G, GH, or GR as described by Aim et al., "Geographical and temporal distribution of SARS- CoV-2 clades in the WHO European Region, January to June 2020". Euro Surveillance: Bulletin European Sur les Maladies Transmissibles = European Communicable Disease Bulletin. 25 (32).

In some embodiments, the SARS-CoV-2 variant is selected from SARS-CoV-2 clade G614, S84, V251, 1378 or D392 as described by Guan et al., A genetic barcode of SARS-CoV-2 for monitoring global distribution of different clades during the COVID-19 pandemic. Int J Infect Dis. 2020 Nov; 100: 216-223.

In some embodiments, the SARS-CoV-2 variant is selected from SARS-CoV-2 clade 19A, 19B, 20A, or 20C as described by Nextstrain: Genomic epidemiology of novel coronavirus - Global sub-sampling. Available from: https://nextstrain.org/ncov.

In some embodiments, the SARS-CoV-2 variant is selected from SARS-CoV-2 lineage A, B, B.l, B.l.l, or B.1.177 as described by Rambaut et al., Phylogenetic Assignment of Named Global Outbreak LINeages (pangolin). San Francisco: GitHub. Available from: https://github.com/cov-lineages/pangolin; Rambaut et al. A dynamic nomenclature proposal for SARS-CoV-2 lineages to assist genomic epidemiology. Nat Microbiol. 2020Nov;5(l 1): 1403-1407; Rambaut et al. SARS-CoV-2 lineages. Available from: https://cov-lineages.org/.

In some embodiments, the SARS-CoV-2 variant is the “Cluster 5” variant, which includes the spike protein amino acid substitution D614G.

In some embodiments, the SARS-CoV-2 variant is VUI 202012/01 (Variant Under Investigation, year 2020, month 12, variant 01) (also known as B.l.1.7 lineage and 20B/501Y.V1), which has been defined by multiple spike protein changes including deletion of the spike protein amino acids 69-70, deletion of the spike protein amino acid Y144, the spike protein amino acid substitution N501 Y, the spike protein amino acid substitution A570D, the spike protein amino acid substitution D614G, the spike protein amino acid substitution P681H, the spike protein amino acid substitution T716I, the spike protein amino acid substitution S982A, the spike protein amino acid substitution D1118H, and a premature stop codon mutation (Q27stop) in the protein product of ORF8.

In some embodiments, the SARS-CoV-2 variant is the B.l.351 lineage variant (also known as 501. V2, 20C/501Y.V2), which includes several mutations in the receptor-binding domain (RBD) in the spike protein: N501Y, K417N, and E484K, which allows the virus to attach more easily to human cells, as well as amino acid substitution D80A in the spike protein, an amino acid substitution D215G in the spike protein, an amino acid substitution A701V in the spike protein, an amino acid substitution L18F in the spike protein, an amino acid substitution R246I in the spike protein, and amino acid deletion at amino acids 242-244 of the spike protein. In some embodiments, the SARS-CoV-2 variant is the 501 Y.V2 lineage variant (also known as 501Y.V2, 20C/20H/501 Y.V2), which also includes the spike protein mutations N501 Y, K417N, and E484K.

In some embodiments, the SARS-CoV-2 variant is the P.l lineage variant (also known as the Brazil(ian) variant), which includes ten mutations in the spike protein mutations, including N501Y and E484K.

In some embodiments, the SARS-CoV-2 variant is the B.1.1.207 lineage variant, which includes a P681H mutation in the spike protein.

In some embodiments, the SARS-CoV-2 variant is the danish mink variant which includes an amino acid deletion of H69 and Y70 in the spike protein, and an amino acid substitution Y453F in the spike protein.

In some embodiments, the SARS-CoV-2 variant is the danish mink cluster 5 variant, which includes an amino acid deletion of H69 and V70 in the spike protein, an amino acid substitution Y453F in the spike protein, an amino acid substitution I692V in the spike protein, and an amino acid substitution M1229I in the spike protein.

In some embodiments, the SARS-CoV-2 variant includes an amino acid deletion of H69 and V70 in the spike protein, and an amino acid substitution N439K in the spike protein.

In some embodiments, the SARS-CoV-2 variant is the Nexstrain cluster 20A.EU1 variant, which includes an amino acid substitution A222V in the spike protein.

In some embodiments, the SARS-CoV-2 variant is the Nexstrain cluster 20A.EU2 variant, which includes an amino acid substitution S477N in the spike protein, and an amino acid substitution A376T in the nucleocapsid protein.

In some embodiments, the SARS-CoV-2 variant has one or more of the following mutations selected from: an amino acid substitution T1001I in the protein product of ORFlab; an amino acid substitution A1708D in the protein product of ORFla; an amino acid substitution I2230T in the protein product of ORFlab; a deletion of amino acids SGF at 3675-3677 in the protein product of ORFlab; an amino acid substitution G251V in the protein product of ORF3a; an amino acid substitution S24L in the protein product of ORF8; an amino acid substitution R52I in the protein product of ORF8; an amino acid substitution Y73C in the protein product of ORF8; an amino acid substitution L84S in the protein product of ORF8; an amino acid substitution P323L in the nspl2 domain; an amino acid substitution Y455I in the nspl2 domain; an amino acid substitution Q57H in the protein product of ORF3a; an amino acid substitution R27C in nsp2; an amino acid substitution VI 981 in nsp2; an amino acid substitution T85I in nsp2; an amino acid substitution P585S in nsp2; an amino acid substitution I559V in nsp2; an amino acid substitution M33I in nsp4; an amino acid substitution G15S in nsp5; an amino acid substitution L37F in nsp6; an amino acid substitution Y541C in nspl3; an amino acid substitution P504L in nspl3; an amino acid substitution S477N in the spike protein; an amino acid substitution N439K in the spike protein; an amino acid substitution N501Y in the spike protein; an amino acid substitution Y453F in the spike protein; an amino acid substitution K417N in the spike protein; an amino acid substitution E484K in the spike protein; an amino acid substitution A222V in the spike protein; an amino acid substitution S98F in the spike protein; an amino acid substitution D80Y in the spike protein; an amino acid substitution A626S in the spike protein; an amino acid substitution VI 122L in the spike protein; an amino acid substitution A570D in the spike protein; an amino acid substitution P681H in the spike protein; an amino acid substitution VI 122L in the spike protein; an amino acid substitution T716I in the spike protein; an amino acid substitution S982A in the spike protein; an amino acid substitution D1118H in the spike protein; an amino acid substitution E583D in the spike protein; an amino acid substitution V483A in the spike protein; an amino acid substitution Q675R in the spike protein; an amino acid substitution A344S in the spike protein; an amino acid substitution T345S in the spike protein; an amino acid substitution R346K in the spike protein; an amino acid substitution A348S in the spike protein; an amino acid substitution A348T in the spike protein; an amino acid substitution N354K in the spike protein; an amino acid substitution S359N in the spike protein; an amino acid substitution V367F in the spike protein; an amino acid substitution V382L in the spike protein; an amino acid substitution P384L in the spike protein; an amino acid substitution P384S in the spike protein; an amino acid substitution T385S in the spike protein; an amino acid substitution V395I in the spike protein; an amino acid substitution R403K in the spike protein; an amino acid substitution D405V in the spike protein; an amino acid substitution Q414P in the spike protein; an amino acid substitution Q414E in the spike protein; an amino acid substitution 1418V in the spike protein; an amino acid substitution L441I in the spike protein; an amino acid substitution R457K in the spike protein; an amino acid substitution K458Q in the spike protein; an amino acid substitution P463S in the spike protein; an amino acid substitution A475V in the spike protein; an amino acid substitution G476S in the spike protein; an amino acid substitution T478A in the spike protein; an amino acid substitution P479L in the spike protein; an amino acid substitution V483A in the spike protein; an amino acid substitution F490L in the spike protein; an amino acid substitution Q493L in the spike protein; an amino acid substitution A520S in the spike protein; an amino acid substitution L5F in the spike protein; an amino acid substitution P521R in the spike protein; an amino acid substitution A522S in the spike protein; an amino acid substitution A83 IV in the spike protein; an amino acid substitution D839Y in the spike protein; an amino acid substitution D839N in the spike protein; an amino acid substitution D839E in the spike protein; an amino acid substitution L8V in the spike protein; an amino acid substitution L8W in the spike protein; an amino acid substitution FI49Y in the spike protein; a deletion of amino acid FI69 in the spike protein; a deletion of amino acid V70 in the spike protein; a deletion of amino acid Y144 in the spike protein; an amino acid substitution D3L in the nucleocapsid protein; an amino acid substitution S253F in the nucleocapsid protein; an amino acid substitution RG203KR in the nucleocapsid protein; an amino acid substitution G214C in the nucleocapsid protein; an amino acid substitution S194L in the nucleocapsid protein; an amino acid substitution F377L in the nspl4 protein; an amino acid substitution K1186R in nsp3; or an amino acid substitution A58T in nsp3.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the envelope (E) protein: S68F; L73F; P71L; S55F; R69I; T9I; V24M; D72H; T30I; S68C; V75L; V58F; V75F; or L21F; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the membrane (M) protein: T175M; D3G; V23L; W31C; A2V; V70F; W75L; Ml 091; I52T; L46F; V70I; D3Y; K162N; H125Y; K15R; D209Y; R146H; R158C; L87F; A2S; A69S; S214I; T208I; L124F; or S4F; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nucleocapsid (N) protein: RG203KR; S194L; S197L; P13L; D103Y; S 1931; S188L; I292T; S202N; D401Y; S190I; D22G; A208G; T205I; S183Y; S33I; D81Y; T393I; A119S; D377Y; S37P; T247I; A156S; D128Y; P199L; R195I; P207L; E62V; R209T; T362I; G18C; T24N; R185C; SI 801; M234I; Q9H; P383L; A35S; P383S; D348H; K374N; R32H; S327L; G179C; G238C; A55S; S190G; H300Y; A119V; D144Y; L139F; P199S; P344S; P6L; R203K; P364L; R209I; S188P; A35V; K387N; P122L; R191C; R195K; T391I; A252S; Q418L; T271I; T325I; G18V; L161F; Q289H; R203S; P162L; D340N; K373N; P168Q; A211V; D3L; G212V; K370N; P151L; T334I; A359S; G34W; P67T; R203M; D144N; R191L; S232I; D402Y; P168S; S187L; T366I; A152S; A381T; N140T; T198I; A251V; A398V; A90S; D348Y; D377G; G204R; G243C; G34E; Q229H; R185L; T24I; T379I; A134V; N196I; P365S; Q384H; R276I; S235F; D216A; M210I; M322I; P20S; Q389H; R209 deletion; or V246I; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nspl protein: M85; D75E; G82 deletion; V84 deletion; P80 deletion; H83 deletion; V86 deletion; H81 deletion; E87 deletion; L88 deletion; K141 deletion;A79 deletion;V89 deletion; V56I; R124C; D75G; A90 deletion; Y118C; D139N; Y136 deletion; G30D; R24C; D139Y; E37K; H45Y; H110Y; G52S; I71V; D156 deletion; A76T; E37D; S135 deletion; S166G; A138T; F157 deletion; G49C; M85I; or D144A; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsplO protein: D64E; P136S; A104V; A32V; T12I; Ti l II; P84S; T51I; I55V; T102I; or T51A; and combinations thereof. In some embodiments, the SARS-CoV2 variant contains one or more of the following mutations in the nspl2 protein: P323L; T141I; A449V; S434F; M666I; H613Y; S647I; M3801; E922D; M629I; G774S; M601I; E436G; N491S; Q822H, A443V, T85I; A423V; M463I, T26I; A656T; M668I; T806I; T276M; T801N; V588L; K267N; V880I; K718R; L514F; F415S; T252N; Y38H; E744D; H752Q; I171V; S913L; A526V; A382V; G228C; P94L; E84K; K59N; P830S; T908I; P21S; D879Y; G108D; K780N; R279S; D258Y; T259I; K263N; D284Y, Q292H; T293I; N297S; Y299F; D304Y; T319I; F321L; P328S; V330E; I333T; G337C; T344I; Y346H; L351P; V354L; Q357H; E370G; L372F; A400S; T402I; V405F; V410I; D418N; K426N; K430N; V435F; Q444H; D445G; A448V; R457C; P461T, C464F; I466V; V473F; K478N; D481G; D517G; D523N; A529V; P537S; S549N; A555V; C563F; M566I; A581T; G584V; A585T; G596S; T604I; S607I; D608G; V609I; M615V; W617L; M629V; I632V; L636F; L638F; A639V; T643I; T644M; L648F; V667I; A699S; N713S; H725; N734T; D736N; V737F; T739I; V742M; N743S; M756I; L758I; A771V; L775Y; A777T; K780T; F793L; T801I; T803A; H810Y; G823C; D825Y; V827A; Y828H; V848L; T870I; K871R; N874D; Q875R; E876D; H882Y; H892Y; D901Y; M906I; N909D; T912N; P918S; E919D; A923T; F480V; V557L; D484Y; or S433G; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nspl3 protein: Y541C; P504L; A18V; R392C; P47L; S485L; L297P; H290Y; T127I; L176F; V193I; V570L; D260Y; V49I; Q518H; S468L; A598V; D204Y; S74L; T588I; G206C; V226L; V348L; M576I; A302D; P53S; T481M; K524N; A338V; P419S; V479F; P77L; V169F; N124S; P78S; S80G; V496L; A4V; T413I; A296S; A368S; K460R; L297F; P172S; A302S; P402S; T530I; L428F; P504S; A368V; D458Y; P364S; S74P; T416A; A568V; M474I; S166L; S350L; D344N; E341D; I432T; L581F; S38L; T250I; Y253H; A509V; E244D; H164Y; S74A; T141I; V356F; E319D; E365D; G170S; L526F; R155C; or Y396C; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nspl4 protein: A320V; F233L; T250I; V182L; A225V; R289C; A274S; P24L; I150T; S374A; H26Y; L177F; L157F; T16I; A482V; P297S; V120A; S255I; P203L; A23 deletion; K311N; M72I; V290F; F431L; K349N; M58I; P140S; R205C; T193A; L409F; P443S; Y260C; D345G; E204D; R163C; R81K; T524I; T113I; T31I; L493F; A119V; D345Y; M501I; A360V; A371V; T206I; V287F; A360S; I74T; M315I; P142L; or Q343K; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nspl5 protein: V320L; A217V; V22L; V172L; D219N; P205S; V127F; Q19H; M218 deletion; A92V; D282G; I252V; T33I; G129S; L331F; A81V; V69L; S312F; T325I; A171V; R206S; D272Y; D87N; S288F; K109R; P270S; P65S; D267Y; D128Y; E215I; T144I; S261L; S287L; T112I; E260K; P205L; S161I; V66L; D39Y; or T114A; or combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nspl6 protein: S33R; K160R; P134S; Q28K; T195I; Y78G; T35I; G265V; K249N; A204S; K182N; R287I; A188S; A116V; T140I; L111F; M270T; R216N; A188V; A34V; D108N; L163F; L163H; Ml 71; T91M; A226S; G77R; L126F; N298L, R216S; T48I; Q238H; or R279K; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp2 protein: T85I P585S; I559V; D268; G212D; V198I; H237R; F10L; G339S; T166I; R27C; L271F; S211F; P91S; G199E; T371I; A336Y; I120F; S122F; A476V; S138L; V480A; T388I; T634I; P129S; R218C; I188T; T170I; P568L; E574A; I367V; H208Y; S99F; T429I; A306V; M405V; P129L; R222C; T44I; Q275H; R380C; A360V; A361V; G115C; L353F; H237Y; L462F; E261G; R4C; S263F; T573I; A318V; G262V; P624L; S430L; T422I; A357S; I100V; E272G; L400F; A192V; D464A; E172D; G262S; L501F; S369F; E172K; G465S; K219R; A411V; A522V; H194Y; S32L; F437L; P181S; P446L; G115V; H532Y; N92H; P13S; A159V; A184S; A306S; I273T; L274F; P13L; R370H; T223I; T590I; E453D; H145Y; K618N; S301F; T153M; V244I; V530I; A127V; L24F; P191L; Q182L; S196L; S248G; S378F; T139I; T434I; A205V; A375V; A411S; C51Y; F300L; M135T; P568S; Q496H; S348P; T412I; T528I; T547I; V447F; or V577I; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp3 protein: A58T; T1198K; T428I; P153L; S1197R; D218E; S1424F; A1431V; S1285F; P74L; Q1884H; P1326L; L1221F; P141S; P1103S; S126L; Y916H; L557F; E391D; A1311V; S650F; P1103L; Y952H; P340S; A534V; P1787S; L1791F; N1587S; S371N; K1693N; G282V; P278S; T1335I; A1711V; K19R; A994D; K1325R; P822L; K412N; A465V; T1004I; T808I; G489D; S1699F; M1436V; S1265R; V1768G; A231V; M951I; K384N; T1288I; Q966H; R1614K; T1036I; T1306I; A1179V; P395L; N1785D; P679L; S166G; A1769V; T181I; L1718F; P822S; T 10221; A1381V; A602T; I1720V; K837N; T73I; A1033V; S1204; C1223Y; P389L; T398A; M1441I; M494I; T1303I; T181A; P1228L; R1135K; V267F; A1883V; A655V; S1296F; T686I; L198I; P1403S; L781F; T1046A; A1215V; E374D; 1205 deletion; V477F; E324K; I707V; P109L; P1558L; P74S; S1212L; S1807F; T819I; T864I; H1000Y; P340L; S697F; T1189I; A480V; D729Y; K1771R; S1717L; T749I; M829I; Q172R; T1482I; A1395V; I385T; M560I; S1206L; S1699P; T1269I; T779I; V1315I; V1795F; V325F; A1892V; A579V; E493G; H1274Y; S1467F; T 10631; T350I; V61F; A1736V; K1804N; R646W; T583I; T611I; V1243I; V190I; A41V; H290Y; H295Y; H342Y; L1244F; Q128H; V1673I; A1305V; A1526S; E948K; L72F; P125S; P402T; A1766V; D1214N; E1271D; G1440D; G283D; K1211N; K902N; K945N; L1839S; L312F; N1263S; P1292S; S1670F; S743A; T771I; V1936I; A1262V; A1321V; A358Y; A41T; C55Y; G1273S; K463E; K497Q; P1044S; R30K; S1375F; S1682F; T133I; T1348I; 4651; T1830I; T237I; V1248L; A225V; A496V; G1217R; I1816T; L956I; N1369T; N506S; P153S; P2L; T1275I; T1459I; V1234M; E595D; F90L; G1585S; H1307Y; I1409V; L1034V; L1328F; L292F; N1264; P1326T; S1197G; T1456I; T64I; T703I; T720I; T820I; V1229F; V234I; A1279V; A333V; A54S; D1121G; D1761N; E731D; I1672T; I789V, K1037R; K487N; L142F; N1177H; P1228S; P723S; Q180H; Q474R; Q940L; S370L; T1180I; T275I; T422I; T526I; T724I; V1434G; or Y207L; or combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp4 protein: F308Y; T295I; M33I; A307V; A457V; G309C; L360F; A231V; H313Y; K399E; V20F; S137L; S34F; A380V; H470Y; T204I; S336L; L264F; L438F; M33L; S209F; C296S; L475I; G79V; T327N; T350I; L206F; M324I; E230G; L436 deletion; T237I; T492I; A260V; A446V; M458I; S395G; S481L; H36Y; T73I; L323F; L349F; S59F; T214I; or T60I; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp5 protein: G15S; D248E; K90R; L89F; A266V; P108S; A70T; A129V; T45I; G71S; L75F; A191V; L220F; N274D; L67F; P241L; K236R; V157L; K61R; P184S; S62Y; T21I; L50F; P108L; S254F; T93I; A255V; A94V; P132S; A234V; A260V; R60C; P96L; V247F; or T199I; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp6 protein: L37F; G277S; A46V; L75F; F37 deletion; T10I; V149F; L260F; Q208H; M83I; A136V; V145I; N156D; M86I; Y153C; G188V; L230I; F34 deletion; I189V; R233H; V114A; L33F; A287V; H11Y; A287T; A51V; G188S; I162T; M126V; M183I; N40Y; SI 04; F35L; M58L; or V84F; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp7 protein: S25L; S26F; L71F; S15T; M75I; orN78S; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp8 protein: M129I; I156V; T145I; R51C; T123I; L95F; T89I; P133S; S41F; K37N; T141M; V34F; R51L; A14T; A74V; I107V; A16V; P10S; A194V; D30G; A152V; or T187I; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the nsp9 protein: T77I; T109I; L42F; T34I; T19I; M101V; T62I; or T19K; and combinations thereof. In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the protein product of ORFIO: L17P; A28V; P10S; I4L; S23F; R24C; *39Q; Q29 stop; Y14C, R20I; orA8V; and combinations thereof

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the protein product of ORF3a: Q57H; G251V; V13L; G196V; A54S; A99V; H93Y; T14I; L46F; Q185H; T175I; Q213K; L108F; K61N; Y264C; A72S; T151I; A23S; G224C, K67N; S171L; W69L; H78Y; K136E; L86F; W131C; L147F; S58N; Y91H; I63T; D155Y; G172C; P240L; Y189C; W131R; KN136NY; T223I; G100C; S195Y; V112F; W131L; G44V; D27H; G174C; K21N, S165F; L65F; T229I; T89I; S74F; A99S; G254R; H204N; K75N; F43L; L53F; Q38P; S26L; S40L; M260I; V256 deletion; K16N; Q218R; S253P; V163L; W69C; A23V; L41F; L106F; V55F; V88A; A99D; E239D; L52F; T24I; A3 IT; D27Y; I186V; L73F; P104L; D22Y; FI 14V; L95F; P240S; P42L; T268M; or T32I; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the protein product of ORF6: I33T; W27L; D53G; F22 deletion; P57L; D61Y; D61L; K42N; D53Y; H3Y; I32T; or R20S; or combinations thereof

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the protein product of ORF7a: S81L; A8T; L96F; A50V; V104F; Q62 stop; S83L; E16D; T14I; T28I; V93F; G38V; H47Y; T39I; T120S; Q62 deletion; Q62L; S37T; V104; P34S; P99L; T120I; V108L; H73Y; V24F; V29L; A13T; or L5F; or combinations thereof

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the protein product of ORF7b: C41F; T40I; A43V; LI IF; S31L; C41 deletion; H42; H42L; S5L; L20F; L32F; E33 stop; A15S; or F13 deletion; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the protein product of ORF8: El 10 stop; G66 deletion; S69L; Till; F104L; F120L; G8R; P38S; D119E; II OS; or I39V; and combinations thereof.

