Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PAN FILOVIRUS VACCINE COMPOSITIONS AND METHODS OF MAKING
Document Type and Number:
WIPO Patent Application WO/2017/172622
Kind Code:
A1
Abstract:
[0001] Provided herein are methods of incorporating substitutions of specified residues into a filovirus GP in order to increase immunogenicity and/or broaden the cross-reactivity of the protective immune response against other filovirus members. Also provided herein are mutant filovirus GPs comprising such substitutions.

Inventors:
AMAN MOHAMMAD JAVAD (US)
HOWELL KATIE A (US)
DAVIDSON EDGAR (US)
DORANZ BENJAMIN J (US)
Application Number:
PCT/US2017/024320
Publication Date:
October 05, 2017
Filing Date:
March 27, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INTEGRATED BIOTHERAPEUTICS INC (US)
INTEGRAL MOLECULAR INC (US)
International Classes:
A61K39/12; C07K14/08; C07K16/10; C12N7/00; C12N15/40; C12N15/86
Foreign References:
US20090232841A12009-09-17
Other References:
KECK ET AL.: "Macaque Monoclonal Antibodies Targeting Novel Conserved Epitopes within Filovirus Glycoprotein", J VIROL, vol. 90, no. 1, 14 October 2015 (2015-10-14), pages 279 - 291, XP055317321
FUSCO ET AL.: "Protective mAbs and Cross-Reactive mAbs Raised by Immunization with Engineered Marburg Virus GPs", PLOS PATHOG, vol. 11, no. 6, 26 June 2015 (2015-06-26), pages 1 - 17, XP055405438
LEE ET AL.: "Structure of the Ebola virus glycoprotein bound to an antibody from a human survivor", NATURE, vol. 454, no. 7201, 10 July 2008 (2008-07-10), pages 177 - 182, XP055066642
DOWLING ET AL.: "Influences of glycosylation on antigenicity, immunogenicity, and protective efficacy of ebola virus GP DNA vaccines", J VIROL, vol. 81, no. 4, 6 December 2006 (2006-12-06), pages 1821 - 1837, XP055430913
MISASI ET AL.: "Structural and molecular basis for Ebola virus neutralization by protective human antibodies", SCIENCE, vol. 351, no. 6279, 25 February 2006 (2006-02-25), pages 1343 - 1346, XP055430914
Attorney, Agent or Firm:
HOLTZ, William A. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. An immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, wherein the base domain comprises one or more single amino acid substitutions relative to the corresponding wild- type filovirus GP amino acid sequence, and wherein the one or more amino acid substitutions can affect the conformation of a cross-reactive epitope in the head domain, thereby increasing immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

2. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to C53, F183, N521, A562, L569, or L573 of the wild- type Ebola virus (EBOV) GP, or a combination thereof.

3. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to F159, P513, L515, or T565 of the wild-type EBOV GP, or a combination thereof.

4. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to R164, LI 84, 1185, H516, or G546 of the wild-type EBOV GP, or a combination thereof.

5. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to L51, G179, Q508, C511, Y517, R559, or C601 of the wild-type EBOV GP, or a combination thereof.

6. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, or 1610 of the wild-type EBOV GP, or a combination thereof.

7. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of the wild-type EBOV GP, or a combination thereof.

8. The immunogen of claim 1, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to C53, F183, N521, A562, L569, L573, F159, P513, L515, T565, R164, L184, 1185, H516, G546, L51, G179, Q508, C511, Y517, R559, C601, 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, 1610, L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of the wild-type EBOV GP, or a combination thereof.

9. An immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, wherein the base domain comprises one or more single amino acid substitutions relative to the corresponding wild- type filovirus GP amino acid sequence, and wherein the one or more amino acid substitutions can affect the conformation of a species specific epitope in the base domain, thereby masking an immunodominant epitope in the base domain.

10. The immunogen of claim 9, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to E103, F159, E178, F183, C511, L515A, W518, N550, D552, G553, C556, or R559 of the wild-type EBOV GP, or a combination thereof.

11. The immunogen of claim 9 or claim 10, wherein the one or more amino acid substitutions also increases immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadens the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

12. The immunogen of claim 11, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to F159, F183, C511, L515, or R559 of the wild-type EBOV GP, or a combination thereof.