In some embodiments, the SARS-CoV-2 variant contains one or more of the following mutations in the spike protein: D614G; D936Y; P1263L; L5F; N439K; R21I; D839Y; L54F; A879S; L18F; F1121L; R847K; T478I; A829T; Q675H; S477N; H49Y; T29I; G769V; G1124V; V1176F; K1073N; P479S; S1252P; Y145 deletion; E583D; R214L; A1020V; Q1208H; D215G; H146Y; S98F; T95I; G1219C; A846V; I197V; R102I; V367F; T572I; A1078S; A831V; P1162L; T73I; A845S; G1219V; H245Y; L8V; Q675R; S254F; V483A; Q677H; D138H; D80Y; M1237T; D1146H; E654D; H655Y; S50L; S939F; S943P; G485R; Q613H; T76I; V341I; M153I; S221L; T859I; W258L; L242F; P681L; V289I; A520S; V1104L; V1228L; L176F; M1237I; T307I; T716I; L141; M1229I; A1087S; P26S; P330S; P384L; R765L; S940F; T323I; V826L; E1202Q; L1203F; L611F; V615I; A262S; A522V; A688V; A706V; A892S; E554D; Q836H; T 10271; T22I; A222V; A27S; A626V; C1247F; K1191N; M731I; P26L; S1147L; S1252F; S255F; V1264L; Y308L; D80A; I670L; P251L; P631S; *1274Q; A344S; A771S; A879T; D1084Y; D253G; H1101Y; L1200F; Q14H; Q239K; A623V, D215Y; E1150D; G476S, K77M; Ml 771; P812S; S704L; T51I; T547I; T791I; V1122L; Y145H; D574Y; G142D; G181V; I834T; N370S; P812L; S12F; T791P; V90F; W152L; A292S; A570V; A647S; A845V; D1163Y; G181R; L84I; L938F; P1143L; P809S; R78M; T1160I; V1133F; V213L; V615F; A831V; D839Y; D839N; D839E; S943P; P1263L; or V622F; and combinations thereof.

Pharmaceutical Compositions and Dosage Forms

A compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, can be administered in an effective amount for the treatment of the 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus in a host, typically a human, in need thereof. In one embodiment the compound is Compound 1A or Compound 3A or a pharmaceutically acceptable salt thereof, for example Compound 2A or Compound 4A. In one embodiment the compound is Compound IB or Compound 3B or a pharmaceutically acceptable salt thereof, for example Compound 2B or Compound 4B.

The compound or its salt can be provided as the neat chemical but is more typically administered as a pharmaceutical composition that includes an effective amount for a host, typically a human, in need of a treatment for COVID-19. Thus, in one embodiment, the disclosure provides pharmaceutical compositions comprising an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, with at least one pharmaceutically acceptable carrier for the treatment of COVID-19. The pharmaceutical composition may contain a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, as the only active agent, or, in an alternative embodiment, in combination with at least one additional active agent.

A compound of Formula I (including but not limited to Compound 1, 1A or IB), Formula II (including but not limited to Compound 3, 3A or 3B), Formula III (including, Formula Ilia, Formula Illb, Formula IIIc, Formula Hid, Formula Hie, or Formula Illf), Formula IV (including, Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula IVe, or Formula IVf), Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, can be formulated with one or more pharmaceutically acceptable carriers. Oral dosage forms are sometimes selected due to ease of administration and prospective favorable patient compliance. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula CIP or a pharmaceutically acceptable salt thereof, is provided in a solid dosage form, such as a tablet or pill, which are well known in the art and described further below. Enteric coated oral tablets may also be used to enhance bioavailability of the compounds for an oral route of administration. Pharmaceutical compositions (formulations) may be administered via oral, parenteral, intravenous, inhalation, intramuscular, topical, transdermal, buccal, subcutaneous, suppository, or other route, including intranasal spray routes of delivery.

In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is administered intravenously. In one non-limiting embodiment, a compound of the present invention is administered intravenously at a loading dose of 550 mg/day and a maintenance dose of 275 mg/day. In one embodiment, the loading dose is administered once and the maintenance dose is administered twice a day for at least 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 days. In one non-limiting embodiment, an intravenous loading dose is 550 mg/day of Compound 1 (i.e., 600 mg/day hemisulfate salt of Compound 1), and a maintenance dose is 275 mg/day (i.e, 300 mg/day of hemisulfate salt)).

Effective dosage form will depend upon the bioavailability/pharmacokinetic of the particular agent chosen as well as the severity of disease in the patient. A compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, can be administered, for example, in one or more tablets, capsules, injections, intravenous formulations, suspensions, liquids, emulsions, implants, particles, spheres, creams, ointments, suppositories, inhalable forms, transdermal forms, buccal, sublingual, topical, gel, mucosal, and the like.

Intravenous and intramuscular formulations are often administered in sterile saline. One of ordinary skill in the art may modify the formulations to render them more soluble in water or another vehicle, for example, this can be easily accomplished by minor modifications (salt formulation, esterification, etc.).

The pharmaceutical compositions contemplated here optionally include a carrier, as described further below. Carriers must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated. The carrier can be inert or it can possess pharmaceutical benefits of its own. The amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound. Representative carriers include solvents, diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agent, viscosity agents, tonicity agents, stabilizing agents, and combinations thereof. In some embodiments, the carrier is an aqueous carrier.

One or more viscosity agents may be added to the pharmaceutical composition to increase the viscosity of the composition as desired. Examples of useful viscosity agents include, but are not limited to, hyaluronic acid, sodium hyaluronate, carbomers, polyacrylic acid, cellulosic derivatives, polycarbophil, polyvinylpyrrolidone, gelatin, dextin, polysaccharides, polyacrylamide, polyvinyl alcohol (including partially hydrolyzed polyvinyl acetate), polyvinyl acetate, derivatives thereof and mixtures thereof.

Solutions, suspensions, or emulsions for administration may be buffered with an effective amount of buffer necessary to maintain a pH suitable for the selected administration. Suitable buffers are well known by those skilled in the art. Some examples of useful buffers are acetate, borate, carbonate, citrate, and phosphate buffers.

To prepare the pharmaceutical compositions according to the present invention, a therapeutically effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof may be admixed with a pharmaceutically acceptable carrier according to conventional pharmaceutical compounding techniques to produce a dose. A carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral.

In preparing pharmaceutical compositions in oral dosage form, any of the usual pharmaceutical media may be used. Thus, for liquid oral preparations such as suspensions, elixirs, and solutions, suitable carriers and additives including water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like may be used. For solid oral preparations such as powders, tablets, capsules, and for solid preparations such as suppositories, suitable carriers and additives including starches, sugar carriers, such as dextrose, mannitol, lactose, and related carriers, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used. If desired, the tablets or capsules may be enteric-coated or sustained release by standard techniques. The use of these dosage forms may significantly enhance the bioavailability of the compounds in the patient.

For parenteral formulations, the carrier will usually comprise sterile water or aqueous sodium chloride solution, though other ingredients, including those which aid dispersion, also may be included. Of course, where sterile water is to be used and maintained as sterile, the compositions and carriers must also be sterilized. Injectable suspensions may also be prepared, in which case appropriate liquid carriers, suspending agents, and the like may be employed. Liposomal suspensions (including liposomes targeted to viral antigens) may also be prepared by conventional methods to produce pharmaceutically acceptable carriers. This may be appropriate for the delivery of free nucleosides, acyl/alkyl nucleosides or phosphate ester pro-drug forms of the nucleoside compounds according to the present invention.

Amounts and weights mentioned in this disclosure typically refer to the free form (i.e., non salt, hydrate or solvate form). The typically values described herein represent free-form equivalents, i.e., quantities as if the free form would be administered. If salts are administered the amounts need to be calculated in function of the molecular weight ratio between the salt and the free form.

The amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, in the pharmaceutically acceptable formulation according to the present invention is an effective amount to achieve the desired outcome of treating COVID- 19, reducing the likelihood of COVID-19, or the inhibition, reduction, and/or elimination of COVID-19 or its secondary effects, including disease states, conditions, and/or complications which occur secondary to the virus. As non-limiting embodiments, a therapeutically effective amount of the present compounds in a pharmaceutical dosage form may range, for example, from about 0.001 mg/kg to about 100 mg/kg per day or more. A compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, may for example in non limiting embodiments be administered in amounts ranging from about 0.1 mg/kg to about 15 mg/kg per day of the patient, depending upon the pharmacokinetics of the agent in the patient.

The weight of active compound in the dosage form described herein is with respect to either the free form or the salt form of the compound unless otherwise specifically indicated. For example, approximately 600 mg of Compound 2 is the equivalent of approximately 550 mg of Compound 1.

In certain embodiments, the pharmaceutical composition is in a dosage form that contains from about 1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, from about 200 mg to about 600 mg, from about 300 mg to about 500 mg, or from about 400 mg to about 450 mg of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, in a unit dosage form.

In certain embodiments, the pharmaceutical composition is in a dosage form, for example in a solid dosage form, that contains up to about 10, about 50, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 625, about 650, about 675, about 700, about 725, about 750, about 775, about 800, about 825, about 850, about 875, about 900, about 925, about 950, about 975, about 1000 mg, about 1050 mg, about 1100 mg, about 1150 mg, about 1200 mg, about 1250 mg, about 1300 mg, about 1350 mg, about 1400 mg, about 1450 mg, about 1500 mg, about 1550 mg, about 1600 mg, about 1650 mg, about 1700 mg or more of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula LX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, in a unit dosage form.

In certain embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, for example Compound 1 or Compound 2, is administered at an initial dose (or loading dose) followed by a maintenance dose of at least about 300 mg, at least about 350 mg, at least about 400 mg, at least about 450 mg, at least about 500 mg, at least about 550 mg, at least about 650, or at least about 750 and the dose is taken once or twice a day. In one embodiment, the loading dose is about 1.5 times greater, about 2 times greater, about 2.5 times greater, or 3-fold times greater than the maintenance dose. In one embodiment, the loading dose is administered once, twice, three, four, or more times before the first maintenance dose.

In one embodiment, the pharmaceutical composition is in a dosage form, for example in a solid dosage form, that contains at least 500 mg, at least 550 mg, 600 mg, at least 700 mg, at least 800 mg, at least 900 mg, at least 1000 mg, at least 1100 mg, at least 1200, at least 1300 mg, at least 1400 mg, or at least 1500 mg of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, in a unit dosage form.

In certain embodiments, the pharmaceutical composition, for example, a solid dosage form, contains at least about 450 mg, 550 mg, 650 mg, 750 mg or 850 mg of Compound 1 or Compound 3. In one embodiment, the pharmaceutical composition contains at least about 500 mg, at least about 550 mg, or at least about 600 mg of Compound 1 or Compound 3 and the composition is administered twice a day. In one embodiment, the pharmaceutical composition contains at least about 550 mg of Compound 1 and the pharmaceutical composition is administered twice a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 900 mg, 1000 mg, 1100 mg, 1100 mg, or 1200 mg of Compound 1 followed by a dose of at least about 400 mg, at least about 450 mg, at least about 500 mg, at least about 550, at least about 600 mg, or at least about 650 mg of Compound 1 twice a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 1100 mg of Compound 1 followed by a dose of at least about 450 mg, 550 mg, 650 mg, 750 mg, or 850 mg of Compound 1 twice a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 1100 mg of Compound 1 followed by a dose of at least about 550 mg of Compound 1 twice a day. In one embodiment, the maintenance dose is administered for at about 4, 5, 6, 7, 8, 9, 10, or more days. In one embodiment, Compound 1 is Compound 1A. In one embodiment, Compound 1 is Compound IB

In one embodiment, an effective amount of a compound of Formula I: or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered for the treatment of the 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus in a human in need thereof wherein the compound is administered according to the following schedule:

(i) a single loading dose of 1100 mg of free base in one day; followed by

(ii) a maintenance dose of 550 mg of free base per day.

In one embodiment, an effective amount of a compound of the Formula: or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, is administered for the treatment of the 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus in a human in need thereof wherein the compound is administered according to the following schedule:

(i) a single loading dose of 1100 mg of free base in one day; followed by

(ii) a maintenance dose of 550 mg of free base per day. In one embodiment, an effective amount of a compound of the Formula: optionally in a pharmaceutically acceptable carrier, is administered for the treatment of the 2019 coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus in a human in need thereof wherein the compound is administered according to the following schedule:

(i) a single loading dose of 1200 mg of salt in one day; followed by

(ii) a maintenance dose of 600 mg of salt per day.

In certain embodiments, the pharmaceutical composition, for example, a solid dosage form, contains at least about 400 mg, at least about 500 mg, 600 mg, 700 mg, or 800 mg of Compound 2 or Compound 4. In one embodiment, the pharmaceutical composition contains at least about 500 mg, at least about 600 mg, or at least about 700 mg of Compound 2 or Compound 4 and the composition is administered twice a day. In one embodiment, the pharmaceutical composition contains at least about 600 mg of Compound 2 and the pharmaceutical composition is administered twice a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 900 mg, 1000 mg, 1100 mg, 1200 mg, or 1300 mg of Compound 2 followed by a dose of at least about 400 mg, 500 mg, 600 mg, 700 mg, or 800 mg of Compound

2 once, twice, or three times a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 1000 mg, 1200 mg, or 1400 mg of Compound 2 followed by a dose of at least about 600 mg of Compound 2 twice a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 1200 mg of Compound 2 followed by a dose of at least about 400 mg, 500 mg, 600 mg, 700 mg, or 800 mg of Compound 2 twice a day. In one embodiment, the pharmaceutical composition is administered at an initial dose (or loading dose) of at least about 1200 mg of Compound 2 followed by a dose of at least about 600 mg of Compound 2 twice a day. In one embodiment, the maintenance dose is administered for at about 4, 5, 6, 7, 8, 9, 10, or more days. In one embodiment, Compound 2 is Compound 2A. In one embodiment, Compound 2 is Compound IB

In certain embodiments, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered for at least five days, six days, seven days, eight days, nine days, ten days, two weeks, three weeks, one month, at least two months, at least three months, at least four months, at least five months, at least six months or more. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IY, Formula V, Formula

VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered once, twice, three, or more times a day In one embodiment, it is administered orally twice a day.

For purposes of the present invention, a prophylactically or preventive effective amount of the compositions according to the present invention may generally fall within the ranges set out above, and can be determined in the best judgement of the health care provider. In one embodiment, a compound of the present invention is administered seasonally as the risk of the virus increases to prevent infection, or can be administered, for example, before, during and/or after travel or exposure.

One of ordinary skill in the art will recognize that a therapeutically effective amount will vary with the infection or condition to be treated, its severity, the treatment regimen to be employed, the pharmacokinetic of the agent used, as well as the patient or subject (animal or human) to be treated, and such therapeutic amount can be determined by the attending physician or specialist.

Solid Dosage Forms

An aspect of the invention is a solid dosage form that includes an effective amount of a compound of Formula I (including but not limited to Compound 1, 1A, IB, 2, 2A or 2B), Formula II (including but not limited to Compound 3), Formula III (including Formula Ilia, Formula Illb, Formula IIIc, Formula Hid, Formula Hie, or Formula Illf), Formula IV (including Formula IVa, Formula IVb, Formula IVc, Formula IVd, Formula IVe, or Formula IVf), Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.

In one embodiment, the solid dosage form includes a spray dried solid dispersion of a compound of Formula I, Formula II, Formula PI, Formula IV, Formula V, Formula VI, Formula

VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, and the composition is suitable for oral delivery. In another embodiment, the solid dosage form is a granulo layered solid dispersion of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, and the composition is suitable for oral delivery. In other embodiments, the solid dispersion also contains at least one excipient selected from copovidone, poloxamer and HPMC-AS. In one embodiment the poloxamer is Poloxamer 407 or a mixture of poloxamers that may include Poloxamer 407. In one embodiment HPMC-AS is HPMC- AS-L.

In other embodiments, a solid dosage form prepared from a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, also comprises one or more of the following excipients: a phosphoglyceride; phosphatidylcholine; dipalmitoyl phosphatidylcholine (DPPC); dioleylphosphatidyl ethanolamine (DOPE); dioleyloxypropyltriethylammonium (DOTMA); dioleoylphosphatidylcholine; cholesterol; cholesterol ester; diacylglycerol; diacylglycerolsuccinate; diphosphatidyl glycerol (DPPG); hexanedecanol; fatty alcohol such as polyethylene glycol (PEG); polyoxyethylene-9-lauryl ether; a surface active fatty acid, such as palmitic acid or oleic acid; fatty acid; fatty acid monoglyceride; fatty acid diglyceride; fatty acid amide; sorbitan trioleate (Span®85) glycocholate; sorbitan monolaurate (Span®20); polysorbate 20 (Tween®20); polysorbate 60 (Tween®60); polysorbate 65 (Tween®65); polysorbate 80 (Tween®80); polysorbate 85 (Tween®85); polyoxyethylene monostearate; surfactin; a poloxomer; a sorbitan fatty acid ester such as sorbitan trioleate; lecithin; lysolecithin; phosphatidylserine; phosphatidylinositol; sphingomyelin; phosphatidylethanolamine (cephalin); cardiolipin; phosphatidic acid; cerebroside; dicetylphosphate; dipalmitoylphosphatidylglycerol; stearylamine; dodecylamine; hexadecyl -amine; acetyl palmitate; glycerol ricinoleate; hexadecyl stearate; isopropyl myristate; tyloxapol; poly(ethylene glycol)5000- phosphatidylethanolamine; poly(ethylene glycol)400-monostearate; phospholipid; synthetic and/or natural detergent having high surfactant properties; deoxycholate; cyclodextrin; chaotropic salt; ion pairing agent; glucose, fructose, galactose, ribose, lactose, sucrose, maltose, trehalose, cellbiose, mannose, xylose, arabinose, glucoronic acid, galactoronic acid, mannuronic acid, glucosamine, galatosamine, and neuramic acid; pullulan, cellulose, microcrystalline cellulose, silicified microcrystalline cellulose, hydroxypropyl methylcellulose (HPMC), hydroxycellulose (HC), methylcellulose (MC), dextran, cyclodextran, glycogen, hydroxyethylstarch, carageenan, glycon, amylose, chitosan, N,O-carboxylmethylchitosan, algin and alginic acid, starch, chitin, inulin, konjac, glucommannan, pustulan, heparin, hyaluronic acid, curdlan, and xanthan, mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol, a pluronic polymer, polyethylene, polycarbonate (e.g., poly(l,3-dioxan-2one)), polyanhydride (e g., poly(sebacic anhydride)), polypropylfumerate, polyamide (e g. polycaprolactam), polyacetal, polyether, polyester (e.g., polylactide, polyglycolide, polylactide-co-glycolide, polycaprolactone, polyhydroxyacid (e.g., poly((p-hydroxyalkanoate)|), poly(orthoester), polycyanoacrylate, polyvinyl alcohol, polyurethane, polyphosphazene, polyacrylate, polymethacrylate, polyurea, polystyrene, and polyamine, polylysine, polylysine-PEG copolymer, and poly(ethyleneimine), poly(ethylene imine)-PEG copolymer, glycerol monocaprylocaprate, propylene glycol, Vitamin E TPGS (also known as d-a-Tocopheryl polyethylene glycol 1000 succinate), gelatin, titanium dioxide, polyvinylpyrrolidone (PVP), hydroxypropyl methyl cellulose (HPMC), hydroxypropyl cellulose (HPC), methyl cellulose (MC), block copolymers of ethylene oxide and propylene oxide (PEO/PPO), polyethyleneglycol (PEG), sodium carboxymethylcellulose (NaCMC), or hydroxypropylmethyl cellulose acetate succinate (HPMCAS).

In other embodiments, a solid dosage form prepared from a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, also comprises one or more of the following surfactants: polyoxyethylene glycol, polyoxypropylene glycol, decyl glucoside, lauryl glucoside, octyl glucoside, polyoxyethylene glycol octylphenol, Triton X-100, glycerol alkyl ester, glyceryl laurate, cocamide MEA, cocamide DEA, dodecyldimethylamine oxide, and poloxamers. Examples of poloxamers include, poloxamers 188, 237, 338 and 407. These poloxamers are available under the trade name Pluronic® (available from BASF, Mount Olive, N.J.) and correspond to Pluronic® F-68, F-87, F-108 and F-127, respectively. Poloxamer 188 (corresponding to Pluronic® F-68) is a block copolymer with an average molecular mass of about 7,000 to about 10,000 Da, or about 8,000 to about 9,000 Da, or about 8,400 Da. Poloxamer 237 (corresponding to Pluronic® F-87) is a block copolymer with an average molecular mass of about 6,000 to about 9,000 Da, or about 6,500 to about 8,000 Da, or about 7,700 Da. Poloxamer 338 (corresponding to Pluronic® F-108) is a block copolymer with an average molecular mass of about 12,000 to about 18,000 Da, or about 13,000 to about 15,000 Da, or about 14,600 Da. Poloxamer 407 (corresponding to Pluronic® F-127) is a polyoxyethylene-polyoxypropylene triblock copolymer in a ratio of between about E101 P56 E101 to about E106 P70 E106, or about E101 P56E101, or about El 06 P70 El 06, with an average molecular mass of about 10,000 to about 15,000 Da, or about 12,000 to about 14,000 Da, or about 12,000 to about 13,000 Da, or about 12,600 Da.

In yet other embodiments, a solid dosage form prepared from a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, also comprises one or more of the following surfactants: polyvinyl acetate, cholic acid sodium salt, dioctyl sulfosuccinate sodium, hexadecyltrimethyl ammonium bromide, saponin, sugar esters, Triton X series, sorbitan trioleate, sorbitan mono-oleate, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monooleate, oleyl polyoxyethylene (2) ether, stearyl polyoxyethylene (2) ether, lauryl polyoxyethylene (4) ether, block copolymers of oxyethylene and oxypropylene, diethylene glycol dioleate, tetrahydrofurfuryl oleate, ethyl oleate, isopropyl myristate, glyceryl monooleate, glyceryl monostearate, glyceryl monoricinoleate, cetyl alcohol, stearyl alcohol, cetylpyridinium chloride, benzalkonium chloride, olive oil, glyceryl monolaurate, corn oil, cotton seed oil, and sunflower seed oil.

In alternative embodiments, a solid dosage form prepared from a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is prepared by a process that includes solvent or dry granulation optionally followed by compression or compaction, spray drying, nano-suspension processing, hot melt extrusion, extrusion/spheronization, molding, spheronization, layering (e.g., spray layering suspension or solution), or the like. Examples of such techniques include direct compression, using appropriate punches and dies, for example wherein the punches and dies are fitted to a suitable tableting press; wet granulation using suitable granulating equipment such as a high shear granulator to form wetted particles to be dried into granules; granulation followed by compression using appropriate punches and dies, wherein the punches and dies are fitted to a suitable tableting press; extrusion of a wet mass to form a cylindrical extrudate to be cut into desire lengths or break into lengths under gravity and attrition; extrusion/spheronization where the extrudate is rounded into spherical particles and densified by spheronization; spray layering of a suspension or solution onto an inert core using a technique such as a convention pan or Wurster column; injection or compression molding using suitable molds fitted to a compression unit; and the like.

Exemplary disintegrants include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium, cross-linked sodium carboxymethylcellulose (sodium croscarmellose), powdered cellulose, chitosan, croscarmellose sodium, crospovidone, guar gum, low substituted hydroxypropyl cellulose, methyl cellulose, microcrystalline cellulose, sodium alginate, sodium starch glycolate, partially pregelatinized starch, pregelatinized starch, starch, sodium carboxymethyl starch, and the like, or a combination thereof.

Exemplary lubricants include calcium stearate, magnesium stearate, glyceryl behenate, glyceryl palmitostearate, hydrogenated castor oil, light mineral oil, sodium lauryl sulfate, magnesium lauryl sulfate, sodium stearyl fumarate, stearic acid, zinc stearate, silicon dioxide, colloidal silicon dioxide, dimethyldichlorosilane treated with silica, talc, or a combination thereof.

The dosage form cores described herein may be coated to result in coated tablets. The dosage from cores can be coated with a functional or non-functional coating, or a combination of functional and non-functional coatings. “Functional coating” includes tablet coatings that modify the release properties of the total composition, for example, a sustained-release or delayed-release coating. “Non-functional coating” includes a coating that is not a functional coating, for example, a cosmetic coating. A non-functional coating can have some impact on the release of the active agent due to the initial dissolution, hydration, perforation of the coating, etc., but would not be considered to be a significant deviation from the non-coated composition. A non-functional coating can also mask the taste of the uncoated composition including the active pharmaceutical ingredient. A coating may comprise a light blocking material, a light absorbing material, or a light blocking material and a light absorbing material.

Exemplary polymethacrylates include copolymers of acrylic and methacrylic acid esters, such as a. an ami nomethacrylate copolymer USP/NF such as a poly(butyl methacrylate, (2 -dimethyl aminoethyl)methacrylate, methyl methacrylate) 1:2:1 (e g., EUDRAGIT E 100, EUDRAGIT EPO, andEUDRAGITE 12.5; CAS No. 24938-16-7); b. a poly(methacrylic acid, ethyl acrylate) 1:1 (e g., EUDRAGIT L30 D-55, EUDRAGIT L100-55, EASTACRYL 30D, KOLLICOAT MAE 30D AND 30DP; CAS No. 25212-88-8); c. a poly(methacrylic acid, methyl methacrylate) 1:1 (e g., EUDRAGIT L 100, EUDRAGIT L 12.5 and 12.5 P; also known as methacrylic acid copolymer, type A NF; CAS No. 25806-15-1); d. a poly(methacrylic acid, methyl methacrylate) 1:2 (e g., EUDRAGIT S 100, EUDRAGIT S 12.5 and 12.5P; CAS No. 25086-15-1); e. a poly(methyl acrylate, methyl methacrylate, methacrylic acid) 7:3:1 (e.g., Eudragit FS 30 D; CAS No. 26936- 24-3); f. a poly(ethyl acrylate, methylmethacrylate, trimethylammonioethyl methacrylate chloride) 1:2:02 or 1:2:0.1 (e g , EUDRAGIT S RL 100, RL PO, RL 30 D, RL 12.5, RS 100, RS PO, RS 30 D, or RS 12.5; CAS No. 33434-24-1); g. a poly(ethyl acrylate, methyl methacrylate) 2:1 (e g., EUDRAGIT NE 30 D, Eudragit NE 40D, Eudragit NM 30D; CAS No. 9010-88-2); and the like, or a combination thereof.

Suitable alkylcelluloses include, for example, methylcellulose, ethylcellulose, and the like, or a combination thereof. Exemplary water based ethylcellulose coatings include AQUACOAT, a 30% dispersion further containing sodium lauryl sulfate and cetyl alcohol, available from FMC, Philadelphia, PA; SURELEASE a 25% dispersion further containing a stabilizer or other coating component (e g., ammonium oleate, dibutyl sebacate, colloidal anhydrous silica, medium chain triglycerides, etc.) available from Colorcon, West Point, PA; ethyl cellulose available from Aqualon or Dow Chemical Co (Ethocel), Midland, MI. Those skilled in the art will appreciate that other cellulosic polymers, including other alkyl cellulosic polymers, can be substituted for part or all of the ethylcellulose.

Other suitable materials that can be used to prepare a functional coating include hydroxypropyl methylcellulose acetate succinate (UPMCAS); cellulose acetate phthalate (CAP); a polyvinylacetate phthalate; neutral or synthetic waxes, fatty alcohols (such as lauryl, myristyl, stearyl, cetyl or specifically cetostearyl alcohol), fatty acids, including fatty acid esters, fatty acid glycerides (mono-, di-, and tri-glycerides), hydrogenated fats, hydrocarbons, normal waxes, stearic acid, stearyl alcohol, hydrophobic and hydrophilic materials having hydrocarbon backbones, or a combination thereof. Suitable waxes include beeswax, glycowax, castor wax, camauba wax, microcrystalline wax, candelilla, and wax -like substances, e g., material normally solid at room temperature and having a melting point of from about 30°C to about 100°C, or a combination thereof.

In other embodiments, a functional coating may include digestible, long chain (e g., C8- C50, specifically C12-C40), substituted or unsubstituted hydrocarbons, such as fatty acids, fatty alcohols, glyceryl esters of fatty acids, mineral and vegetable oils, waxes, or a combination thereof. Hydrocarbons having a melting point of between about 25 °C and about 90 °C may be used. Specifically, long chain hydrocarbon materials, fatty (aliphatic) alcohols can be used.

The coatings can optionally contain additional pharmaceutically acceptable excipients such as a plasticizer, a stabilizer, a water-soluble component (e.g., pore formers), an anti-tacking agent (e.g., talc), a surfactant, and the like, or a combination thereof.

A functional coating may include a release-modifying agent, which affects the release properties of the functional coating. The release-modifying agent can, for example, function as a pore-former or a matrix disrupter. The release-modifying agent can be organic or inorganic, and include materials that can be dissolved, extracted or leached from the coating in the environment of use. The release-modifying agent can comprise one or more hydrophilic polymers including cellulose ethers and other cellulosics, such as hydroxypropyl methylcellulose, hydroxypropylcellulose, hydroxyethylcellulose, methyl cellulose, cellulose acetate phthalate, or hydroxypropyl methylcellulose acetate phthalate; povidone; polyvinyl alcohol; an acrylic polymer, such as gastric soluble Eudragit FS 30D, pH sensitive Eudragit L30D 55, L 100, S 100, or L 100- 55; or a combination thereof. Other exemplary release-modifying agents include a povidone; a saccharide (e.g., lactose, and the like); a metal stearate; an inorganic salt (e.g., dibasic calcium phosphate, sodium chloride, and the like); a polyethylene glycol (e.g., polyethylene glycol (PEG) 1450, and the like); a sugar alcohol (e.g., sorbitol, mannitol, and the like); an alkali alkyl sulfate (e.g., sodium lauryl sulfate); a polyoxyethylene sorbitan fatty acid ester (e.g., polysorbate); or a combination thereof. Exemplary matrix disrupters include water insoluble organic or inorganic material. Organic polymers including but not limited to cellulose, cellulose ethers such as ethylcellulose, cellulose esters such as cellulose acetate, cellulose acetate butyrate and cellulose acetate propionate; and starch can function as matrix disrupters. Examples or inorganic disrupters include many calcium salts such as mono-, di- and tri calcium phosphate; silica and, talc.

The coating may optionally contain a plasticizer to improve the physical properties of the coating. For example, because ethylcellulose has a relatively high glass transition temperature and does not form flexible films under normal coating conditions, it may be advantageous to add plasticizer to the ethylcellulose before using the same as a coating material. Generally, the amount of plasticizer included in a coating solution is based on the concentration of the polymer, e.g., can be from about 1% to about 200% depending on the polymer but is most often from about 1 wt% to about 100 wt% of the polymer. Concentrations of the plasticizer, however, can be determined by routine experimentation.

Examples of plasticizers for ethylcellulose and other celluloses include plasticizers such as dibutyl sebacate, diethyl phthalate, triethyl citrate, tributyl citrate, triacetin, or a combination thereof, although it is possible that other water-insoluble plasticizers (such as acetylated monoglycerides, phthalate esters, castor oil, etc.) can be used.

Examples of plasticizers for acrylic polymers include citric acid esters such as triethyl citrate NF, tributyl citrate, dibutyl phthalate, 1,2-propylene glycol, polyethylene glycols, propylene glycol, diethyl phthalate, castor oil, triacetin, or a combination thereof, although it is possible that other plasticizers (such as acetylated monoglycerides, phthalate esters, castor oil, etc.) can be used.

Suitable methods can be used to apply the coating material to the surface of the dosage form cores. Processes such as simple or complex coacervation, interfacial polymerization, liquid drying, thermal and ionic gelation, spray drying, spray chilling, fluidized bed coating, pan coating, or electrostatic deposition may be used.

In certain embodiments, an optional intermediate coating is used between the dosage form core and an exterior coating. Such an intermediate coating can be used to protect the active agent or other component of the core subunit from the material used in the exterior coating or to provide other properties. Exemplary intermediate coatings typically include water-soluble film forming polymers. Such intermediate coatings may include film forming polymers such as hydroxyethyl cellulose, hydroxypropyl cellulose, gelatin, hydroxypropyl methylcellulose, polyethylene glycol, polyethylene oxide, and the like, or a combination thereof; and a plasticizer. Plasticizers can be used to reduce brittleness and increase tensile strength and elasticity. Exemplary plasticizers include polyethylene glycol propylene glycol and glycerin.

Combination and Alternation Therapy

The compounds or their pharmaceutically acceptable salts as described herein can be administered on top of the current standard of care for COVID patients, or in combination or alternation with any other compound or therapy that the healthcare provider deems beneficial for the patient. The combination and/or alternation therapy can be therapeutic, adjunctive, or palliative.

It has been observed that COVID patients can pass through various stages of disease, and that the standard of care can differ based on what stage of illness the patient presents with or advances to. COVID is noteworthy for the development of “cross-talk” between the immune system and the coagulation system. As the disease progresses, the patient can mount an overreaction by the immune system, which can lead to a number of serious implications, including a cytokine storm. Via the cross-talk between the immune system and the coagulation system, the patient can begin clotting in various areas of the body, including the respiratory system, brain, heart and other organs. Multiple clots throughout the body have been observed in COVID patients, requiring anticoagulant therapy. It is considered that these clots may cause long term, or even permanent damage if not treated and disease alleviated.

More specifically, COVID-19 has been described as progressing through three general stages of illness: stage 1 (early infection), stage 2 (pulmonary phase), and stage 3 (hyperinflammation phase / cytokine storm).

Stage 1 is characterized by non-specific, and often mild, symptoms. Viral replication is occurring, and it is appropriate to begin immediate treatment with the compounds described herein and perhaps in combination or alternation with another anti-viral therapy. Interferon-b may also be administered to augment the innate immune response to the virus. In one embodiment, therefore, a compound of Formula I, Formula II, Formula PI, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is used in an effective amount in combination or alternation with interferon-b and or an additional anti-viral drug. Zinc supplements and or Vitamin C is also sometimes administered at this stage or as the illness progresses.

Stage 2 of COVID-19 is the pulmonary phase where patients may experience acute hypoxemic respiratory failure. In fact, the primary organ failure of COVED-19 is hypoxemic respiratory failure. It has been shown that moderate immunosuppression via a steroid, for example, dexamethasone, can be beneficial to patients with acute hypoxemic respiratory failure and/or patients on mechanical ventilation. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof is used in an effective amount in combination with a corticosteroid which may be a glucocorticoid. Non-limiting examples are budesonide (Entocort EC), bethamethasone, (Celestone), prednisone (Prednisone Intensol), prednisolone (Orapred, Prelone), triamcinolone (Aristospan Intra-Articular, Aristospan Intralesional, Kenalog), methylprednisolone (Medrol, Depo-Medrol, Solu-Medrol), hydrocortisone, or dexamethasone (Dexamethasone Intensol, DexPak 10 Day, DexPak 13 Day, DexPak 6 Day).

The NS5B inhibitor Remdesivir has provided mixed results when given to COVID-19 patients. It can only be administered in a hospital setting, and only by intravenous injection, typically three times a day, which makes it inappropriate for mild to moderate COVID-19 patients. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with Remdesivir to amplify the overall antiviral effect.

Stage 3, the final stage of the disease, is characterized by progressive disseminated intravascular coagulation (DIC), a condition in which small blood clots develop throughout the bloodstream. This stage also can include multi-organ failure (e.g. vasodilatory shock, myocarditis). It has also been observed that many patients respond to this severe stage of COVID-19 infection with a “cytokine storm ” There does appear to be a bi-directional, synergistic relationship between DIC and cytokine storm. To combat DIC, patients are often administered an anti -coagulant agent, which may, for example, be an indirect thrombin inhibitor or a direct oral anticoagulant (“DO AC”). Non-limiting examples are low-molecular weight heparin, warfarin, bivalirudin (Angiomax), rivaroxaban (Xarelto), dabigatran (Pradaxa), apixaban (Eliquis), or edoxaban (Lixiana). In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XTTT or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with anti-coagulant therapy. In some severe cases of clotting in COVID patients, TPA can be administered (tissue plasminogen activator).

It has been observed that high levels of the cytokine interleukin-6 (IL-6) are a precursor to respiratory failure and death in COVID-19 patients. To treat this surge of an immune response, which may constitute a cytokine storm, patients can be administered an IL-6-targeting monoclonal antibody, pharmaceutical inhibitor or protein degrader such as a bispecific compound that binds to IL-6 and also to a protein that mediates degradation. Examples of antibodies include tocilizumab, sarilumab, siltuximab, olokizumab and clazakizumab. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with tocilizumab or sarilumab. Additional nonlimiting examples of immunosuppressant drugs used to treat the overreacting immune system include Janus kinase inhibitors (tofacitinib (Xeljanz)); calcineurin inhibitors (cyclosporine (Neoral, Sandimmune, SangCya)), tacrolimus (Astagraf XL, Envarsus XR, Prograf)); mTOR inhibitors (sirolimus (Rapamune), everolimus (Afinitor, Zortress)); and, IMDH inhibitors (azathioprine (Azasan, Imuran), leflunomide (Arava), mycophenolate (CellCept, Myfortic)). Additional antibodies and biologies include abatacept (Orencia), adalimumab (Humira), anakinra (Kineret), certolizumab (Cimzia), etanercept (Enbrel), golimumab (Simponi), infliximab (Remicade), ixekizumab (Taltz), natalizumab (Tysabri), rituximab (Rituxan), secukinumab (Cosentyx), tocilizumab (Actemra), ustekinumab (Stelara), vedolizumab (Entyvio), basiliximab (Simulect), and daclizumab (Zinbryta)).

IL-1 blocks the production of IL-6 and other proinflammatory cytokines. COVID patients are also sometimes treated with anti-IL-1 therapy to reduce a hyperinflammatory response, for example, an intravenous administration of anakinra. Anti-IL-1 therapy generally may be for example, a targeting monoclonal antibody, pharmaceutical inhibitor or protein degrader such as a bispecific compound that binds to IL-1 and also to a protein that mediates degradation.

Patients with COVID often develop viral pneumonia, which can lead to bacterial pneumonia. Patients with severe COVID-19 can also be affected by sepsis or “septic shock”. Treatment for bacterial pneumonia secondary to COVID or for sepsis includes the administration of antibiotics, for example a macrolide antibiotic, including azithromycin, clarithromycin, erythromycin, or roxithromycin. Additional antibiotics include amoxicillin, doxycycline, cephalexin, ciprofloxacin, clindamycin, metronidazole, sulfamethoxazole, trimethoprim, amoxicillin, clavulanate, or levofloxacin. In one embodiment, thus a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with an antibiotic, for example, azithromycin. Some of these antibiotics such as azithromycin have independent anti-inflammatory properties. Such dmgs may be used both as anti-inflammatory agents for COVID patients and have a treatment effect on secondary bacterial infections.

A unique challenge in treating patients infected with COVID-19 is the relatively long-term need for sedation if patients require mechanical ventilation which might last up to or greater than 5, 10 or even 14 days. For ongoing pain during this treatment, analgesics can be added sequentially, and for ongoing anxiety, sedatives can be added sequentially. Non-limiting examples of analgesics include acetaminophen, ketamine, and PRN opioids (hydromorphone, fentanyl, and morphine). Non-limiting examples of sedatives include melatonin, atypical antipsychotics with sedative- predominant properties (olanzapine, quetiapine), propofol or dexmedetomidine, haloperidol, and phenobarbital. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with a pain reliever, such as acetaminophen, ketamine, hydromorphone, fentanyl, or morphine. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XIII or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with a sedative, such as melatonin, olanzapine, quetiapine, propofol, dexmedetomidine, haloperidol, or phenobarbital. Investigational drugs for COVID-19 include chloroquine and hydroxychloroquine. In one embodiment, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula CIP or a pharmaceutically acceptable salt thereof, is administered in combination or in alternation with chloroquine or hydroxychloroquine.

A protease inhibitor such as lopinavir or ritonavir, previously approved for HIV, may also be administered.

Additional drugs that may be used in the treatment of a COVID patient include, but are not limited to favipiravir, fingolimod (Gilenya), methylprednisolone, bevacizumab (Avastin), Actemra (tocilizumab), umifenovir, losartan and the monoclonal antibody combination of REGN3048 and REGN3051 or ribavirin. Any of these drugs or vaccines can be used in combination or alternation with an active compound provided herein to treat a viral infection susceptible to such.

In one embodiment, a compound of the present invention is used in an effective amount in combination with anti-coronavirus vaccine therapy, including but not limited to mRNA-1273 (Moderna, Inc.), AZD-1222 (AstraZeneca and University of Oxford), BNT162 (Pfizer and BioNTech), CoronaVac (Sinovac), NVX-CoV 2372 (NovoVax), SCB-2019 (Sanofi and GSK), ZyCoV-D (Zydus Cadila), and CoVaxin(Bharat Biotech). In another embodiment, a compound of the present invention is used in an effective amount in combination with passive antibody therapy or convalescent plasma therapy.

SAR.S-CoV-2 is constantly mutating, which many increase virulence and transmission rates. Drug-resistant variants of viruses may emerge after prolonged treatment with an antiviral agent. Drug resistance may occur by mutation of a gene that encodes for an enzyme used in viral replication. The efficacy of a drug against an RNA virus infection in certain cases can be prolonged, augmented, or restored by administering the compound in combination or alternation with another, and perhaps even two or three other, antiviral compounds that induce a different mutation or act through a different pathway, from that of the principle drug.