13. The immunogen of claim 11, wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to F 183 of the wild-type EBOV GP.

14. The immunogen of any one of claims 1 to 13, wherein the immunogen further comprises the glycan cap of the filovirus GP1 subunit or an immunogenic fragment thereof.

15. An immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or an immunogenic fragment thereof, wherein the glycan cap comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP, and wherein the one or more amino acid substitutions reduce glycosylation of the glycoprotein, thereby increasing

immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

16. The immunogen of claim 15, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises an amino acid substitution at a position corresponding to N238, T240, N257, T259, N268, or T270 of the wild- type EBOV GP, or a combination thereof.

17. The immunogen of claim 16, wherein the amino acid substitution in the glycan cap comprises at least two single amino acid substitutions selected from a group consisting of amino acid substitutions at a position corresponding to N238 and/or T240, at a position corresponding to N257 and/or T259, and at a position corresponding to N268 and/or T270 of the wild-type EBOV GP, wherein the substitutions disrupt at least two glycosylation sites.

18. An immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or an immunogenic fragment thereof, wherein the glycan cap comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence within the disordered loop connecting the β17 to β18 strands, and wherein the one or more amino acid substitutions can affect the conformation of a cross-reactive epitope in the glycan cap, thereby increasing the immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

19. The immunogen of claim 18, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises an amino acid substitution at a position corresponding to F290, W291, or E292 of the wild-type EBOV GP, or a combination thereof.

20. The immunogen of claim 18, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises an amino acid substitution at a position corresponding to E292 of the wild-type EBOV GP.

21. The immunogen of any one of 15 to 20, wherein the wild-type filovirus is a wild- type Ebola virus (EBOV), and wherein the glycan cap comprises: (i) an amino acid substitution at a position selected from the group consisting of N238, T240, N257, T259, N268, and T270 of the wild-type EBOV GP, or a combination thereof; and (ii) an amino acid substitution at a position selected from the group consisting of F290, W291, and E292 of the wild-type EBOV GP, or a combination thereof.

22. An immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the GP internal fusion loop domain or an immunogenic fragment thereof, wherein the GP comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence, and wherein the one or more amino acid substitutions increases immunogenicity of the immunogen against the corresponding wild-type filovirus GP internal fusion loop domain, and/or broadens the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

23. The immunogen of claim 22, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the GP amino acid substitution comprises an amino acid substitution at a position corresponding to N40, T42, D192, F193, F194, or Q595 of the wild-type Ebola virus (EBOV) GP, or a combination thereof.

24. The immunogen of claim 22, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the GP amino acid substitution comprises an amino acid substitution at a position corresponding to N40, D 192, F 193, or F 194 of the wild-type Ebola virus (EBOV) GP, or a combination thereof.

25. The immunogen of any one of claims 22 to 24, further comprising one or more GP amino acid substitutions of an immunogen of any one of claims 1 to 21.

26. The immunogen of any one of claims 1 to 25, wherein the wild-type Ebola virus is EBOV strain Mayinga-76.

27. The immunogen of any one of claims 1 to 26, wherein the other filovirus species or strain comprises EBOV, SUDV, BDBV, RESTV, TAFV, MARV, any strain thereof, or a combination thereof.

28. The immunogen of any one of claims 1 to 27, wherein the wild-type amino acid is substituted with an alanine (A) residue, except where the wild-type amino acid is A.

29. The immunogen of any one of claims 1 to 27, wherein the wild-type amino acid is A and the wild-type amino acid is substituted with a serine (S) residue.

30. The immunogen of any one of claims 1 to 27, wherein the wild-type amino acid is substituted with a glycine (G) residue, except where the wild-type amino acid is G.

31. The immunogen of any one of claims 1 to 30, wherein the immunogen lacks the mucin like domain (MLD) of the filovirus GP1 subunit.

32. An immunogen comprising a GP filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the immunogen is a cleaved GP (GPCL) lacking the filovirus GPl subunit MLD and glycan cap.

33. The immunogen of claim 32, wherein the GPCL comprises one or more GP amino acid substitutions of an immunogen of any one of claims 1 to 30.