Alternatively, the pharmacokinetics, bio distribution, half-life, or other parameter of the drug can be altered by such combination therapy (which may include alternation therapy if considered concerted). Since the disclosed purine nucleotides are polymerase inhibitors, it may be useful to administer the compound to a host in combination with, for example a:

(1) Protease inhibitor;

(2) Another polymerase inhibitor;

(3) Allosteric polymerase inhibitor;

(4) Interferon alfa-2a, which may be pegylated or otherwise modified, and/or ribavirin;

(5) Non-substrate-based inhibitor; (6) Helicase inhibitor;

(7) Antisense oligodeoxynucleotide (S-ODN);

(8) Aptamer;

(9) Nuclease-resistant ribozyme;

(10) iRNA, including microRNA and SiRNA;

(11) Antibody, partial antibody or domain antibody to the virus, or

(12) Viral antigen or partial antigen that induces a host antibody response.

EXAMPLES General Methods

'hi, 19 F and 31 P NMR spectra were recorded on a 400 MHz Fourier transform Briicker spectrometer. Spectra were obtained DMSO-dr, unless stated otherwise. The spin multiplicities are indicated by the symbols s (singlet), d (doublet), t (triplet), m (multiplet) and, br (broad). Coupling constants ( ) are reported in Hz. The reactions were generally carried out under a dry nitrogen atmosphere using Sigma-Aldrich anhydrous solvents. All common chemicals were purchased from commercial sources.

The following abbreviations are used in the Examples:

BID: Twice a day DCM: Dichloromethane EtOAc: Ethyl acetate EtOH: Ethanol GT: Genotype

HPLC: High pressure liquid chromatography LD: Loading dose NaOH: Sodium hydroxide NaiSCU: Sodium sulphate (anhydrous)

MeOH: Methanol

NaiSCE: Sodium sulfate

NH4CI: Ammonium chloride

PE: Petroleum ether

Silica gel (230 to 400 mesh, Sorbent) t-BuMgCl: /-Butyl magnesium chloride

THF: Tetrahydrofuran (THF), anhydrous

TP: Triphosphate Example 1. Synthesis of Compound 1A and Compound 2A

Part A: Synthesis of (2R,3R,4R,5R)-5-(2-Amino-6-(methylamino)-9H-purin-9-yl)-4-fl uoro-2-

(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (1-7)

Potassium i-butoxide CbzO F HO' F i-BuOH 1-6 Compound 1-7 Step 5 Step 6

In Step 1, Compound 1-1 is dissolved in DCM and the reaction is cooled to 10 °C before benzyl chloroformate is added followed by NEt 3. The reaction is allowed to cool to room temperature and stir for 12-14 hours. Following appropriate work-up and purification conditions, Compound 1-2 is isolated. In Step 2, Compound 1-2 is dissolved in acetonitrile and cooled to -15 to 5 °C before Morpho DAST is added. The reaction is allowed to stir for 6 hours. Following appropriate work-up and purification conditions, Compound 1-3 is isolated. In Step 3, Compound 1-3 is dissolved in toluene and the reaction is cooled to 0 -10 °C before Red A1 is added. Following appropriate work-up and purification conditions, Compound 1-4 is isolated as the diastereomer with (R)-stereochemistry at the hydroxyl position. In Step 4, Compound 1-4 is dissolved in acetonitrile and cooled to -15 to 5 °C before CBr4 and PPh 3 are added. Following appropriate work-up and purification conditions, Compound 1-5 is isolated. In Step 5, Compound 1-5 is dissolved is acetonitrile and t-BuOH, t-BuOK, and 6-chloro-9H-purin-2-amine are added. The reaction is heated to 40 - 50 °C. Following appropriate work-up and purification conditions, Compound 1-6 is isolated. In Step 6, Compound 1-6 is dissolved in MeOH and MelNTH is added. The reaction is heated to 20 - 30 C. Following appropriate work-up and purification conditions, Compound 1-7 is isolated.

Part B: Synthesis of dihydroquinine salt of isopropyl (hydroxy(phenoxy)phosphoryl)-L- alaninate (1-12)

Phenyl dichlorophosphate (1-8, 150 g, 1.0 eq.) was added into 1300 mL of isopropyl acetate. The solution was cooled to -10°C ± 5°C and then a solution of benzyl alcohol (1-9, 80.6 g, 1.05 eq.) and Et 3 N (86.3 g, 1.2 eq.) was added. The mixture was stirred for 3 hours at -10 ± 5°C. The end point of reaction was monitored by TLC.

L-Alanine isopropyl ester hydrochloride (1-10, 125 g, 1.05 eq.) and Et 3 N (152 g, 2.1 eq.) were added at -10°C ± 5°C. The reaction mixture was stirred at -10 ± 5°C for 2 hours. The end point of reaction was monitored by TLC.

The reaction mixture was filtered, and the filter cake was washed with 20 mL of isopropyl acetate. The filtrate was washed with IN HC1, water, and aqueous sodium bicarbonate. The separated organic layer was dried with anhydrous NazSCL and then concentrated to dryness under vacuum at 40°C-50°C to give 240 g of crude product 1-11 as a diastereomeric mixture (approximately, 1:1). (Pale yellow oil; yield: 89.6% mol/mol; HPLC purity: 83.4% by area; HPLC assay: 86.2% w/w). The product contained around 6%-7% residual benzyl alcohol. The crude 1-4 was used directly in the next step.

Compound 1-11 (135 g, 1.0 eq., 86.2% assay) and quinine (100 g, 1.0 eq.) were added into 650 mL of i-PrOH. After 5% Pd/C (19.2g, 60% water by KF) was added, hydrogenation was performed at 20°C -25°C for 8 hours using a hydrogen bag in a closed system After completion of reaction, the mixture was filtered through a Biichner funnel. The filtrate was concentrated under vacuum to remove the solvent. To the above residue, 300 mL of TBME was added. The mixture was concentrated to remove the solvent under vacuum at 40°C-45°C, and then this step was repeated with another 300 mL of MTBE. To the above, 600 mL of MTBE was added, and the mixture was stirred at 40°C- 45°C for 1 hour and then stirred at 0°C-5°C for additional 1 hour. The mixture was filtered, and the filter cake was washed with 100 mL of MTBE. The cake was dried at 45°C for 16 hours without vacuum to give 152 g of the dihydroquinine salt of isopropyl (hydroxy(phenoxy)phosphoryl)-L- alaninate (1-12, white solid; yield: 69.5% mol/mol; HPLC Purity: 97.91%).

Part C: Synthesis of Compound 1A

The dihydroquinine salt of isopropyl (hydroxy(phenoxy)phosphoryl)-L-alaninate (1-12, 5.9 g, 1.5 eq.), Compound 1-7 (2.0 g, 1.0 eq), DIPEA (0.83 g, 1.0 eq), and HATU (3.65 g, 1.5 eq) were added into 100 mL of dichloromethane. The mixture was heated to 40°C and stirred for 18 hours. The reaction was monitored by TLC and HPLC.

After the reaction was completed, the reaction mixture was cooled to room temperature, washed with IN hydrochloric acid (100 mL x 2), water (100 mL x 2), and 5% aqueous sodium bicarbonate 15 mL x 1). The separated organic phase was dried with 2 g of anhydrous sodium sulfate, filtered, and concentrated at 40°C-45°C under vacuum to give a yellow oil.

Isopropyl acetate (10 mL of) was added. After stirring, the mixture was concentrated under vacuum. Then, 25 mL of isopropyl acetate was added. The mixture was heated to 45°C to afford a clear solution. After stirring at room temperature for 2 hours, the solid precipitate was filtered and dried without vacuum at 45°C for 15 hours to give 2.0 g of crude Compound 1A (yield: 53.8% mol/mol; HPLC purity: 93.1% by area (containing 3.7% of Rp-Compound IB).

The mixture of crude Compound 1A (2.0 g) and 15 mL of isopropyl acetate was heated to 80°C-85°C to afford a solution. The solution was cooled to 20°C-25°C and stirred for 1 hour. The precipitated solid was filtered, washed with isopropyl acetate (1 mL), and dried without vacuum at 50°C for 16 hours to give 1.7 g of Compound 1A (yield: 45.7% mol/mol, HPLC purity: 98.99%). 'H NMR, 19 F NMR, and 31 P NMR spectra confirmed the structure of Compound 1A.

Part D. Synthesis of Hemi-Sulfate Salt Compound 2A

A 250 mL flask was charged with MeOH (151 mL) and the solution was cooled to 0-5 °C. A concentrated solution of H2SO4 was added dropwise over 10 minutes. A separate flask was charged with Compound 1A (151 g) and acetone (910 mL), and the HiSCL/MeOH solution was added dropwise at 25-30 °C over 2.5 hours. A large amount of solid was precipitated. After the solution was stirred for 12-15 hours at 25-30 °C, the mixture was filtered, washed with MeOH/acetone (25 mL/150 mL), and dried at 55-60 °C in vacuum to afford Compound 2A (121 g, 74%). 1 HNMR: (400 MHz, DMSO-rfc): d 8.41 (br, 1H), 7.97 (s, 1H), 7.36 (t, J= 8.0 Hz, 2H), 7.22 (d, J= 8.0 Hz, 2H ), 7.17 (t, J= 8.0 Hz, 1H), 6.73 (s, 2H), 6.07 (d, J= 8.0 Hz, 1H), 6.00 (dd, J = 12.0, 8.0 Hz, 1H), 5.81(br, 1H), 4.84-4.73 (m, 1H), 444-4.28 (m, 3H), 4.10 (t, J = 8.0 Hz, 2H), 3.85-3.74 (m, 1H), 2.95 (s, 3H), 1.21 (s, J= 4.0 Hz, 3H), 1.15-1.10 (m, 9H). Example 2. Synthesis of Select Compounds of the Present Invention

NMR spectra were recorded on a 400 MHz Fourier transform Briicker spectrometer. Spectra were obtained from samples prepared in 5 mm diameter tubes in CDCh, CD 3 OD or DMSO-d6. The spin multiplicities are indicated by the symbols s (singlet), d (doublet), t (triplet), m (multiplet) and, br (broad). Coupling constants (./) are reported in Hz. MS spectra were obtained using electrospray ionisation (ESI) on an Agilent Technologies 6120 quadrupole MS apparatus. The reactions were generally carried out under a dry nitrogen atmosphere using Sigma- Aldrich anhydrous solvents. All common chemicals were purchased from commercial sources.

Synthesis 2. Isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)(pheno xy)phosphoryl)-L- alaninate (Compound 5)

Compound 5 Step 1: N-(6-Chloro-9-((6«/?,#/?,9/?,9«/? -2,2,4,4-tetraisopropyl-9-

((trimethylsilyl)oxy)tetrahydro-6H-furo[3,2-f][l,3,5,2,4] trioxadisilocin-8-yl)-9H-purin-2- yl)acetamide: A solution of intermediate 2-1 (50.2 g, 9225 mmol) in DCM (500 mL) and pyridine (75 ml) was added TMSC1 (30.1 g, 276.74 mmol) at 0 °C and stirred for 0.5 h at 0 °C. To the solution was added acetic anhydride (28.2 g, 276.74 mmol) and the reaction was stirred for 0.5 h at 0 °C and then at room temperature for 1 hour. To the reaction mixture was added LLO and EtOAc. The organic phase was washed with saturated brine, dried over anhydrous Na 2 SC> 4 , and concentrated. The residue was purified by column chromatography (silica gel, EA/PE= 0 to 10%) to afford intermediate 2-2 (53 g, 51%) as a yellow solid.

Step 2: N-(6-Chloro-9-( 6«/?,&R,9/?,9aA -9-hydroxy-2,2,4,4-tetraisopropyltetrahydro-6H- furo[3,2-f [l,3,5,2,4]trioxadisilocin-8-yl)-9H-purin-2-yl)acetamide: To a solution of intermediate 2-2 (53 g, 82.25 mmol) in THF (530 mL) was added p-toluenesulfonic acid monohydrate (5.9 g, 36.67 mmol) at room temperature. The solution was stirred for 1 h at room temperature. Then, TEA (8.3 g, 82.25 mmol) was added and the reaction mixture was concentrated. The residue was purified by column chromatography (silica gel, EA/PE= 10% to 20%) to afford intermediate 2-3 (47 g, 99.5%) as a white solid.

Step 3: N-(6-Chloro-9-( 6a/?,8/?,9a/f -2,2,4,4-tetraisopropyl-9-oxotetrahydro-6H-furo[3,2- f][l,3,5,2,4]trioxadisilocin-8-yl)-9H-purin-2-yl)acetamide: Chromium trioxide (18.9 g, 189 36 mmol) was dispersed in DCM (280 mL), and pyridine (15.0 g, 189.36 mmol) and acetic anhydride (19.3 g, 189.36 mmol) were added dropwise. The reaction mixture turned black. Intermediate 2-3 (37 g, 63.12 mmol) in DCM (380 ml) was added at room temperature and the reaction was stirred for 0.5 h. EtOAc (1200 mL) was added. The reaction mixture was filtered and concentrated, azeotropic with toluene (200 mL). The residue was purified by column chromatography (silica gel, EA/PE= 33% to 100%) to afford intermediate 2-4 (34.8 g, 94%) as a white solid.

Step 4: N-(6-Chloro-9-( 6«/?,#/?,9,V,9rt/? -9-hydroxy-2,2,4,4-tetraisopropyl-9- methyltetrahydro-6H-furo[3,2-f][l,3,5,2,4]trioxadisilocin-8- yl)-9H-purin-2-yl)acetamide: Magnesium chips (6.7 g, 278.66 mmol) were dispersed in anhydrous ether (380 mL), deuterated methyl iodide (9.2 g, 63.33 mmol) was added, and the reaction was stirred for 0.5 h at room temperature before being brought to room temperature. Deuterated methyl iodide (27.5 g, 190.00 mmol) was added dropwise, and the reaction was stirred for 1 h before being brought to below 10 °C and adding intermediate 2-4 (37 g, 63.33 mmol) in DCM (380 ml). The reaction was stirred for 1 hour and then quenched by saturated NaHCCL and extracted with EtOAc. The organic phase was washed with saturated brine, dried over anhydrous Na2S04, and concentrated. The residue was purified by column chromatography (silica gel, EA/PE= 33% to 50%) to afford intermediate 2-5 (22 g,57.9%) as a solid. Step 5: N-(6-Chloro-9-((2/?,3,V, /?,5/?)-3,4-dihydroxy-5-(hydroxymethyl)-3- methyltetrahydrofuran-2-yl)-9H-purin-2-yl)acetamide: To a solution of intermediate 2-5 (22 g,

36.65 mmol) in THF (150 mi .) was added tetrabutylammonium fluoride (23.12 g, 73.30 mmol) in THF (70 ml). The solution was stirred for 1 h at room temperature and concentrated. The residue was purified by column chromatography (silica gel, MeOH/DCM= 2% to 5%) to afford intermediate 2-6 (20 g, >100%) as a solid.

Step 6: (2/?,3/?,4V,5/? -5-(2-Acetamido-6-chloro-9H-purin-9-yl)-2-(acetoxymethyl)-4- hydroxy-4-methyltetrahydrofuran-3-yl acetate: To a solution of intermediate 2-6 (20 g, 55.90 mmol) in dry DCM (400 mL) and pyridine (8 ml) was added acetic anhydride (22.8 g, 223.62 mmol). The solution was stirred overnight at 0 °C. Then, EtOAc was added and the suspension was filtered. The solution was washed brine, dried over anhydrous NaiSCL and concentrated. The residue was purified by column chromatography to afford intermediate 2-7 (8.4 g, 29.8% yield over 3 steps) as a solid.

Step 7: 27?,5 f, ?,5 f -5-(2-Acetamido-6-chloro-9H-purin-9-yl)-2-(acetoxymethyl)-4- fluoro- 4-methyltetrahydrofuran-3-yl acetate: To a solution of intermediate 2-7 (5.4 g, 12.22 mmol) in DCM (220 mL) was added DAST (5.9 g, 36.67 mmol) dropwise at -65 °C and the reaction was stirred for 0.5 h. The reaction was allowed to warm to room temperature and stirred for 1 hours. The reaction was quenched by saturated NaHCCh and separated. The organic phase was washed with saturated brine, dried over anhydrous Na SCri, and concentrated. The residue was purified by column chromatography (silica gel, EA/PE= 50% to 100%) to afford intermediate 2-8 (1.54 g, 30%) as a solid.

Step 8: (2/?,i/?, /?,5/? -5-(2-Amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-2-

(hydroxymethyl)-4-methyltetrahydrofuran-3-ol: A solution of intermediate 2-8 (890 mg, 2.01 mmol) in methylamine (18% in EtOH) (36 mL) in a sealed container was stirred overnight at room temperature and concentrated. Additional sodium methoxide (325.0 mg, 6.02 mmol) was then added before the solution stirred for 1 additional hour at RT. Acetic acid was added to adjust to pEl of 7 and the solution was concentrated. The residue was purified by column chromatography (silica gel, MeOH/DCM= 0 to 2%) to afford intermediate 2-9 (400 mg, 64.5%) as a solid. 'H NMR (400 MHz, CD 3 OD) f¾.057(s, 1H), 6.103(d, J=18.4 Hz, 1H), 4.047-4.015(m, 2H), 3.878-3.845(m, lH), 3.030(s, 3H). 19 F NMR (400 MHz, CD,OD) d 164.35 l(s). MS (ESI) m!z calcd. for CnHwDsFNeCri [M+H] + 316.13; found 316.4.

Step 9: Isopropyl((S)-(( 2/?,J f, /?,5 ?)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro- 3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)(phenoxy)phos phoryl)-L-alaninate: To a solution of intermediate 2-9 (230 mg, 0.736 mmol) and isopropyl ((R)-

(perfhiorophenoxy)(phenoxy)phosphoryl)-L-alaninate (334 mg, 0.736 mmol) in TEIF (4 ml) was added t-BuMgCl (1.7 N in THF) (912 pL, 1.55 mmol) dropwise at -5 °C. The solution stirred for 0.5 h at 0 °C. The reaction mixture was then quenched by saturated NH4CI aqueous solution. The residue was purified by column chromatography (silica gel, MeOH/DCM= 0 to 3.3%) to afford Compound 5 (130 mg, 30.5%) as a solid. ¾NMR (400 MHz, DMSO) d 7.81 (s, 1H), 7.32-7.16 (m, 5H), 6.13-6.08(d, J = 18.8, 8.0 Hz, 1H), 4.87 (s, 1H), 4.50(m, 3H), 4.19(m, 1H), 3.89(m,

1H), 3.02(s, 1H), 1.30-1.27 (m, 3H), 1.17-1.13(m, 9H). 19 F NMR (400 MHz, DMSO) d -163.42(s). MS (ESI) m!z calcd. for C24H30D3FN7O7P [M+H] + 585.22; found 585.5.

Synthesis 3. Isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- f]uoro-4-(fluoromethyl)-3-hydroxytetrahydrofuran-2-yl)methox y)(phenoxy)phosphoryl)-L- alaninate (Compound 6)

Step 1: (6rt/?,#/?,9/?,9rt,y -8-(2-Ainino-6-chloro-9H-purin-9-yl)-2,2,4,4- tetraisopropyltetrahydro-6H-furo[3,2-f|[l,3,5,2,4]trioxadisi locin-9-ol: To a solution of intermediate 3-1 (81.8g, 0.27mol) in pyridine (330 mL) was added TPDSCh (102.6 g, 0.32 mol) drop-wise at 0+5 °C. The solution was stirred for 2h at 0 °C and quenched by water. Then, EtOAc was added, and the phases were separated and dried over anhydrous Na 2 SC> 4 . The solution was concentrated and azeotropic with toluene three times to remove the pyridine. The residue was purified by column chromatography (DCM/MeOH=200: 1-100: 1) to afford intermediate 3-2 (123.3 g, 83.6% yield) as an oil.

Step 2: N-(6-Chloro-9-((6tf/?,#/?,9/?,9tf/?)-2,2,4,4-tetraisopropyl- 9-

((trimethylsilyl)oxy)tetrahydro-6H-furo[3,2-f][l,3,5,2,4] trioxadisilocin-8-yl)-9H-purin-2- yl)isobutyramide: To a solution of intermediate 3-2 (122.6 g 0.225mol) in DCM (1300 mL) and pyridine (184 mL) was added TMSC1 (48.7 g, 0.45mol) drop-wise at 0~5°C. The reaction mixture was stirred for 10 min at 0°C. Then, Isobutyryl chloride (36 g, 0.337 mol) was added and the reaction was stirred for another 10 minutes at 0°C. Water was added, and the phases were separated and washed with CuSCri aqueous solution. The organic phase was washed with brine, dried over anhydrous NaiSCri and concentrated to obtain the crude intermediate 3-3 (151.7 g).

Step 3: N-(6-Chloro-9-( ba/?,&R,9/?,9aA -9-hydroxy-2,2,4,4-tetraisopropyltetrahydro-6H- furo[3,2-f][l,3,5,2,4]trioxadisilocin-8-yl)-9H-purin-2-yl)is obutyramide: To a solution of intermediate 3-3 (151.7 g, 0.22mol) in THF (1500 mL) was added />-toluenesulfonic acid monohydrate (29 g, 0.155mol). The mixture was stirred at room temperature for 10 minutes and then quenched by addition of triethylamine (35 ml). The solution was concentrated and purified by column chromatography (DCM/MeOH=200: 1-100:1) to obtain the intermediate 3-4 (81 g, 58.5% yield over 2 steps).