34. A method of increasing immunogenicity of and/or broadening the cross-reactive immunogenicity of an immunogen comprising a filovirus GP or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, the method comprising making one or more single amino acid substitutions in the base domain relative to the corresponding wild-type filovirus GP amino acid sequence, thereby affecting the conformation of a cross-reactive epitope in the head domain, thereby increasing immunogenicity against the corresponding wild-type filovirus GP and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

35. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to C53, F183, N521, A562, L569, or L573 of the wild-type Ebola virus (EBOV) GP, or a combination thereof.

36. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to F159, P513, L515, or T565 of the wild-type EBOV GP, or a combination thereof.

37. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to R164, L184, 1185, H516, or G546 of the wild-type EBOV GP, or a combination thereof.

38. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to L51, G179, Q508, C511, Y517, R559, or C601 of the wild-type EBOV GP, or a combination thereof.

39. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, or 1610 of the wild-type EBOV GP, or a combination thereof.

40. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of the wild-type EBOV GP, or a combination thereof.

41. The method of claim 34, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to C53, F183, N521, A562, L569, L573, F159, P513, L515, T565, R164, L184, 1185, H516, G546, L51, G179, Q508, C511, Y517, R559, C601, 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, 1610, L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of the wild-type EBOV GP, or a combination thereof.

42. A method of masking an immunodominant epitope in a immunogen comprising a filovirus GP or a fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, the method comprising making one or more single amino acid substitutions in the base domain relative to the corresponding wild-type filovirus GP amino acid sequence, thereby affecting the

conformation of a species specific epitope in the base domain, thereby masking the

immunodominant epitope in the base domain.

43. The method of claim 42, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to E103, F159, E178, F183, C511, L515A, W518, N550, D552, G553, C556, or R559 of the wild-type EBOV GP, or a combination thereof.

44. The method according to claim 42 or claim 43, wherein the one or more amino acid substitutions also increases immunogenicity of the immunogen against the corresponding wild-type filovirus GP and/or broadens the immunogenetic cross-reactivity of the immunogen against other filovirus species or strains.

45. The method of claim 44, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to F159, F183, C511, L515, or R559 of the wild-type EBOV GP, or a combination thereof.

46. The method of claim 44, wherein the amino acid substitution in the GP base domain comprises an amino acid substitution at a position corresponding to F 183 of the wild-type EBOV GP.

47. The method of any one of claims 34 to 46, wherein the immunogen further comprises the glycan cap of the filovirus GP1 subunit or an immunogenic fragment thereof.

48. A method of increasing the immunogenicity of and/or broadening the cross-reactive immunogenicity of an immunogen comprising a filovirus GP or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or an immunogenic fragment thereof, the method comprising making one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP in the glycan cap, thereby reducing

glycosylation of the glycoprotein, thereby increasing the immunogenicity of the immunogen against the corresponding wild-type virus GP, and/or broadening the immunogenetic cross- reactivity of the immunogen against other filovirus species or strains.

49. The method of claim 48, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises an amino acid substitution at a position corresponding to N238, T240, N257, T259, N268, or T270 of the wild- type EBOV GP, or a combination thereof.

50. The method of claim 49, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises at least two single amino acid substitutions selected from a group consisting of amino acid substitutions at a position corresponding to N238 and/or T240, a position corresponding to N257 and/or T259, and a position corresponding to N268 and/or T270 of the wild-type EBOV GP.

51. A method of increasing the immunogenicity of and/or broadening the cross-reactive immunogenicity of an immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or immunogenic fragment thereof, the method comprising making one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence within the disordered loop connecting the β17 to β18 strands, thereby affecting the conformation of a cross- reactive epitope in the glycan cap, thereby increasing immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

52. The method of claim 51, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises an amino acid substitution at a position corresponding to F290, W291, or E292 of the wild-type EBOV GP, or a combination thereof.

53. The method of claim 52, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution in the glycan cap comprises an amino acid substitution at a position corresponding to E292 of the wild-type EBOV GP.