Step 4: N-(6-Chloro-9-( 6a/?,8/?,9a/? -2,2,4,4-tetraisopropyl-9-oxotetrahydro-6H-furo[3,2- f [l,3,5,2,4]trioxadisilocin-8-yl)-9H-purin-2-yl)isobutyramide : To a solution of intermediate 3-

4 (40 g, 0.065mol) in DCM (300 mL) was added DMP (56 g, 0.13mol) at 0 °C. The reaction mixture was stirred at room temperature overnight. Then, ether (2000 mL) was added and the suspension was filtered. The solution was washed with saturated NaHCCb aqueous solution, saturated Na 2 S 2 0 3 aqueous solution and brine successively. The organic phase was dried over anhydrous Na 2 SC> 4 , concentrated and azeotropic with toluene to obtain the intermediate 3-5, which was used in the next step directly.

Step 5: N-(6-Chloro-9-((¾f//?,#ft , ,9,Y,9«/? -9-hydroxy-2,2,4,4-letraisopropyl-9-

((trimethylsilyl)ethynyl)tetrahydro-6H-furo[3,2-f][l,3,5, 2,4]trioxadisilocin-8-yl)-9H-purin-2- yl)isobutyramide: To a solution of trimethylsilylacetylene (52 g, 0.529 mol) in dry THF (350 ml) was added n-butyllithium (204 ml, 0.51mol) drop-wise at -15— 20°C. The solution was stirred for 30 mins at -15— 20°C. Then, the reaction mixture was cooled down to -70 °C and a solution of intermediate 3-5 (54 g, 0.088mol) in dry DMF (200mL) was added drop-wise. The solution was stirred for 20 mins at -70°C. The resulting solution was slowly warmed up to 0 °C and quenched by addition of saturated NH 4 CI aqueous solution. EtOAc was added, the organic phases were separated and washed with brine, dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-100: 1) to afford intermediate 3-6 (35. lg, 56% yield over 2 steps) as a yellow solid.

Step 6: N-(6-Chloro-9-((¾«/?,#/?,9/?,9«/? -9-fhioro-2,2,4,4-tetraisopropyl-9-

((trimethylsilyl)ethynyl)tetrahydro-6H-furo[3,2-f [l,3,5,2,4]trioxadisilocin-8-yl)-9H-purin-2- yl)isobutyramide: To a solution of intermediate 3-6 (2 g, 2.81mmol) in DCM (20 mL) and pyridine (0.703 g, 8.85mmol) was added DAST (2.27 g, 14.05 mmol) at -70°C. The reaction mixture was slowly warmed up to -30°C and stirred for 5 mins at -30°C. Then, the solution was added into the saturated NaHCCh aqueous solution slowly and DCM was added. The organic phases were separated and washed with brine, dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-100:1) to afford intermediate 3-7 (1.257 g, 62.85% yield) as a white solid.

Step 7: N-(9-((6n/?,/?/?,9/?,9n/? -9-Ethynyl-9-fluoro-2,2,4,4-tetraisopropyltetrahydro-6H- furo[3,2-f] [l,3,5,2,4]trioxadisilocin-8-yl)-6-(methylamino)-9H-purin-2- yl)isobutyramide: To a solution of intermediate 3-7 (14 g, 19.65mmol) in methanol (30 mL) was added 30% methylamine methanol solution (30ml) at 0°C and the reaction was stirred at room temperature for 5 min. The solution was then concentrated to remove the methylamine at RT. Then, the resulting solution was concentrated and purified by column chromatography (DCM/MeOH=200: 1-100:1) to afford intermediate 3-8 (8.8g, 70.4% yield) as a white solid.

Step 8: N-(9-((6tf/?, ?,9/?,9tf/?)-9-Fluoro-2,2,4,4-tetraisopropyl-9-vinyltetrahyd ro-6H- furo[3,2-f] [l,3,5,2,4]trioxadisilocin-8-yl)-6-(methylamino)-9H-purin-2- yl)isobutyramide. To a solution of intermediate 3-8 (8.8 g, 13.86 mmol) in EtOAc (88 mL) was added Lindlar catalyst ( 3.52 g). Then, the solution was stirred overnight under hydrogen atmosphere (0.8 MPa) atRT. The reaction mixture was filtered to remove the catalyst, concentrated, and purified by column chromatography (DCM/MeOH=200: 1-100:1) to afford intermediate 3-9 (7.5g, 85.2% yield) as a solid.

Step 9: N-(9-((2/?,3/?,¥/?,5/? -3-Fluoro-4-hydroxy-5-(hydroxymethyl)-3-vinyltetrahydrofuran - 2-yl)-6-(methylamino)-9H-purin-2-yl)isobutyramide. To a solution of intermediate 3-9 ( 7.5 g,

11.77 mmol) in THF (40ml) was added the solution of TBAF (5.56 g, 17.65 mmol) in THF (35ml) at 0°C. When TLC indicated complete conversion of the starting material, the solution was concentrated and purified by column chromatography (DCM/MeOH=200: 1-10:1) to afford intermediate 3-10 (3.8g, 82.6% yield) as a solid.

Step 10: ((2 f,2 ?, i?,5/?)-3-(Benzoyloxy)-4-fluoro-5-(2-isobutyramido-6-(methyl amino)-9H- purin-9-yl)-4-vinyltetrahydrofuran-2-yl)methyl benzoate. To a solution of compound intermediate 3-10 (3.7g, 9.38mmol) in pyridine (19ml) was added BzCl (2.9 g, 20.64 mmol) at 0°C. The reaction mixture was stirred overnight at RT. Then, the reaction mixture was quenched by methanol and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-100:1) to afford intermediate 3-11 (4.68g, 82.8% yield) as a white solid. Step 11: ((2/?,3/?,4/?,5/?)-3-(Benzoyloxy)-4-((S)-l,2-dihydroxyethyl) -4-fluoro-5-(2- isobutyramido-6-(methylamino)-9H-purin-9-yl)tetrahydrofuran- 2-yl)methyl benzoate. To a solution of intermediate 3-11 (4.68 g, 7.77mmol) in THF (46.8 ml) and H2O (9.36ml) was added NMO ( 1.82 g, 15.53mmol) and OsCri (0.65 g, 2.56mmol. The reaction mixture was stirred overnight at RT. Then, the reaction mixture was quenched by saturated Na 2 S 2 C> 3 aqueous solution and EtOAc was added. The organic phases were separated and washed with brine, dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-50:1) to afford intermediate 3-12 (4g, 82% yield) as a white solid.

Step 12: ((2 /?,.?/?, /?, 5/? -3-(Benzoyloxy)-4-fluoro-4-(hydroxymethyl)-5-(2-isobutyramid o-6- (methylamino)-9H-purin-9-yl)tetrahydrofuran-2-yl)methyl benzoate. Intermediate 3-12 (4 g, 6.28mmol ) was dissolved in methanol (40 ml), THF (11.7 ml) and water (7 ml). Then, sodium metaperiodate (2 g, 9.42 mmol ) was added to the solution and reaction mixture was stirred for 3h at RT. The reaction mixture was filtered and washed with methanol and THF. NaB¾ (0.382 g, 10.05 mmol) was added to the resulting solution in portions and the reaction was stirred for 10 minutes. The reaction mixture was quenched by ice water and EtOAc was added. The organic phases were separated and washed with brine, dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-50:1) to afford intermediate 3-13 (3.17g, 83.2% yield) as a white solid. Step 13: ((2/?,.?/?,4 ?,5/?)-3-(Benzoyloxy)-4-fluoro-4-(fluoromethyl)-5-(2-isobiit yramido-6- (methylamino)-9H-purin-9-yl)tetrahydrofuran-2-yl)methyl benzoate. To a solution of intermediate 3-13 (1000 mg, 1.65mmol) in DCM (10 ml) and pyridine (650 mg, 8.24 mmol) was added trifluoromethanesulfonic anhydride (838 mg, 2.97 mmol) at -15— 20°C. The reaction mixture was stirred for 10 min at -15— 20°C. The solution was quenched by saturated NaHCCL aqueous solution and washed with CuSCL aqueous solution. Then, the organic phase was washed with brine, dried over anhydrous Na2S04 and concentrated. The crude product was dissolved in acetonitrile (10ml) and TBAF (1M, 8.25ml) in THF was added to the resulting solution. The reaction mixture was stirred for 5 min and monitored by TLC. The reaction was then diluted with EtOAc and water. The phases were separated and washed with brine, dried over anhydrous NaiSCU and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-100:1) to afford intermediate 3-14 (113mg, 37.7% yield) as a white solid.

Step 14: (2/?,i/?, /?,5/? -5-(2-Amino-6-(methylamino)-9H-purin-9-yl)-4-fhioro-4-

(fluoromethyl)-2-(hydroxymethyl)tetrahydrofuran-3-ol. Intermediate 3-14 (0.51 g, 0.84 mmol) was dissolved in 20% methylamine methanol solution and stirred overnight at 100°C in a sealed steel reactor. The reaction mixture was concentrated and purified by column chromatography (DCM/MeOH=200: 1-10: 1) to afford intermediate 3-15 (241mg, 87.6% yield ) as a white solid. ¾ NMR (400 MHz, CD 3 OD) d 7.94 (s, 1H), 6.28 (d, J= 20.0 Hz, 1H), 4.84-4.56 (m, 1H), 4.46 (t, J = 24.0Hz, 1H), 4.35-3.89 (m, 1H), 3.86 (m, 2H), 3.30(d, J= 20.0 Hz, 1H), 3.03(s, 3H). 19 F NMR (400 MHz, CD3OD) d 179.67(s), 80.15 (s).

Step 15 : Isopropyl((S)-(((2/?,d/?,4/?,5/? -5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4- fluoro-4-(fluoromethyl)-3-hydroxytetrahydrofuran-2-yl)methox y)(phenoxy)phosphoryl)-L- alaninate. Intermediate 3-15 (150 mg, 0.45mmol) was dissolved in acetonitrile (6 mL), NMI (895.6 mg, 10.80 mmol) was added at RT. The solution was cooled down to 0 °C and a solution of isopropyl (chloro(phenoxy)phosphoryl)-L-alaninate ( 1247.7 mg, 4.05mmol) in acetonitrile (2 mL) was added drop-wise. The reaction mixture was stirred at 0°C for 5 min and monitored by TLC. Then, the resulting solution was quenched by water and diluted with EtOAc. The phases were separated and washed with brine, dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by column chromatography (DCM/MeOH=200: 1-30:1) to afford Compound 6 (lllmg, 40.8% yield) was obtained as a white solid.

¾ NMR (400 MHz, CD 3 OD) d 7.78 (s, 1H), 7.31-7.16 (m, 5H), 6.27(dd, J= 20.0, 8.0 Hz, 1H), 4.91-4.84 (m, 1H), 4.56(m, 1H), 4.51(m, 4H), 4.50(m, 1H), 3.87(dd, J= 20.0, 8.0 Hz, 1H), 3.03 (s, 3H), 1.27-1.13(m, 9H). 19 F NMR (400 MHz, CD 3 OD) d 179.09(s). 31 P NMR (400 MHz, CD 3 OD) d 3.86, 3.79 (d). MS (ESI) m!z calcd. for C 24 H 32 F 2 N 7 O 7 P [M+H] + 600.2; found 600.2. Synthesis 4. Isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- ethynyl-4-fluoro-3-hydroxytetrahydrofuran-2-yl)methoxy)(phen oxy)phosphoryl)-L- alaninate (Compound 7)

Compound 7 NH 2 Step 1: To a solution of intermediate 4-1 (30 mg, 0.09 mmol) in dry THF (2 mL) was added tert- butylmagnesium chloride (1 M in THF) (112 pL, 0.11 mmol) drop-wise at 0 °C. The solution was stirred for 15 mins at 0 °C and for 45 mins at RT. Then, the reaction mixture was cooled down to 0 °C and a solution of isopropyl ((/LV)-(pentafluorophenoxy)-phenoxy-phosphoryl)-/.-alaninate (51 mg, 0.11 mmol) in dry THF (1 mL) was added drop-wise. The resulting solution was slowly warmed up to room temperature and stirred for 15 h. The reaction mixture was then diluted with EtOAc (10 mL) and saturated MLCl aqueous solution (8 mL). The phases were separated and the aqueous layer was back-extracted with EtOAc (2 x 5 mL). The combined organics were washed with saturated MLCl aqueous solution (10 mL) and brine (10 mL), dried overNa 2 S0 4 and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 10%) and by reverse phase column chromatography (C-18 silica, H 2 0/MeOH 0 to 100%). Compound 7 (9 mg, 16%) was obtained as a white solid. ¾NMR (300 MHz, CD 3 OD) d 7.81, 7.79 (s+s, 1H), 7.36-7.14 (m, 5H), 6.26 (d, J= 17.4 Hz, 0.1H), 6.24 (d, J= 17.4 Hz, 0.9H), 4.93-4.89 (overlapped with H 2 0, m, 1H), 4.80-4.78 (m, 1H), 4.53-4.49 (m, 2H), 4.21-4.18 (m, 1H), 3.95-3.84 (m, 1H), 3.23-3.20 (m, 1H), 3.04 (bs, 1H), 1.31-1.14 (m, 9H). 31 P NMR (121 MHz, CD 3 OD) d 4.06 (s), 3.97 (s). MS (ESI) m!z calcd. for C25H32FN7O7P [M+H] + 592.2; found 592.2.

Synthesis 5. Isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- fluoro-3-hydroxy-4-vinyltetrahydrofuran-2-yl)methoxy)(phenox y)phosphoryl)-L-alaninate (Compound 8)

Step 1 : (2/?,i/f, /?,5/?)-5-(2-Amino-6-(methylamino)-9//-puriii-9-yl)-4-fluoro -2-

(hydroxymethyl) -4-vinyltetrahydrofuran-3-ol (3). To a solution of intermediate 4-1 (255 mg, 0.79 mmol) in EtOH (4 mL) was added palladium (5% on BaSCE) (60 mg) and quinoline (2 drops). The solution was put under an atmosphere of H 2 and stirred for 1 h at RT Then, the mixture was filtered on Celite and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 10%). Intermediate 5-1 (177 mg, 70%) was obtained as a white solid. 1 HN R (400 MHz, CD3OD) d 7.98 (s, 1H), 6.12 (d, J = 18.6 Hz, 1H), 5.52-5.35 (m, 2H), 5.25-5.19 (m, 1H), 4.86-4.70 (m, 1H, overlapped with H 2 0), 4.13-3.86 (m, 3H), 3.03 (br. s, 3H). MS (ESI) m!z calcd. for C^HisFNeOi [M+H] + 325.1; found 325.2.

Step 2: (2.S)- Isopropyl 2-(((((2/?,i f,4/?,5/?)-5-(2-amino-6-(melhylamino)-9//-purin-9-yl)-4- vinyl-4-fluoro-3-hydroxytetrahydrofuran-2-yl)methoxy)(phenox y)phosphoryl)amino) propanoate. To a solution of intermediate 5-1 (150 mg, 0.47 mmol) in dry THF (10 mL) was added /erZ-butylmagnesium chloride (1 M in THF) (560 pL, 0.55 mmol) drop-wise at 0 °C. The solution was stirred for 15 mins at 0 °C and for 45 mins at RT. Then, the reaction mixture was cooled down to 0 °C and a solution of isopropyl ((7?,ri)-(pentafluorophenoxy)-phenoxy- phosphoryl)-/.-alaninate (250 mg, 0.55 mmol) in dry THF (5 mL) was added drop-wise. The resulting solution was slowly warmed up to room temperature and stirred for 15 h. The reaction mixture was then diluted with EtOAc (50 mL) and saturated H4CI aqueous solution (40 mL). The phases were separated and the aqueous layer was back-extracted with EtOAc (2 x 25 mL). The combined organics were washed with saturated H4CI aqueous solution (50 mL) and brine (50 mL), dried over Na 2 S0 4 and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 10%) and by reverse phase column chromatography (C-18 silica, H 2 0/MeOH 0 to 100%). Compound 8 (45 mg, 16%) was obtained as a white solid. ¾ NMR (400 MHz, CD 3 OD) d 7.81 (s, 1H), 7.36-7.14 (m, 5H), 6.12 (d, J = 19.1 Hz, 1H), 5.54-5.547 (m, 2H), 5.28-5.23 (m, 1H), 4.91-4.81 (m, 1H, overlapped with H 2 0), 4.58-4.47 (m, 2 H), 4.28-4.22 (m, 1H), 3.95-3.85 (m, 1H), 3.05 (br. s, 3H), 1.32-1.13 (m, 9H). 31 P NMR (162 MHz, CD 3 OD) d 3 86 (s). MS (ESI) m!z calcd. for C 25 H 34 FN 7 O 7 P [M+H] + 594.2; found 594.2.

Synthesis 6. Isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- f]uoro-3-hydroxy-4-(hydroxymethyl)tetrahydrofuran-2-yl)metho xy)(phenoxy)phosphoryl)- L-alaninate (Compound 9) Step 5

Compound 9

Step 1 : (2/?,7/?,4/?,5/?)-5-(2-Aniino-6-(methylamino)-9//-purin-9-yl )-2-(((/i77- butyldiphenylsilyl) oxy)methyl)-4-fluoro-4-vinyltetrahydrofuran-3-ol (5). To a solution of intermediate 5-1 (185 mg, 0.57 mmol) in dry DMF (7 mL) was added imidazole (232 mg, 3.40 mmol) and TDPSC1 (445 pL, 1.70 mmol) at 0 °C. The solution was stirred for 1 h at 0 °C. The reaction mixture was then diluted with EtOAc (50 mL) and saturated NH 4 CI aqueous solution (40 mL). The phases were separated and the organic layer was washed with saturated MLCl aqueous solution (4 x 30 mL), dried over Na 2 SC> 4 and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 5%) to afford intermediate 6-1 (352 mg, 84%) as a white solid. Step 2: (2/?,3/?,¥/?,5/?)-5-(2-Aiiiino-6-(methylamino)-9//-purin-9- yl)-2-(((to7- butyldiphenylsilyl) oxy)methyl)-4-fluoro-4-(hydroxymethyl)tetrahydrofuran-3-ol (6). To a solution of intermediate 6-1 (200 mg, 0 36 mmol) in dioxane (5 mL) was added N- methylmorpholine-/V-oxide (220 mg, 1.87 mmol) and OsC> 4 (4% in ThO) (210 pL). The solution was stirred for 15 h at room temperature in the dark. Then, the mixture was diluted with EtOAc (25 mL) and fdtered on Celite. The solution was washed brine (20 mL), dried over Na 2 SC> 4 and concentrated. The residue was dissolved in DCM (5 mL) and added on NalCri (adsorbed on silica) (2.3 g). The resulting slurry was triturated for 15 h at RT. Then, the silica was fdtered and washed thoroughly with DCM The combined fdtrates were concentrated. The residue was dissolved in EtOH (5 mL) and NaBTL (225 mg, 6.0 mmol) was added at 0 °C. The solution was stirred for 3 h at 0 °C and then quenched with saturated NH4CI aqueous solution (15 mL). The mixture was extracted with EtOAc (2 x 20 mL). The combined organics were washed with brine (10 mL), dried over Na 2 S0 4 and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 5%) to afford intermediate 6-2 (143 mg, 70%) as a white solid.

Step 3: 9-((2/?,T/?,4 ?,3/?)-5-(((tert-Butyldiphenylsilyl)oxy)methyl)-3-fluoro-4-( (tetrahydro- 2//-pyran-2-yl)oxy)-3-(((tetrahydro-2//-pyran-2-yl)oxy)methy l)tetrahydrofuran-2-yl)-/V' i - methyl-9//-purine-2, 6-diamine (7). To a solution of intermediate 6-2 (130 mg, 0.23 mmol) in dry DCM (10 mL) was added 3,4-dihydro-2//-pyran (360 pL, 4.0 mmol) and camphorsulfonic acid (100 mg, 0.43 mmol). The solution was stirred for 2 h at room temperature and diluted with DCM (20 mL). Saturated NaHCCL aqueous solution (20 mL) was added and the phases were separated. The organics were washed with brine (20 mL), dried over Na 2 SC> 4 and concentrated. The residue was purified by column chromatography (silica gel, PE/EtOAc 0 to 40%) to afford intermediate 6-3 (137 mg, 81%) as a white solid.

Step 4: ((2/?,3 ?,¥/?,57?)-5-(2-Amino-6-(methylamino)-9/ -purin-9-yl)-4-fluoro-3-((tetrahydro- 2//-pyran-2-yl)oxy)-4-(((tetrahydro-2//-pyran-2-yl)oxy)methy l)tetrahydrofuran-2- yl)methanol (8). To a solution of intermediate 6-3 (130 mg, 0.18 mmol) in dry THF (5 mL) was added TBAF (1 M in THF) (360 pL, 0.36 mmol). The solution was stirred for 1 h at room temperature and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 5%) to afford intermediate 6-4 (81 mg, 92%) as a white solid.

Step 5: (2A)-Isopropyl 2-(((((2/?,d/?,4/?,5/?)-5-(2-Amino-6-(methylamino)-9//-purin -9-yl)-4- fluoro -3-hydroxy-4-(hydroxymethyl)tetrahydrofuran-2-yl)methoxy)(ph enoxy)phosphoryl) amino) propanoate (9). To a solution of intermediate 6-4 (50 mg, 0.10 mmol) in dry THF (3 mL) was added /e/V-butyl magnesium chloride (1 M in THF) (130 pL, 0.13 mmol) drop-wise at 0 °C. The solution was stirred for 15 mins at 0 °C and for 45 mins at RT. Then, the reaction mixture was cooled down to 0 °C and a solution of isopropyl ((A,ri)-(pentafluorophenoxy)-phenoxy- phosphoryl)-L-alaninate (52 mg, 0.11 mmol) in dry THF (1 mL) was added drop-wise. The resulting solution was slowly warmed up to room temperature and stirred for 15 h. The reaction mixture was then diluted with EtOAc (10 mL) and saturated NH 4 CI aqueous solution (8 mL) The phases were separated and the aqueous layer was back-extracted with EtOAc (2x 5 mL). The combined organics were washed with saturated H 4 CI aqueous solution (10 mL) and brine (10 mL), dried over Na 2 S0 4 and concentrated. The residue was dissolved in HC1 (1.25 M in z-PrOH) (2 mL). The solution was stirred for 2 h at room temperature and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 10%) and by reverse phase column chromatography (C-18 silica, ThO/MeOH 0 to 100%). Compound 9 (29 mg, 49%) was obtained as a white solid. ¾ NMR (400 MHz, CD 3 OD) d 7.80 (s, 1H), 7.36-7.13 (m, 5H), 6.17 (d, J= 18.3 Hz, 1H), 4.92-

4.73 (m, 2H, overlapped with H 2 0), 4.55-4.49 (m, 2 H), 4.26-4.18 (m, 1H), 3.93-3.80 (m, 2H), 3.57-3.43 (m, 1H), 3.03 (br. s, 3H), 1.28 (d, .7= 7.3 Hz, 3H), 1.15 (t, = 6.6 Hz, 6H). 31 P NMR(162 MHz, CD 3 OD) d 3.90 (s). MS (ESI) miz calcd. for C 24 H 34 FN 7 O 8 P [M+H] + 598.2; found 598.2. Synthesis 7. Isopropyl ((((2R,3R,4R,5R)-5-(2-amino-6-(cyclopropylamino)-9H-purin-9- yl)-4- f]uoro-3-hydroxy-4-(hydroxymethyl)tetrahydrofuran-2-yl)metho xy)(phenoxy)phosphoryl)- L-alaninate (Compound 10) Step 1 : (2/?,7/?,4/?,5/?)-5-(2-Aniino-6-(cyclopropyla ino)-9//-purin-9-yl)-4-fluoro-2-

(hydroxymethyl)-4-vinyltetrahydrofuran-3-ol (11). To a solution of intermediate 7-1 (232 mg, 0.67 mmol) in EtOH (4 mL) was added palladium (5% on BaSOf) (50 mg) and quinoline (2 drops). The solution was put under an atmosphere of ¾ and stirred for 1 h at RT. Then, the mixture was filtered on Celite and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 10%) to afford intermediate 7-2 (170 mg, 73%) as a white solid.

Step 2: (2/?,3/?, /?,5/?)-5-(2-Amino-6-(cyclopropylamino)-9//-purin-9-yl)-2-(( (to7- butyldiphenylsilyl)oxy)methyl)-4-fluoro-4-vinyltetrahydrofur an-3-ol (12). To a solution of intermediate 7-2 (168 mg, 0.48 mmol) in dry DMF (5 mL) was added imidazole (200 mg, 291 mmol) and TDPSC1 (375 pL, 1.46 mmol) at 0 °C. The solution was stirred for 1 h at 0 °C. The reaction mixture was then diluted with EtOAc (40 mL) and saturated MLCl aqueous solution (30 mL). The phases were separated and the organic layer was washed with saturated LLCl aqueous solution (4 x 20 mL), dried over Na 2 SC> 4 and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 5%) to afford intermediate 7-3 (254 mg, 89%) as a white solid.

Step 3: (2/?,3/?, /?,5/?)-5-(2-Amino-6-(cyclopropylamino)-9//-purin-9-yl)-2-(( (to7- butyldiphenylsilyl)oxy)methyl)-4-f1uoro-4-(hydroxymethyl)tet rahydrofuran-3-ol (13). To a solution of intermediate 7-3 (250 mg, 0.42 mmol) in dioxane (6 mL) was added N- methylmorpholine-A-oxide (257 mg, 2.19 mmol) and Os0 4 (4% in ¾0) (245 pL). The solution was stirred for 15 h at room temperature in the dark. Then, the mixture was diluted with EtOAc (30 mL) and filtered on Celite. The solution was washed brine (20 mL), dried over Na 2 S0 4 and concentrated. The residue was dissolved in DCM (6 mL) and added on NaI0 4 (adsorbed on silica) (2.7 g). The resulting slurry was triturated for 15 h at RT. Then, the silica was filtered and washed thoroughly with DCM. The combined filtrates were concentrated. The residue was dissolved in EtOH (6 mL) and NaBH4 (263 mg, 7.0 mmol) was added at 0 °C. The solution was stirred for 3 h at 0 °C and then quenched with saturated NH4CI aqueous solution (20 mL). The mixture was extracted with EtOAc (2 x 25 mL). The combined organics were washed with brine (10 mL), dried over Na 2 S0 4 and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 5%) to afford intermediate 7-4 (159 mg, 64%) as a white solid.

Step 4: 9-((2/?,J/?,4/?,5/?)-5-(((/cv/-Butyldiphenylsilyl)oxy)methyl )-3-fluoro-4-((tetrahydro- 2//-pyran-2-yl)oxy)-3-(((tetrahydro-2//-pyran-2-yl)oxy)methy l)tetrahydrofuran-2-yl)-M- cyclopropyl-9/T-purine-2, 6-diamine (14). To a solution of intermediate 7-4 (150 mg, 0.25 mmol) in dry DCM (10 mL) was added 3 ,4-di hydro-2//-py ran (360 pL, 4.0 mmol) and camphorsulfonic acid (100 mg, 0.43 mmol). The solution was stirred for 2 h at room temperature and diluted with DCM (20 mL). Saturated NaHCCL aqueous solution (20 mL) was added and the phases were separated. The organics were washed with brine (20 mL), dried over Na 2 S0 4 and concentrated. The residue was purified by column chromatography (silica gel, PE/EtOAc 0 to 40%) to afford intermediate 7-5 (146 mg, 76%) as a white solid.

Step 5: ((2/?,7/?,4/?,5/?)-5-(2-Amino-6-(cyclopropylamino)-9//-purin -9-yl)-4-fluoro-3-

((tetrahydro-2//-pyran-2-yl)oxy)-4-(((tetrahydro-2//-pyra n-2-yl)oxy)methyl) tetrahydrofuran-2-yl)methanol (15). To a solution of intermediate 7-5 (140 mg, 0 18 mmol) in dry THF (5 mL) was added TBAF (1 M in THF) (360 pL, 0.36 mmol). The solution was stirred for 1 h at room temperature and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 5%) to afford intermediate 7-6 (86 mg, 90%) as a white solid.

Step 6: (2S)-Isopropyl 2-(((((2/?, .?/?, /?, 5/i)-5-(2-am ino-6-(methylamino)-9//-purin-9-yl)-4- fluoro -3-hydroxy-4-(hydroxymethyl)tetrahydrofuran-2-yl)methoxy)(ph enoxy)phosphoryl) amino) propanoate (16). To a solution of intermediate 7-6 (51 mg, 0.10 mmol) in dry THF (3 mL) was added /c/V-butyl magnesium chloride (1 M in THF) (130 pL, 0.13 mmol) drop-wise at 0 °C. The solution was stirred for 15 mins at 0 °C and for 45 mins at RT. Then, the reaction mixture was cooled down to 0 °C and a solution of isopropyl ((A%)-(pentafluorophenoxy)-phenoxy- phosphoryl)-L-alaninate (52 mg, 0.11 mmol) in dry THF (1 mL) was added drop-wise. The resulting solution was slowly warmed up to room temperature and stirred for 15 h. The reaction mixture was then diluted with EtOAc (10 mL) and saturated NH 4 CI aqueous solution (8 mL). The phases were separated and the aqueous layer was back-extracted with EtOAc (2x 5 mL). The combined organics were washed with saturated MLCl aqueous solution (10 mL) and brine (10 mL), dried over Na 2 S0 4 and concentrated. The residue was dissolved in HC1 (1.25 M in /-PrOH) (2 mL). The solution was stirred for 2 h at room temperature and concentrated. The residue was purified by column chromatography (silica gel, DCM/MeOH 0 to 10%) and by reverse phase column chromatography (C-18 silica, H 2 0/MeOH 0 to 100%). Compound 10 (25 mg, 41%) was obtained as a white solid. 1 H NMR (400 MHz, CD 3 OD) d 7.81 (s, 1H), 7.33-7.14 (m, 5H), 6.19 (d, J = 18.3 Hz, 1H), 4.93- 4.81 (m, 2H, overlapped with H 2 0), 4.53-4.50 (m, 2 H), 4.24-4.20 (m, 1H), 3.92-3.80 (m, 2H), 3.55-3.44 (m, 1H), 2.91-2.88 (br. m, 1H), 1.27 (d, J= 7.2 Hz, 3H), 1.15 (t, J= 7.0 Hz, 6H), 0.86- 0.81 (m, 2H), 0.61-0.57 (m, 2H). 31 P NMR (162 MHz, CD 3 OD) d 3.85 (s). MS (ESI) m/z calcd. for C 26 H 36 FN 7 O 8 P [M+H] + 624.2; found 624.3. Synthesis 8. Isopropyl ((R)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- fluoro-3-hydroxy-4-(trifluoromethyl)tetrahydrofuran-2-yl)met hoxy)(phenoxy)phosphoryl)-

L-alaninate (Compound 11B)

8-1 ep -

Cl

CIBzO^°r / <' 1 7 ^ °' .N'^

, CIBzO ' N ^ N H 2

\ — r o c H . ^Q: CF 3

' ' ^ CF3 Step 4 CIBzO CIBz0 ’ —Z b*rcf 3

CIBzO' F " F " Step 5

8-5 CIBzO' F 8-6

8-4

^ CH

HN > 3

NH

N

NH l 2

Step 6 ^° N^ N ^ N H 2 - _

) \_r Step 7 , - CF 3

- — HO' 'F

' CFS 0-7 Compound 11 B

Step 1: To a 3-neck round bottom flask was charged with anhydrous tetrahydrofuran (100 mL) and LDA (25 mL, 50 mmol). The mixture was stirred and cooled to -75°C. To this mixture was then slowly added intermediate 8-1 (8.7 g, 50mmol) while maintaining the batch temperature below - 74°C. The mixture was stirred at -76°C for 60 minutes and a solution of freshly distilled D- glyceraldehyde (6.5 g, 50 mmol) in anhydrous THF (20mL) was slowly added while maintaining batch temperature below -74°C. After the addition, the mixture was stirred for approximately 60 minutes and 100 g of 20% MLCl solution was added. The mixture was slowly warmed to ambient temperature and transferred to a separatory funnel. The aqueous phase was separated and extracted with dichloromethane. The organic phases were combined, dried over MgSCL, filtered, and concentrated to afford crude intermediate 8-2 and the residue was purified by column chromatography to afford the product as a yellow oil (7.3g, 50% yield). 'H MR (CDCh, 400 MHz): d (ppm) 1.33-1.43 (m, 9H), 4.06-4.42 (m, 6H); 19 F NMR (CDCb, 376.5 MHz) d (ppm) - 73.83 (s, lF),-73.00(t, 3F).

Step 2: A mixture of intermediate 8-2 (3.0 g, lOmmol), 20 g EtOH, and 2 g of 12% sulfuric acid was refluxed at 78°C for 5 hours. The mixture was cooled to ambient temperature and 1 g of triethylamine was added to neutralize the acid. The mixture was concentrated to dryness. The residue was mixed with 20 g of toluene and the mixture was again concentrated to dryness. The residue was dissolved in 15 g of acetonitrile. To the solution was added a catalytic amount of 4- dimethylaminopyridine (DMAP) and 4-chlorobenzoylchloride (5g, 36mmol) at room temperature. The mixture was cooled in an ice water bath and then triethylamine (7.3g, 72mmol) was added. The mixture was stirred at room temperature overnight. The reaction was quenched by adding water (60 mL) and the resulting solution was concentrated under reduced pressure. The residue was diluted with ethyl acetate (100 mL), washed with water, and brine (each 2 c 50mL). The organic layer was concentrated under reduced pressure and the residue was purified by column chromatography to afford intermediate 8-3 as a pale yellow solid (2.5g, 50% yield). ¾ NMR (CDCb, 400 MHz) d (ppm) 4.60-4.68 (m, 1H), 4.75-4.84 (m, 1H), 4.92-5.00(m, 1H), 6,06-6.16(m, lH),7.40-7.53(m, 4H),7.91-7.98(m, 4H); 19 F NMR (CDCb, 376.5 MHz) d (ppm) -78.63 (s, lF),-74.74(t,3F).

Step 3: A dried round bottom flask was charged with intermediate 8-3 (5g, lOmmol) and the solid was dissolved in anhydrous THF (50mL). The solution was cooled to -20° C. Lithium tri-tert- butoxyaluminumy solution (1.0 M in THF) (17mL, 17mmol) was then added via addition funnel for 20 minutes and the resulting mixture was stirred for 1 hour at -20°C. Ethyl acetate (12 mL) was added and the mixture was allowed to warm slowly to 0 ° C. A saturated aqueous solution of ammonium chloride (4.5mL) was then added and the mixture was concentrated in vacuo and diluted with EtOAc (lOOmL). Aqueous HC1 (3N, 30 mL) was added to dissolve the solids. After the phase separation, the organic layer was washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure to give a solid. The residue solid was purified by column chromatography to afford intermediate 8-4 as a white solid (3.5g, 70% yield). 'H NMR (CDCb, 400 MHz) d (ppm) 4.65-4.74 (m, 3H), 5.57-6.17 (m, 2H), 7.31-7.47(m, 4H),7.91-8.01(m, 4H); 19 F NMR (CDCb, 376.5 MHz) d (ppm) -79.25 (d, lF),-75.14(d,lF),-74.88(d,lF),-72.20(d,lF).

Step 4: Under nitrogen, intermediate 8-4 (1.5 g, 3mmol) was dissolved in dichloromethane (18 ml) at -20°C and PPI13 (1.5 g, 4.5 mmol) was added. The reaction mixture was stirred for 15 minutes and CBr4 (2.4 g, 4.5mmol) was added portion-wise. The reaction was stirred for 1.5 hours between -20° C and -15° C. The reaction mixture was purified (without work-up and concentration) by chromatography on a silica gel column to afford intermediate 8-5 (1.18 g,70% yield). 'H NMR (CDCb, 400 MHz) d (ppm) 4.58-4.85 (m, 3H), 5.75-5.78(m, 0.5H),6.41-6.70(m, 1.4 H), 7.38- 7.47(m, 4H),7.94-8.04(m, 4H); 19 F NMR (CDCb, 376.5 MHz) d (ppm) -79.27 (d, 1F),- 75.23(d,lF),-72.81(d,lF),-70.46(d,lF).

Step 5: A three-neck round-bottomed flask was charged with 6-chloro-2-aminopurine (1.1 g,6.5 mmol) followed by anhydrous tBuOH (45 mL) with stirring. To the above stirred solution was added potassium tert-butoxide (1.5 g, 7mmol) portion-wise at room temperature. After 30 min, a solution of intermediate 8-5 (l.lg, 2 mol) in anhydrous acetonitrile (4 mL) was added at room temperature. The mixture was slowly heated to 50° C and stirred for 22 h. Saturated aqueous solution of ammonium chloride (4.5mL) was added and the solution was diluted with ethyl acetate (60 mL), and washed with water and brine (each 2 c 30mL). The organic layer was concentrated under reduced pressure and the residue was purified by column chromatography to afford the intermediate 8-6 (0.5 g, 40% yield).

Step 6: A 5 mL flask was charged with methanamine (3 mL, 30% in methanol) and stirred at 10±5° C. Intermediate 6 (325mg, 0.5mmol) was added in batches at 20±5° C and the reaction was stirred for 1 hour to obtain a clear solution. The reaction was stirred for an additional 6-8 hours, at which point TLC indicated that the intermediate was less than 1% of the solution. The reactor was charged with solid NaOH (0.2 g), stirred for 30 minutes and concentrated The resulting residue was purified by column chromatography (DCM:MeOH=50:l to 20:1) to afford intermediate 7 (120 mg, 65% yield).

Step 7: A solution of intermediate 8-7 (110 mg, 0.3 mmol) and (S)-2-[((S)-(2,3,4,5,6- pentafluorophenoxyXphenoxy) phosphoryl)- amino]propionic acid isopropyl ester (150 mg, 0.33 mmol) were suspended in THF (2 mL) and stirred under nitrogen. The suspension was then cooled to a temperature below -15° C and a 1.7 M solution of t-BuMgCl solution (0.5 mL, 0.85 mool) was slowly added while a temperature of -15 to -10° C was maintained. The reaction was stirred for an additional 16 hours, at which point TLC indicated that the intermediate was less than 5% of the solution. A saturated aqueous solution of ammonium chloride (4.5mL) was added to the suspension at room temperature, and the solution was diluted with ethyl acetate (60 mL), and washed with water and brine (each 2 c 30mL). The organic layer was concentrated under reduced pressure and the residue was purified by column chromatography (DCM:MeOH=150:l to 50:1) to afford the Compound 11B (lOOmg, 53% yield). ^NMR^DCb, 400 MHz) d (ppm) 1.71-1.18 (m,6H), 1.29- 1.32(m,3H), 3.05(s,3H), 3.87-3.91(m,lH), 4.11-4.12 (bs,lH), 4.37-4.40 (m,2H), 4.70-4.77 (m, 1H), 4.87-4.95(m,lH), 6.45-6.49(d,lH), 7.20-7.38(m, 5H), 7.73(d, 1H); 19 F NMR (CDCh, 376.5 MHz) d (ppm) -178.22 (s, lF),-76.37(d,3F).

Synthesis 9. Isopropyl ((S)-(((2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4- difluoro-3-hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phos phoryl)-L-alaninate Isopropyl ((S)-(((2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4-difluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-a laninate can be synthesized as described in in Hertel et al. J. Org Chem. 1988, 53, 2406 and Example 1. A non-limiting example is described below:

Using Reformatskii conditions, ethyl 2-bromo-2,2-difluoroacetate is coupled to (R)-2,2- dimethyl-l,3-dioxolane-4-carbaldehyde I the presence of activated Zinc to afford ethyl (3R)-3-(2,2- dimethyl-l,3-dioxolan-4-yl)-2,2-difluoro-3-hydroxypropanoate , which is then subjected to hydrolytic removal of the isopropylidine group and lactone closure as described in Hertel et al. Following protection with a CBz group, (2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4-difluoro-2-(hydroxymethyl)tetrahydrofuran-3-ol can be synthesized using the same procedures as described in Example 1, Part A, Steps 3-6.

(2R,3R,5R)-5-(2-Amino-6-(methylamino)-9H-purin-9-yl)-4,4- difluoro-2- (hydroxymethyl)tetrahydrofuran-3-ol is then coupled to the dihydroquinine salt of isopropyl (hydroxy(phenoxy)phosphoryl)-L-alaninate (1-12) as described in Example 1, Part C to afford isopropyl ((S)-(((2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4-difluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-a laninate:

Synthesis 10. Isopropyl ((S)-(((2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4- dichloro-3-hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phos phoryl)-L-alaninate

Isopropyl ((S)-(((2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4-dichloro-3- hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-a laninate can be synthesized as described in Pinho et al. J. Org. Chem. 2017, 27, 3468 and Example 1. A non-limiting example is described below:

Isopropyl 2,2,2-trichloroacetateis is coupled to (R)-2, 2-dimethyl- 1,3 -dioxolane-4- carbaldehyde in the presence of Turbo Grignard to afford ethyl (3 R)-3 -(2, 2-dimethyl- 1,3 -dioxolan- 4-yl)-2,2-dichloro-3-hydroxypropanoate, which is then subjected to acetic conditions and lactone closure as described in Pinho et al. Following protection with a CBz group, (2R,3R,5R)-5-(2-amino- 6-(methylamino)-9H-purin-9-yl)-4,4-difluoro-2-(hydroxymethyl )tetrahydrofuran-3-ol can be synthesized using the same procedures as described in Example 1, Part A, Steps 3-6.

(2R,3R,5R)-5-(2-Amino-6-(methylamino)-9H-purin-9-yl)-4,4- dichloro-2- (hydroxymethyl)tetrahydrofuran-3-ol is then coupled to the dihydroquinine salt of isopropyl (hydroxy(phenoxy)phosphoryl)-L-alaninate (1-12) as described in Example 1, Part C to afford isopropyl ((S)-(((2R,3R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)- 4,4-dichloro-3- hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-a laninate:

Synthesis 11. Isopropyl ((S)-(((2R,3R,4S,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4- chloro-4-fluoro-3-hydroxytetrahydrofuran-2-yl)methoxy)(pheno xy)phosphoryl)-L-alaninate

(2R,3R,4R,5R)-5-(2-Amino-6-(methylamino)-9H-purin-9-yl)-4 -chloro-4-fluoro-2- (hydroxymethyl)tetrahydrofuran-3-ol can be synthesized as described in US 20150175648 and the Example 1. A non-limiting example is described below: f-BuOH

(3S,4R)-3-hydroxy-4-(hydroxymethyl)cyclopentan-l-one is first CBz-protected to afford benzyl (((lR,2S)-2-(((benzyloxy)carbonyl)oxy)-4-oxocyclopentyl)meth yl) carbonate and then subjected to NFSI (N-fluorobenzenesulfonimide) to afford benzyl (((lR,2R,3S)-2-

(((benzyloxy)carbonyl)oxy)-3-fluoro-4-oxocyclopentyl)meth yl) carbonate, an intermediate that is then subjected to NCS (N-chlorosuccinimide) to afford benzyl (((lR,2R,3S)-2-

(((benzyloxy)carbonyl)oxy)-3-fluoro-4-oxocyclopentyl)meth yl) carbonate as described in U.S. Patent ‘648.