54. The method of any one of 48 to 53, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the glycan cap comprises: (i) an amino acid substitution at a position selected from the group consisting of N238, T240, N257, T259, N268, and T270 of the wild-type EBOV GP, or a combination thereof; and (ii) an amino acid substitution at a position selected from the group consisting of F290, W291, and E292 of the wild-type EBOV GP, or a combination thereof.

55. A method of increasing immunogenicity of and/or broadening the cross-reactive immunogenicity of an immunogen comprising a filovirus GP or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the GP internal fusion loop domain or an immunogenic fragment thereof, the method comprising making one or more single amino acid substitutions in the filovirus GP amino acid sequence relative to the corresponding wild-type filovirus GP amino acid sequence, thereby increasing immunogenicity against the corresponding wild-type filovirus GP internal fusion loop domain and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

56. The method of claim 55, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the GP amino acid substitution comprises an amino acid substitution at a position corresponding to N40, T42, D192, F193, F194, or Q595 of the wild-type Ebola virus (EBOV) GP, or a combination thereof.

57. The method of claim 55, wherein the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the GP amino acid substitution comprises an amino acid substitution at a position corresponding to N40, D 192, F 193, or F 194 of the wild-type Ebola virus (EBOV) GP, or a combination thereof.

58. The method of any one of claims 55 to 57, wherein the immunogen further comprises one or more GP amino acid substitutions of an immunogen of any one of claims 1 to 21.

59. The method of any one of claims 34 to 58, wherein the wild-type Ebola virus is EBOV strain Mayinga-76.

60. The method of any one of claims 34 to 59, wherein the other filovirus species or strain comprises EBOV, SUDV, BDBV, RESTV, TAFV, MARV, any strain thereof, or a combination thereof.

61. The method of any one of claims 34 to 60, wherein the wild-type amino acid is substituted with an alanine (A) residue except where the wild-type amino acid is A.

62. The method of any one of claims 34 to 60, wherein the wild-type amino acid is A and the wild-type amino acid is substituted with a serine (S) residue.

63. The method of any one of claims 34 to 60, wherein the wild-type amino acid is substituted with a glycine (G) residue except where the wild-type amino acid is G.

64. The method of any one of claims 34 to 63, wherein the immunogen lacks the mucin like domain (MLD) of the filovirus GP 1 subunit.

65. A method of increasing immunogenicity of and/or broadening the cross-reactive immunogenicity of an immunogen comprising a GP filovirus spike glycoprotein (GP) or immunogenic fragment thereof, the method comprising using an immunogen that is a cleaved GP (GPCL) lacking the filovirus GP 1 subunit MLD and glycan cap, thereby increasing immunogenicity against the corresponding wild-type filovirus GP and/or broadening the cross-reactive

immunogenicity of the immunogen against other filovirus species or strains.

66. The method of claim 65 comprising making one or more GP amino acid

substitutions of an immunogen of any one of claims 1 to 30 in the GPCL, thereby increasing immunogenicity against the corresponding wild-type filovirus GP and/or broadening the cross- reactive immunogenicity of the immunogen against other filovirus species or strains.

67. A composition comprising the immunogen of any one of claims 1 to 33 and an adjuvant.

68. A composition comprising the immunogen of any one of claims 1 to 33 incorporated into a filovirus virus-like particle (VLP).

69. The composition of claim 68, wherein the filovirus VLP comprises a the

immunogen and a filovirus VP40.

70. The composition of claim 69, wherein the filovirus VLP further comprises a filovirus nucleoprotein ( P) and/or filovirus VP24.

71. An isolated polynucleotide comprising a nucleic acid encoding the immunogen or fragment thereof of any one of claims 1 to 33 or a subunit thereof.

72. A vector comprising the polynucleotide of claim 71.

73. A composition comprising the polynucleotide of claim 71 or the vector of claim 72.

74. A host cell comprising the polynucleotide of claim 71 or the vector of claim 72.

75. A method of making the immunogen of any one of claims 1 to 33, the method comprising:

(a) culturing the host cell of claim 74; and

(b) recovering the immunogen.

Description:
PAN FILOVIRUS VACCINE COMPOSITIONS AND METHODS OF MAKING

CRO S S -REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. Provisional Application No. 62/314,009 filed March 28, 2016 and U.S. Provisional Application No. 62/423,584, filed November 17, 2016, both of which are incorporated by reference herein in their entireties.

STATEMENT REGARDING FEDERALLY-SPONSORED

RESEARCH AND DEVELOPMENT

[0002] Statement under MPEP 310. This invention was made with government support under HDTRA1-13-C-0015 awarded by US Defense Threat Reduction Agency (DTRA), HHSN272201400058C awarded by US Department of Health and Human Services (HHS) and NIAID grant AI098178 awarded by National Institute of Allergy and Infectious Diseases (NIAID). The government has certain rights in the invention."

BACKGROUND

[0003] Filoviruses, the ebolaviruses and marburgvirus, cause severe hemorrhagic fevers in humans, with mortality rates reaching 88% (Feldmann, et al., 2003, Nat Rev Immunol, 3 (8):677-685) as well as epizootics in nonhuman primates and probably other mammals. There are currently no vaccines or therapeutics approved against filoviruses. Filoviruses consist of five Ebolavirus species and a single species of Marburgvirus (Kuhn, et al., 2014, Viruses, 24; 6(11):4760-99). The main ebolavirus species causing outbreaks in humans are Ebola virus (EBOV; formerly known as Zaire Ebola virus), Sudan virus (SUDV) and Bundibugyo virus (BDBV) (Kuhn, et al, 2014, Viruses, 24; 6(11):4760-99). Filoviruses are enveloped, single-stranded, negative sense RNA filamentous viruses and encode seven proteins, of which the spike glycoprotein (GP) is considered the main protective antigen and vaccine target (Feldmann, et al., 2003, Nat Rev Immunol, 3 (8):677-685; Feldmann, et al., 2005, Curr Opin Investig Drugs, 6 (8):823-830; Geisbert, et al, 2010, Rev Med Virol, 20(6):344-57).

[0004] Four vector-based approaches and DNA vaccine strategies against filoviruses have been reported including replication-incompetent Venezuelan equine encephalitis virus (VEE) replicons, adenoviral vectors, live recombinant vesicular stomatitis virus (VSV), and parainfluenza (PIV) vectored vaccines (Geisbert, et al., 2010, Rev Med Virol, 20(6):344-57; Marzi and Feldmann, 2014, Expert Rev Vaccines, 13(4): p. 521-31). The VSV-based vaccine was recently tested with apparent success for efficacy in humans infected with EBOV during the 2014 West Africa Ebola virus disease (EVD) outbreak (Henao-Restrepo, 2015, Lancet, 386(9996):857-66). The current vaccine candidates are designed based on the wild-type sequence of GP of specific filovims species or strain. However, these vaccines are unable to cross-protect against multiple species of ebolavirus or Marburgvirus (Geisbert, et al., 2010, Rev Med Virol, 20(6):344-57). It is also not clear if a simple mixing of vaccines for different species will provide broad protection against multiple species of filoviruses. Thus, vaccines designed to induce broadly protective immune responses are highly desired.

[0005] GP is proteolytically cleaved by furin protease into two subunits linked by a disulfide linkage: GPl (-140 kDa) and GP2 (-38 kDa) (Manicassamy, et al, 2005, J Virol, 79 (8):4793-4805). Three GP1-GP2 units form the trimeric GP envelope spike (-550 kDa) on the viral surface (Feldmann, et al., 1993, Arch Virol Suppl, 7:81-100; Feldmann, et al., 1991, Virology, 182 (l):353-356; Geisbert and Jahrling, 1995, Virus Res, 39 (2-3): 129-150; Kiley, et al, 1988a, J Gen Virol, 69 (Pt 8): 1957-1967). GPl mediates cellular attachment (Kiley, et al, 1988b, J Gen Virol, 69 (Pt 8): 1957-1967; Kuhn, et al, 2006, J Biol Chem, 281 (23): 15951-15958), and contains a mucin-like domain (MLD) which is heavily glycosylated and variable and has little or no predicted secondary structure (Sanchez, et al, 1998, J Virol, 72 (8):6442-6447).