(2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4 -chloro-4-fluoro-2- (hydroxymethyl)tetrahydrofuran-3-ol is then coupled to the dihydroquinine salt of isopropyl (hydroxy(phenoxy)phosphoryl)-L-alaninate (1-12) as described in Example 1, Part C to afford isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)-4-chloro-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-a laninate:

1-12

In one embodiment, the fluorination reaction affords a mixture of “a-fluoro” and “b-fluoro” lactone derivatives and the compounds are separated by conventional methods known to a skilled artisan, for example, column chromatography or crystallization, to isolate the two diastereomers. In this embodiment, both diastereomers are carried forward to afford the “a-fluoro” and “b-fluoro” final products:

In an alternative embodiment, the fluorination reaction is conducted with A'-fluoro-o- benzenedisulfonimide (NFOBS) or Selectfluor.

Example 3. Activity of Compound 1A against Coronavirus in Huh7 cells

The activity of Compound 1A was tested against the human coronaviruses alpha-229E and beta-OC43 in Huh7 cells.

Compound 1A

Huh7 cells were seeded in 96-well plates at a concentration that yielded 80-100% confluent monolayers in each well after overnight incubation. Compound 1A was dissolved in DMSO to 10 mg/mL and 8 half-log serial dilutions in test medium (modified Eagle’s medium containing 5% fetal bovine serum and 50 pL gentamicin) were prepared with the highest concentration of 50 pg/mL. 100 pL of each concentration were added to 5 test wells on the 96-well plate and 3 wells were infected with test virus in test medium (<100 CCID50 per well). An equivalent amount of test medium was added to the remaining test wells to assess toxicity to uninfected cells. Six wells were infected to serve as untreated virus controls. Media only was added to 6 wells to serve as cell controls. Plates were incubated at 37°C in a humidified 5% CO2 atmosphere until cytopathic effect (CPE) was observed microscopically.

To obtain the CPE endpoint, wells were stained with 0.011% neutral red dye for approximately 2 hours. The dye was siphoned off and wells were rinsed once with phosphate- buffered saline to remove residual, unincorporated dye. 200 pL of 50:50 Sorensen citrate buffer/ethanol was added for >30 min with agitation and then light absorbance at 540 nm was measured on a spectrophotometer.

To obtain the virus yield reduction (VYR) endpoint, supernatant fluid from 3 replicate wells of each compound concentration were pooled and virus titer was measured using a standard endpoint dilution CCID50 assay and titer calculations using the Reed Muench (1948) equation (Reed, LJ and Muench, H. Am. J. Hygiene 27:493-497 (1948)). The concentration of compound required to reduce virus yield by 1 logio (EC90) was determined using regression analysis.

As shown in Table 1, Compound 1A is potent against both the alpha-229E coronavirus and the beta-OC43 coronavirus. Compound 1A exhibits an EC90 value of 0.71 pM against alpha-229E in the virus yield reduction assay and an EC90 value of 0.29 pM against beta-OC43. Additionally, Compound 1A exhibits high CC50 values and selectivity indexes (SI) against both the alpha and beta coronaviruses. For example, against the beta coronavirus, Compound 1A has a selectivity index of greater than 170 when measured using the viral yield reduction assay and a CC50 value of greater than 50 pM when measured in neutral red assay. Table 1. Activity of Compound 1A against Coronaviruses Alpha-229E and Beta-OC43

Visual and neutral red SI: CC50/EC50 VYR SI: CC 5 O/EC9O NT : not tested

Example 4. Activity of Compound 1A and IB against Coronavirus in BHK-21 and MES-21 cells

Compound 1A and Compound IB were tested for activity against human coronavirus in BHK-21 cells (Table 2A and Table 2B) and MES-1 cells (Table 3A and Table 3B). The ECso and the CC50 was determined and compared to Sofosbuvir.

Compound activity against coronavirus was based on inhibition of virus induced cytopathogenicity acutely infected with a multiplicity of infection (m.o.i.) of .01. After a 3-day incubation at 37 °C cell viability was determined by the MTT method as described by Pauwels et al (J. Virol. Methods 1988, 20, 309-321).

To determine the cytotoxicity, cells were seeded at an initial density of 1 x 106 cells/mL in 96 well plates containing Minimum Essential Medium with Earles’ s salts (MEM-E), L-glutamine, 1 mM sodium pyruvate and 25 mg/L kanamycin, supplemented with 10% fetal bovine serum. Cell cultures were then incubated at 37 °C in a humidified 5% CO2 atmosphere in the absence or presence of serial dilutions of test compounds. Cell viability was determined by the MTT method.

Table 2A. Activity of Select Compounds against HCoV in BHK-21 Cells a Compd cone. (mM) required to reduce the viability of mock infected BHK cells by 50% as determined by the MTT method after 3 days of incubation

¾ Compd cone. (mM) required to achieve 50% protection of BHK cells from virus-induced cytopathogenicity as determined by the MTT method at day 3 post-infection

Table 2B. Activity of Select Compounds against HCoV in BHK-21 Cells a Compd cone. (mM) required to reduce the viability of mock infected BHK cells by 50% as determined by the MTT method after 3 days of incubation

¾ Compd cone. (mM) required to achieve 50% protection of BHK cells from virus-induced cytopathogenicity as determined by the MTT method at day 3 post-infection

Table 3A. Activity of Select Compounds against HCoV in MES-1 Cells c Compd cone. (mM) required to reduce viability of mock infected MES-1 cells by 50% as determined by the MTT method after 3 days of incubation d Compd cone. (mM) required to achieve 50% protection of MES-1 cells from virus-induced cytopathogenicity as determined by the MTT method at day 3 post-infection

Table 3B. Activity of Select Compounds against HCoV in MES-1 Cells c Compd cone. (mM) required to reduce viability of mock infected MES-1 cells by 50%, as determined by the MTT method after 3 days of incubation. d Compd cone. (mM) required to achieve 50% protection of MES-1 cells from virus-induced cytopathogenicity as determined by the MTT method at day 3 post-infection

Example 5. Activity of Compound 1A against SARS-CoV and SARS-CoV-2 Compound 1A was tested against SARS-CoV in Huh7 cells and SARS-CoV-2 in differentiated normal human bronchial epithelial (dNHBE, also referred to as HAE (human airway epithelial)) cells and the results are provided in Table 4. The CC50 was determined using the neutral red assay and the EC90 and SI were determined using the virus yield reduction assay. The EC90 is provided in pg/mL and mM. Compound 1A exhibits an EC90 of 0.34 mM against SARS-CoV and an EC90 of 0.64 mM against SARS-CoV-2. Table 4. Activity of Compound 1A Against SARS-CoV and SARS-CoV-2

^Cso was estimated by visual inspection of the cells

2 Value represents the mean of two replicate EC90 determinations, 0.33 and 0.41 pg/mL

The activity of Compound 1A was evaluated in Huh-7 cells infected with SARS-CoV (Urbani) in a neutral red (NR) assay to assess cytotoxicity and then tested using a virus yield reduction (VYR) assay to assess antiviral activity.

Neutral red assay: Compound 1A was dissolved in 100% DMSO at a concentration of 10 mg/mL and serially diluted using eight half-log dilutions in test medium (Minimum Essential Medium supplemented with 5% FBS and 50 pg/mL gentamicin). The starting (high) test concentration was 50 pg/mL. Each dilution was added to 5 wells of a 96-well plate with 80-100% confluent Huh7 or RD cells (hCoY beta OC43 only). Three wells of each dilution were infected with virus and two wells remained uninfected as toxicity controls. Six untreated wells were infected as virus controls and six untreated wells were left uninfected to use as cell controls. Viruses were diluted to a specific 50% cell culture infectious dose (CCID50) per mL to achieve the lowest possible multiplicity of infection (MOI) that would yield >80% toxicity within 5-7 days. The MOI was 0.03 CCIDso/cell. Plates were incubated at 37±2°C, 5% C02.

On day 7 post-infection (p.i ), the plates were stained with neutral red dye for approximately 2 hours (+15 minutes). Supernatant dye was removed, wells were rinsed with PBS, and the incorporated dye was extracted in 50:50 Sorensen citrate buffer/ethanol for >30 minutes and the optical density was read on a spectrophotometer at 540 nm. Optical densities were converted to percent of cell controls and the concentration of Compound 1A required to cause 50% cell death in the absence of virus was calculated (CC 50 ). The selective index (SI) is the CC 50 divided by the EC50.

Virus yield reduction assay: Vero76 cells were seeded in 96-well plates and grown overnight (37°C) to 80% confluency. A sample of the supernatant fluid from each compound concentration was collected on day 3 post-infection (3 wells pooled) and tested for virus titer using a standard endpoint dilution CCID50 assay and titer calculations using the Reed-Muench (1948) equation (Reed, LJ and Muench, H. Am. J Hygiene 27:493-497 (1948)). The concentration of compound required to reduce virus yield by 1 log 10 (EC90) was calculated by regression analysis.

The antiviral activity of Compound 1A was next evaluated against SARS-CoV-2 (WA1) using differentiated normal human bronchial epithelial (dNHBE; HAE (human airway epithelial) cells made to order by MatTek Corporation (Ashland, MA).

Cell Culture: dNHBE cells were grown on 6mm mesh disks and arrived in kits with either 12- or 24-well transwell inserts. During transportation the tissues were stabilized on a sheet of agarose, which was removed upon receipt. One insert was estimated to consist of approximately 1.2 x 106 cells. Kits of cell inserts (EpiAirwayTM AIR-100, AIR-112) originated from a single donor, # 9831, a 23 -year old, healthy, non-smoking, Caucasian male. The cells have unique properties in forming layers, the apical side of which is exposed only to air and that creates a mucin layer. Upon arrival, the cell transwell inserts were immediately transferred to individual wells of a 6-well plate according to manufacturer’s instructions, and 1 mL of MatTek’ s proprietary culture medium (AIR-100-MM) was added to the basolateral side, whereas the apical side was exposed to a humidified 5% CO2 environment. Cells were cultured at 37°C for one day before the start of the experiment. After the 24 hour equilibration period, the mucin layer, secreted from the apical side of the cells, was removed by washing with 400 pL pre-warmed 30 mM HEPES buffered saline solution 3X. Culture medium was replenished following the wash steps.

Viruses: Virus was diluted in AIR-IOO-MM medium before infection, yielding a multiplicity of infection (MOI) of approximately 0.0015 CCID50 per cell.

Experimental design: Each compound treatment (120 pL) and virus (120 pL) was applied to the apical side. At the same time, the compound treatment (1 mL) was applied to the basal side for a 2-h incubation. As a virus control, some of the cells were treated with placebo (cell culture medium only). Following the 2-h infection, the apical medium was removed, and the basal side was replaced with fresh compound or medium (1 mL). The cells were maintained at the air-liquid interface. On day 5, cytotoxicity (CC50 values) in the placebo-treated inserts was estimated by visual inspection, and the medium was removed from all inserts and discarded from the basal side. Virus released into the apical compartment of the dNHBE cells was harvested by the addition of 400 pL of culture medium that was pre-warmed at 37°C. The contents were incubated for 30 minutes, mixed well, collected, thoroughly vortexed and plated on Vero 76 cells for VYR titration. Duplicate wells were used for virus control and cell controls.

Determination of virus titers from each treated cell culture: Vero 76 cells were seeded in 96-well plates and grown overnight (37°C) to confluence. Samples containing virus were diluted in 10-fold increments in infection medium and 200 pL of each dilution transferred into respective wells of a 96-well microtiter plate. Four microwells were used for each dilution to determine 50% viral endpoints. After 5 days of incubation, each well was scored positive for virus if any cytopathic effect (CPE) was observed as compared with the uninfected control and counts were confirmed for endpoint on days 6 and 7. The virus dose that was able to infect 50% of the cell cultures (CCID50 per 0.1 mL) was calculated by the Reed-Muench method (1948) (Reed, LJ and Muench, H. Am. J. Hygiene 27:493-497 (1948)) and the 90% effective concentration (EC90; concentration to reduce virus yield by 1 log 10) was determined by regression analysis. The day 5 values were reported. Untreated, uninfected cells were used as the cell controls.

Example 6. In Vitro Activity of Compound 1A and other Oral Antiviral Drugs Against Various Human Coronaviruses

Compound 1A and other oral antiviral drugs were tested against various human coronaviruses (Table 5) in various cell lines. The data demonstrate the potent in vitro activity of Compound 1A against several CoVs, with individual EC90 values ranging from 0.34 to 1.2 m\1 against HCoV-229E, HCoV-OC43, SARS-CoV-1 and SARS-CoV-2 and less activity against MERS-CoV (average EC 90 = 36 mM).

Table 5. Activity of Compound 1A and Other Oral Antiviral Drugs Against Human

Coronaviruses a Average of 2 experiments (1.6 and 2.0 mM) ¾ EC 5O determined by dye staining (virus yield reduction substantially overestimates antiviral potency of cytotoxic compounds) C CC 50 /EC 5O d Not determined (no cytopathic effect with this virus in this cell line) e Cytotoxicity assessed by visual inspection of cell monolayers f Average of two replicates (0.57 and 0.70 mM) g Average of two replicates (0.52 and 0.42 mM) h Average of two replicates (4.7 and 3.1 pM)

BHK-21, baby hamster kidney cell line

Huh-7, human hepatocyte carcinoma cell line (established ability to form triphosphate from Compound 1A)

RD, human rhabdomyosarcoma cell line (unknown ability to form triphosphate from Compound 1A)

HAE, human airway epithelial cell culture (established ability to form triphosphate from Compound 1A) (established ability to form triphosphate from Compound 1A)

In an initial screening, BHK-21 cells acutely infected with a seasonal human alpha coronavirus, HCoV-229E, were exposed to serial dilutions of Compound 1A. After a 3 -day incubation, the effective concentration of Compound 1A required to achieve 50% inhibition (ECso) of the virus-induced cytopathic effect (CPE) from two independent experiments averaged 1.8 pM. In contrast, the 2 , -fluoro-2’-methyl uridine nucleotide prodrug sofosbuvir did not inhibit HCoV- 229E replication at concentrations as high as 100 pM (Table 5). No toxicity was detected from either drug.

The in vitro potency of Compound 1A against HCoV-229E, HCoV-OC43 (another seasonal human coronavirus strain), MERS-CoV and SARS-CoV-1 was then evaluated in Huh-7 cell-based assays. This human hepatocarcinoma cell line was selected based on its ability to activate Compound 1A intracellularly to its triphosphate metabolite, unlike MRC-5 cells in which Compound 1A lacked activity against HCoV-229E (ECso >100 pM) as reported in Good, S.S. etal. PLoS One 15(1), e0227104 (2020)). Antiviral activity was assessed by two different methods after exposure of Huh-7 cells to virus and serial dilutions of test compound by determining 1) the ECso for virus-induced CPE by neutral red dye staining after a 5-day (229E and OC43) or 7-day (MERS and SARS) incubation and 2) the effective concentration required to reduce secretion of infectious virus into the culture medium by 90% (EC90) after a 3-day incubation using a standard endpoint dilution CCID50 assay to determine virus yield reduction (VYR). Half-maximal cytotoxicity (CC50) was measured by neutral red staining of compound-treated duplicates in the absence of virus. Although a robust VYR endpoint was obtained in Huh-7 cells infected with HCoV-OC43 or SARS- CoV-1, CPE was not observed and ECso values using neutral red staining were not obtained with these viruses. Individual determinations of EC90 values for Compound 1A against HCoV-229E, HCoV-OC43 and SARS-CoV-1 ranged from 0.34 to 1.2 pM, whereas the value against MERS- CoV averaged 37 pM (Table 5). No cytotoxicity was detected with Compound 1A up to 86 pM, the highest concentration tested.

Chloroquine and hydroxychloroquine appeared to be quite potent against HCoV-229E and HCoV-OC43 based on their EC90 values of <0.05 pM obtained using VYR measurements (Table 5). The respective EC50 values for these two drugs (8.1 and 7.4 mM), obtained using the neutral red assay, were substantially higher and only 2.6- to 3.6-fold less than the corresponding CC50 values, indicating considerably lower potencies and poor selectivity indices. These differences illustrate an inherent error in assessing antiviral activities of cytotoxic compounds using only measurements of VYR. When cells are poisoned by toxic drugs and are progressing toward death, their ability to support viral replication and propagation in addition to their own health likely is greatly diminished. At the point when cell death is detected by staining, viral yield reduction measurements likely reflect a combination of antiviral activity and cytotoxicity, thus overestimating antiviral potencies.

In contrast to data published in Wang, M. et al. ( Cell Research 2020, 30, 269), Huh-7 cells were not permissive for replication of SARS-CoV-2. An assay was developed using human airway epithelial (HAE) cell preparations, a highly relevant in vitro model of the lung, which has been established as a more representative system than cell lines for SARS-CoV-2 replication (Jomsdottir, H.R., Virol. ./. 13, 24 (2016)). These primary cells form polarized monolayers, the apical side of which is exposed to air and produces a mucin layer, consistent with the physiology of the human airways (Jomsdottir, H R., Virol. J. 13, 24 (2016)). Average EC90 and CC50 values for Compound 1A against SARS-CoV-2 from two separate HAE assays (0.5 and >86 mM, respectively) were in the same range as those obtained for HCoV-OC43 and SARS-CoV-1 (Table 5).

In the second HAE assay, the activity of Compound 1A was tested in parallel with N 4 - hydroxycytidine with recently reported in vitro and in vivo activity against SARS-CoV-2 (Sheahan, T.P. et al. Sci. Transl. Med. 12, eabb5883 (2020)). The potency of N 4 -hydroxycytidine against SARS-CoV-2 (EC90 = 3.9 mM) was 8 times less than that of Compound 1A in the same experiment.

A 30-fold difference of Compound 1A activity between MERS-CoV and other CoVs was observed. Nucleotide and nucleotide analogue selection is achieved at the CoV RdRp active site, the nspl2 gene product activated by its processivity co-factors nsp7 and nsp8 (Subissi, L., Proc. Natl. Acad. Sci. USA 111 (37) 3900-9 (2014)). Conserved amino acid motifs A and C are involved in phosphodiester bond formation, whereas motifs F and B participate in nucleotide channeling and binding at the active site, respectively. No significant structural differences are apparent between MERS-CoV and other CoVs in these essential motifs. With a similar ribose modification between Compound 1A and sofosbuvir, it is unlikely that the selective lack of activity of sofosbuvir would be due to excision by the CoV exonuclease carried by nspl4 (Ferron, F., Proc. Natl. Acad. Sci. USA 115 (2) 162-171 (2018)).

Cells, antivirals and viruses

BHK-21 (baby hamster kidney) cells , Huh-7 (human hepatocarcinoma) cells, RD (human rhabdomyosarcoma) cells and the seasonal human coronaviruses (HCoV-229E and HCoV-OC43) were obtained from American Type Culture Collection, Manassas, VA. MERS-CoV (EMC), SARS-CoV-1 (Urbani) and SARS-CoV-2 (USA-WA1/2020) were supplied by The Centers for Disease Control and Prevention, Atlanta, GA The HAE cell preparations (EpiAirwayTM AIR- 100 or AIR-112) were purchased from MatTek Corporation, Ashland, MA. Compound 1A and N4- hydroxycytidine were prepared for Atea Pharmaceuticals by Topharman Shanghai Co., Ltd., Shanghai, China and Oxeltis, Montpellier, France, respectively. Chloroquine and hydroxychloroquine were purchased from Mason-Chem, Palo Alto, CA and sofosbuvir was purchased from Pharma Sys, Inc., Cary, NC.

Antiviral assays

BHK-21 cells: Test compounds were dissolved in DMSO at 100 mM and then diluted in Minimum Essential Medium with Earle’s salts (MEM-E) containing 1 mM sodium pyruvate and 25 pg/mL kanamycin, supplemented with 10% FBS (growth medium) to final concentrations of 100, 20, 4 and 0.8 mM (two 24-well replica plates each). After BHK-21 cells were grown to confluency in 96-well plates, growth medium was replaced with fresh maintenance medium (growth medium with 1% inactivated FBS in place of 10% FBS) containing serially diluted test compound and HCoV-229E at a multiplicity of infection (MOI) of 0.01. Uninfected cells in the presence of serially diluted compound were used to assess the cytotoxicity of compounds. After a 3 -day incubation at 37°C in a humidified 5% CO2 atmosphere, cell viability was determined by the MTT method (Pauwels, R el al. ./. Virol. Methods 20(4): 309-321 (1988)). The effective concentration of test compound required to prevent virus-induced cytopathic effect (CPE) by 50% (EC50) and to cause 50% cell death in the absence of virus (CC50) were calculated by regression analysis.

Huh-7 and RD cells: The antiviral activities of test compounds were evaluated against human coronaviruses alpha (229E), beta (OC43), MERS (EMC) and SARS (Urbani) using a neutral red assay to determine inhibition of virus-induced and compound-induced CPE and using a virus yield reduction (VYR) assay as a second, independent determination of the inhibition of virus- induced CPE.

Neutral red assay: Test compounds were dissolved in DMSO at a concentration of 10 mg/mL and serially diluted using eight half-log dilutions in test medium (Minimum Essential Medium supplemented with 5% FBS and 50 pg/mL gentamicin) so that the highest test concentration was 50 pg/mL. Each dilution was added to 5 wells of a 96-well plate with 80-100% confluent Huh-7 or RD cells (OC43 only). Three wells of each dilution were infected with virus, and two wells remained uninfected as toxicity controls. Six untreated wells were infected as virus controls and six untreated wells were left uninfected to use as virus controls. Viruses were diluted to achieve MOIs of 0.003, 0.002, 0.001 and 0.03 CCID50 per cell for 229E, OC43, MERS and SARS, respectively. Plates were incubated at 37±2°C in a humidified atmosphere containing 5% CO 2.

On day 5 (229E and OC43) or day 7 (MERS and SARS) post-infection, when untreated virus control wells reached maximum CPE, the plates were stained with neutral red dye for approximately 2 hours (±15 minutes). Supernatant dye was removed, wells were rinsed with PBS, and the incorporated dye was extracted in 50:50 Sorensen citrate buffer/ethanol for >30 minutes and the optical density was read on a spectrophotometer at 540 nm. Optical densities were converted to percent of controls and the concentrations of test compound required to prevent virus-induced CPE by 50% (ECso) and to cause 50% cell death in the absence of virus (CC50) were calculated.

Virus yield reduction assay: Vero 76 cells were seeded in 96-well plates and grown overnight (37°C) to confluence. A sample of the supernatant fluid from each compound concentration was collected on day 3 post infection (3 wells pooled) and tested for virus titer using a standard endpoint dilution CCID50 assay and titer calculations using the Reed-Muench equation (1948) (Reed, LJ and Muench, H. Am. J. Hygiene 27:493-497 (1948)) and the concentration of compound required to reduce virus yield by 90% (EC90) was determined by regression analysis.

HLE cell preparations

The antiviral activities of test compounds were evaluated against SARS-CoV-2 (USA- WA1/2020) using made to order human airway epithelial (HAE) cells.

Cell Culture: HAE cells were grown on 6mm mesh disks and arrived in kits with either 12- or 24-well transwell inserts. During transportation the tissues were stabilized on a sheet of agarose, which was removed upon receipt. One insert was estimated to consist of approximately 1.2 x 10 6 cells. Kits of cell inserts (EpiAirway™ AIR-100 or AIR-112) originated from a single donor, # 9831, a 23-year old, healthy, non-smoking, Caucasian male. The cells form polarized monolayers, the apical side of which is exposed to air and creates a mucin layer. Upon arrival, the cell transwell inserts were immediately transferred to individual wells of a 6-well plate according to the manufacturer’s instructions, and 1 mL of MatTek’s proprietary culture medium (AIR-100-MM) was added to the basolateral side, whereas the apical side was exposed to a humidified 5% CO2 environment. Cells were cultured at 37°C in a humidified atmosphere containing 5% CO2 for one day before the start of the experiment. After the 24-h equilibration period, the mucin layer, secreted from the apical side of the cells, was removed by washing with 400 pL pre-warmed 30 mM HEPES buffered saline solution 3X. Culture medium was replenished following the wash steps.

Viruses: Virus was diluted in AIR-IOO-MM medium before infection to yield a MOI when added to cultures of approximately 0.0015 CCID 50 per cell. Experimental design : Each compound treatment (120 pL) and virus (120 pL) was applied to the apical side, and the compound treatment (1 mL) was applied to the basal side As a virus control, some of the cells were treated with cell culture medium only. After a 2-h infection incubation, the apical medium was removed, and the basal medium was replaced with fresh compound or medium (1 mL). The cells were maintained at the air-liquid interface. On day 5, cytotoxicity (CC50 values) in the uninfected, compound-treated inserts was estimated by visual inspection, and the basal medium was removed from all inserts and discarded. Virus released into the apical compartment of the HAE cells was harvested by the addition of 400 pL of culture medium that was pre-warmed at 37°C. The contents were incubated for 30 min, mixed well, collected, thoroughly vortexed and plated on Vero 76 cells for VYR titration. Separate wells were used for virus control and duplicate wells were used for untreated cell controls. Virus titers from each treated culture were determined as described above.

Example 7. Compound 1A Triphosphate Levels in Human Nasal and Bronchial Cells Compound 1A (10 pM) was incubated in triplicate with human nasal and bronchial epithelial cells for 8 hours. At the end of the 8-hr exposure to individual test articles, the incubation medium was removed and the cell layer was washed with Hepes buffered saline solution (HBSS). HBSS was removed, followed by the addition of fresh cell culture medium without test article. At 0, 15, 24, 48, and 72 hours after the removal of test article, the extracellular medium was removed, and the cell layer was rinsed with HBSS. The cells were scraped off from plates and suspended in cold 60% methanol in water containing the internal standard AT 9005 and stored at ca. -20 °C, followed by centrifugation for LC/MS/MS analysis for the formation of the corresponding triphosphate metabolite of Compound 1A, Compound 1-6 (Scheme 1). Table 6 provides the mean intracellular concentration of the triphosphate metabolite Compound 1-6 at each of the time points. FIG. 1 is a graph of the concentration of Compound 1-6 at each time point post-exposure in bronchial cells and nasal cells. The half-life (ti / 2) in nasal cells was 38 hours and 39 hours in bronchial cells. The triphosphate level in bronchial cells was greater than in nasal cells, but substantial levels of triphosphate were formed in both. The half-life in both cells was over 1.5 days and no toxicity was observed up to 100 mM. Table 6. Intracellular Concentration of Triphosphate Concentrations in Bronchial and Nasal

Cells

Calculated using an average volume of 1320 microns 3 for alveolar type I and II epithelial cells (Crapo, J.D. et al. 1982 Am. Rev. Respir. Dis. 126(2): 332-7. doi:10.1164/arrd.l982.126.2.332.)

FIG. 2 and FIG. 3 are bar graphs of the triphosphate levels in bronchial and nasal cells, respectively, at each of the time points. FIGS. 2 and 3 compare the triphosphate level formed from Compound 1A to the triphosphate level formed from ALS-8112 (shown below) and the 4’-Me substituted prodrug (isopropyl ((S)-(((2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-y l)- 4-fluoro-3-hydroxy-2,4-dimethyltetrahydrofuran-2-yl)methoxy) (phenoxy)phosphoryl)-L- alaninate, shown below on the left). ALS-8112 is a clinically effective drug against RSV with an in vitro EC 9O of 1.3 - 2.7 mM in RSV-infected HAE cells (Deval, J. et al. 2015 PLoS Pathog 11(6): el004995).

As shown in FIGS. 2 and 3, substantially more triphosphate is formed from Compound 1A compared to ALS-8112 or the 4’-Me substituted prodrug in both human bronchial and nasal cells. At end of 8 hour incubation with each drug at a concentration of 10 mM, ratios of triphosphate from Compound 1A vs. ALS-8112 and the 4’-Me substituted prodrug were 2 and 84 in bronchial cells and 1 and 14 in nasal cells, respectively. Ratios of triphosphate from Compound 1A vs. ALS-8112 and the 4 ’-Me substituted prodrug were 5 and 54 in bronchial cells and 3 and 8 in nasal cells, respectively, 15 hours after washout.

Example 8. Triphosphate Levels in Tissues of Non-Human Primates after Oral Administration of Compound 2A

Non-human primates were administered a three-day oral dosing regimen of Compound 2A to achieve steady state levels. The primates were given one 60 mg/kg dose followed by five 30 mg/kg doses every 12 hours (doses were allometrically scaled from clinical dosing regimen of 100 mg loading dose + 550 mg twice a day (BED)).

The plasma PK of metabolites Compound 1A, Compound 1-2, and the triphosphate surrogate Compound 1-7 were determined. Just prior to the penultimate dose and at 0.5, 1, 2, 4, 6, 8 and 12 h (just prior to the last dose) thereafter, blood samples were obtained from 3 monkeys and mixed with EDTA. Plasmas were then prepared by centrifugation and analyzed for concentrations of Compound 1A, Compound 1-2, and Compound 1-7 by LC-MS/MS. (Triphosphate Compound 1-6 is produced in the cell and does not leave. It is therefore not measurable in the plasma. However, the 5'-OH metabolite Compound 1-7 (see Scheme 1) is exported from the cell, and therefore is measurable in plasma and can act as a surrogate for the intracellular active metabolite Compound 1-6.) The plasma PK data for the metabolites are given in Table 7.

Table 7. Plasma PK data for Metabolites 1A, 1-2, and 1-7 following Compound 2A Dosing FIGS. 4, 5, and 6 are graphs of the mean plasma profile of Compound 1A, Compound 1-2, and Compound 1-7, respectively, in the non-human primates following administration of oral doses of Compound 2A (30 mg/kg BID for 3 days). Plasma trough concentrations (mean of 0- and 12 h time points) for Compound 1-2 (intermediate prodrug) and Compound 1-7 (surrogate for intracellular TP levels) are 0.20 and 0.11 mM, respectively. The profiles show rapid conversion of Compound 1A to Compound 1-2 and Compound 1-7 (a surrogate for triphosphate Compound 1-6).

Lung, kidney, and liver tissue levels were determined for triphosphate Compound 1-6, Compound 1A, and other metabolites at 12, 24, and 48 hours post last dose (3 males per time point) as shown in FIG. 7A and FIG. 7B. As described above, three male non-naive cynomolgus monkeys were administered a 60 mg/kg loading dose followed by five 30 mg/kg doses every 12 h of Compound 2A. Samples of plasma, lung, kidney and liver tissue were collected at 12, 24, and 48 hours post last dose from the three anesthetized animals at each time point and immediately flash- frozen in liquid nitrogen. Approximately 0.5 g of each sample of tissue was homogenized using a Polytron in 5 volumes (5 mL/g) 70% methanol:30% 268 mM EDTA adjusted to pH 7.8 and containing appropriate internal standards in tubes immersed in a dry ice:ethanol bath. Homogenates were analyzed for concentrations of Compound 1A, Compound 1-2, Compound 1-6, and Compound 1-7 by LC-MS/MS.

The trough (12 h) levels of active metabolite triphosphate Compound 1-6 in lung, kidney and liver non-human primate tissues was 0.14 mM, 0.13 pM, and 0.09 pM, respectively. The triphosphate species concentration was 1.6-fold greater in the lung compared to the liver at 12-hour steady-state trough levels. Table 8 provides the mean intracellular concentration of the triphosphate metabolite Compound 1-6 in lung, kidney, and liver tissue at each of the time points. As shown in FIG. 7A, the half-life of Compound 1-6 in lung, kidney, and liver was 9.4 hours, 8.0 hours, and 4.3 hours, respectively. The half-life of Compound 1-6 was determined by dividing ln(2) by k, the rate constant for the decrease in Compound 1-6 concentration obtained from the slope of the plot of In (tissue concentration) vs. time after linear regression analysis.

Table 8. Intracellular Concentration of Compound 2A Triphosphate Concentrations in Lung, Kidney, and Liver Cells

Calculated using non-interstitial volumes of 0.75 and 0.9 mL/g lung and liver, respectively, and an assumed volume of 0.83 mL/g kidney (Mandikian, D. et al. 2018 AAPS Journal 20(6): 107. doi.org/ 10.1208/sl2248-018-0264-z.)

Example 9. Prediction of Human Lung and Kidney Concentrations of Triphosphate Compound 1-6 based on Compound 1-6 Tissue Levels in Non-Human Primates

As described in (Good, S.S. et al. 2020 PLoS ONE 15(l):e0227104), the levels of triphosphate Compound 1-6 in human and monkey hepatocytes incubated with 10 mM of Compound 1A were determined. FIG. 8 is a graph comparing the levels in the two species, and as shown in the Figure, triphosphate Compound 1-6 concentrations in human hepatocytes is 7-fold greater than in non-human primate (monkey) hepatocytes assessed by AUCo- 24 values. The data is also presented in Table 9. Using a hepatocyte volume of 3.4 x 10-9 mL (14) to calculate the intracellular concentration, Compound 1-6 peaked at 8 hours in human hepatocytes at 26 + 1 mM, while the highest concentration in monkey hepatocytes was 3.1 + 0.1 mM at 4 hours. Table 9. Intracellular Concentration of Triphosphate Concentrations in Monkey and Human Hepatocytes

Calculated using a hepatocyte volume of 3.4 xlO 9 mL (Lodish, H. et al. 2000 Molecular Cell Biology (fifth edition), W.H. Freeman and Co.. New York. P. 10.

Based on the ratio (7.0) of human to monkey concentrations of triphosphate Compound 1- 6 as assessed by its in vitro formation in primary hepatocytes, the predicted human tissue level was determined. The actual tissue levels of triphosphate Compound 1-6 in monkeys and the predicted tissue levels in humans is shown in Table 10. A clinical dosing regimen of 1100 mg LD (loading dose) + 550 mg BID (twice a day) is predicted to achieve lung intracellular levels of triphosphate Compound 1-6 at trough (12 h) above the in vitro EC90 of Compound 2A against SARS-CoV-2 replication in HAE cell cultures. Triphosphate levels during this dosing regimen are predicted to consistently remain above the in vitro EC90 of Compound 2A against SARS-CoV-2. Predictions are conservative (at least for the lung) because the ling triphosphate half-life in human bronchial/nasal cells (38 hours) as compared to the non-human primate lung tissue (9.4 hours) predicts higher steady-state levels in humans with BID dosing, but this was not factored into predictions. Table 10. Actual (Monkey) and Predicted (Human) Tissue Levels of Compound 1-6

Example 10. Simulation of Intracellular Concentrations of Triphosphate Compound 1-6 in Human Lung Tissue

Plasma conc-time profiles of Compound 1-7 (a surrogate for triphosphate Compound 1-6 because Compound 1-6 cannot be measured in plasma) was obtained from subjects (N=18) who received 600 mg QD dose of Compound 2A (equivalent to 553 mg of free base Compound 1A) for 7 days as part of the AT-OlB-001 study (as described in Fig 1 and Table 3 of Berliba, e. et al. 2019 Antimicrob. Agents Chemother. 63(12):e01201-19). The profiles were subjected to population pharmacokinetic (PPK) analysis using Monolix Suite 2019 Suite 2019 (Lixosoft, Antony, France). The obtained PPK parameters together with the associated variance and covariance matrices were then used to simulate, using the Simulx module of Monolix Suite, plasma PK profiles of Compound 1-7 for various dosing regimens.

It was assumed that 1) lung triphosphate levels of Compound 1-7 were 1.6 the plasma levels of its nucleoside metabolite Compound 1-7 based on the ratio obtained in monkeys for lung-to-liver concentrations of Compound 1-6; and 2) the average in vitro EC90 of Compound 1A against SARS- CoV-2 was about 0.55 uM in HAE cell preparations. Under these assumptions, the simulated lung Compound 1-6 levels would exceed the in vitro EC90 shortly after administration of a loading dose of 1100 mg and remain above EC90 with the 550 BID maintenance doses for the remaining days. The simulated lung Compound 1-6 concentration over the course of 5 days is shown in FIG. 9.

Additional simulations were also conducted. The kinetics of human lung Compound 1-6 levels were simulated for a Compound 2A 550 mg BID dose regimen for 5 days (FIG. 10) using published plasma Compound 1-7 data from subjects given daily 550 mg doses of Compound 2A (described in Berliba, e. et al. 2019 Antimicrob. Agents Chemother. 63(12):e01201-19) amplified by a factor of 1.6 based on the assumption that the observed lung-to-liver Compound 1-6 concentration ratio in monkys is applicable to humans as well. The resulting predicted steady-state peak and trough levels for the active triphosphate Compound 1-6 in human lung for this dose regimen are 1.5 and 0.9 mM, respectively. A second approach to predicting human lung Compound 1-6 concentrations used the same simulated plasma Compound 1-7 data but corrected the plasma values by a factor of 1.2, which is the ratio of the mean steady-state 12-h lung Compound 1-6 concentration in monkies to that of Compound 1-7 in plasma. This prediction provided respective estimates of 1.1 and 0.7 mM for peak and trough human lung triphosphate concentrations.

According to both methods, the predicted human lung levels of Compound 1-6 exceed the EC90 value observed against SARS-CoV-2 replication in HAE cells from within a few hours after the first dose through the end of the dosing period.

FIG. 10 shows the stimulated Compound 1-6 concentration in human lung using both approaches. The solid curve represents predicted lung concentrations of the active triphosphate Compound 1-6 metabolite after correcting for the Compound 1-6 lung-to-liver concentration ratio of 1.6. The dotted curve represents predicted lung concentrations of the active triphosphate Compound 1-6 metabolite after correcting for the Compound 1-6 lung-to- Compound 1-7 plasma ratio of 1.2. The horizontal line represents the EC90 of Compound 1A against SARS-CoV-2 in HAE cells in vitro (0.47 pM).

Example 11. In Vitro Activity of Select Compounds Against SARS-CoV-2 Select compounds of the present invention were tested in a SARS-CoV-2 assay. The assay was conducted using human airway epithelial (HAE; dNHBE) cells as described in Example 6. Remdesivir was the positive control and the compounds were compared to Sofosbuvir. Compounds were tested in singlet using 4 serial dilutions of each compound (10, 2.5, 0.625 and 0.156 pg/mL. Sofosbuvir was only tested in duplicate at 10 ug/mL, Compound 1A in singlet at 1 and 0.1 ug/mL, and remdesivir in singlet at 4 serial log dilutions with top concentration of 1 ug/mL. The results are shown in Table 11. Compound 5 with deuterium substitution at the 2’-position exhibited an EC90 of 0.31 mM and Compound 6 with 2’-CH2F substitution exhibited an EC90 of 1.8 pM. None of the compounds showed visual toxicity. Table 11. Activity of Select Compounds Against SARS-CoV-2

Table 11B. Activity of Additional Select Compounds Against SARS-CoV-2

Example 12. Formulation Description and Manufacturing of Compound 2A

A representative non-limiting batch formula for Compound 2A tablets (50 mg and 100 mg) is presented in Table 12. The tablets were produced from a common blend using a direct compression process. The active pharmaceutical ingredient (API) was adjusted based on the as-is assay, with the adjustment made in the percentage of microcrystalline cellulose. The API and excipients (microcrystalline cellulose, lactose monohydrate, and croscarmellose sodium) were screened, placed into a V-blender (PK Blendmaster, 0.5L bowl) and mixed for 5 minutes at 25 rpm. Magnesium Stearate was then screened, added and the blend was mixed for an additional 2 minutes. The common blend was divided for use in producing 50 mg and 100 mg tablets. The lubricated blend was then compressed at a speed of 10 tablets/minutes using a single punch research tablet press (Korsch XP1) and a gravity powder feeder. The 50 mg tablets were produced using round standard concave 6 mm tooling and 3.5 kN forces. The 100 mg tablets were produced using 8 mm round standard concave tooling and 3.9-4.2 kN forces.

Table 12. Formulation of 50 mg and 100 mg Compound 2 Tablets Compound 2A was adjusted based on the as-is assay, with the adjustment made in the percentage of microcrystalline cellulose. Compound 2A and excipients (microcrystalline cellulose, lactose monohydrate, and croscarmellose sodium) were screened, placed into a V-blender (PK Blendmaster, 0.5L bowl) and mixed for 5 minutes at 25 rpm. Magnesium stearate was then screened, added and the blend was mixed for an additional 2 minutes. The common blend was divided for use in producing 50 mg and 100 mg tablets. The lubricated blend was then compressed at a speed of 10 tablets/minutes using a single punch research tablet press (Korsch XP1) and a gravity powder feeder. The 50 mg tablets were produced using round standard concave 6 mm tooling and 3.5 kN forces. The 100 mg tablets were produced using 8 mm round standard concave tooling and 3.9-4.2 kN forces. The specifications of the 50 mg and 100 mg tablets are shown in Table 13.

Table 13. Specifications of 50 mg and 100 mg Tablets of Compound 2A

The 50 mg and 100 mg tablets produced as described above were subjected to 6 month stability studies under three conditions: 5°C (refrigeration), 25°C/60% RH (ambient), and 40°C/75% RH (accelerated). Both the 50 mg and 100 mg tablets were chemically stable under all three conditions tested.

Under refrigeration conditions (5 °C), both the 50 mg and 100 mg tablets remained white solids that did not change in appearance from T=0 to T=6 months. Throughout the 6-month study, no impurities were reported that were greater than 0.05% for either the 50 mg tablets or the 100 mg tablets. The water content after 6 months was also less than 3.0 % w/w for both tablets. Similar results were reported when the tablets were subjected to ambient conditions (25 °C/60% RH); no impurities that were greater than 0.05% were reported throughout the 6 months for both tablets and the water content did not exceed 3.0 % w/w at the 6-month mark. When the tablets were subjected to accelerated conditions (40 °C/75% RH), the appearance of the 50 mg and 100 mg tablets did not change from a white, round tablet. One impurity was reported after 3 months, but the impurity was only 0.09%. A second impurity was reported after 6 months, but the total impurity percentage was only 0.21% for both the 50 mg and 100 mg tablets. Water content was 3.4 % w/w at 6 months for the 50 mg tablets and 3.2 % w/w for the 100 mg tablets.

In a separate study, the stability of 50 mg and 100 mg tablets of Compound 2A at ambient conditions (25 °C/60% RH) was measured over 9 months. The appearance of the 50 mg and 100 mg tablet did not change from a white round tablet over the course of 9 months. Impurities in the 50 mg tablet were less than 0.10% after 9 months and impurities in the 100 mg tablet were less than 0.05%. The water content of the 50 mg tablet and the 100 mg tablet after 9 months was only 2.7 % w/w and 2.6 % w/w, respectively.

This specification has been described with reference to embodiments of the invention. Given the teaching herein, one of ordinary skill in the art will be able to modify the invention for a desired purpose and such variations are considered within the scope of the invention.