[0006] A specific region of the MARV and EBOV GPl consisting of -150 amino acids has been identified (Kuhn, et al, 2006, JBiol Chem, 281 (23): 15951-15958) that binds filovirus receptor-positive cells, but not receptor-negative cells, more efficiently than GPl, and compete with the entry of the respective viruses (Kuhn, et al, 2006, J Biol Chem, 281 (23): 15951-15958). These properties are similar to regions defined for SARS coronavirus and Machupo arenavirus (Li, et al, 2003, Nature, 426 (6965):450-454; Radoshitzky, et al, 2007, Nature, 446 (7131):92-96; Wong, et al, 2004, J Biol Chem, 279 (5):3197-3201). This region of GP is referred to as receptor binding region (RBR) and is part of a larger domain that excludes the highly variable, glycosylated, and bulky mucin-like domain (MLD). The RBR shows the highest level of homology between Filovirus glycoproteins (Kuhn, et al,

2006, J Biol Chem, 281 (23): 15951-15958).

[0007] The crystal structure of the trimeric, pre-fusion conformation of EBOV GP (lacking MLD) in complex with the EBOV-specific neutralizing antibody KZ52 was solved at 3.4 A (Lee, et al, 2008, Nature, 454 (7201): 177-182). This suggests a GP structure where three GPl subunits assemble to form a chalice, cradled in a pedestal of the GP2 fusion subunits, while the MLD restricts access to the conserved RBR, sequestered in the GP chalice bowl (Figure 1). The Base of the structure consists of the entire GP2 as well as residues 33-69, 95-104, 159-168, and 177-189 of GPl, referred to here as the GPl base. The residues within the GPl base closely interact with GP2 residue and contribute to the integrity of the trimeric structure (Lee, et al, 2008, Nature, 454 (7201): 177-182). The chalice itself consists of the so called core GPl or GPl head domain consisting of residues 70-94, 105-158,169-176, and 214-226. An additional highly glycosylated GPl domain called the glycan cap (residues 227-313) is located on the rim of the chalice.

[0008] Filovirus GPs are cleaved by cathepsin proteases as a step in entry, reducing GPl to a ~18kDa product (Chandran, et al, 2005, Science, 308 (5728): 1643-1645; Kaletsky, et al,

2007, J Virol, 81 (24): 13378-13384; Schornberg, et al, 2006, J Virol, 80 (8):4174-4178). The structures suggest that the most likely site of cathepsin cleavage is the flexible and structurally disordered β13-β14 loop (residues 190-213) of GPl and illustrate how cleavage there would release the heavily glycosylated regions from GP, leaving just the core of GPl, encircled by GP2 that interacts with the GPl base residues, with the RBR now well exposed. Cathepsin cleavage enhances infection, presumably as a result of better exposing the RBR for interaction with cell surface factors trafficked with the virus into the endosome (Dube, et al, 2009, J Virol, 83 :2883-2891). On the surface of the authentic virus, the MLD probably dominates host-interaction surfaces of filovirus GP, and indeed, antibodies against the MLD have been frequently identified. The seclusion of the receptor binding region (RBR) in the full length GP and its exposure during entry in the endosome suggest that targeting of neutralizing antibodies that recognize RBR to the endosomes may be useful in achieving effective neutralization of the filoviruses. The monoclonal antibody FVM04 is a prototypic inhibitor of receptor binding and consistent with the conserved nature of the RBR, FVM04 cross neutralizes multiple ebolaviruses and protects against Ebola virus and Sudan virus infections in animal models (Howell, et al., 2016, Cell Rep, 15(7): 1514-26). GPc L -NPCl interaction positions the internal fusion domain (IFL) of GP to interact with the endosomal membrane and trigger viral membrane fusion. While GPC L -NPC I interaction is required for membrane fusion, it is not sufficient. (Aman, 2016, MBio, 7(2):e00346-16). This process of fusion triggering involves major conformational rearrangement that are only partially understood likely dependent of acid and protease dependent processes that still remain to be defined in details. The trigger unwinds the GP2 helical structure from around the GP1 positioning IFL next to the endosomal membrane and allowing it to penetrate the endosomal membrane. As a result the pre-hairpin intermediate pulls together the viral and endosomal membrane, leading to hemifusion followed by formation of a fusion pore and post-fusion six helix bundle structure (Lee and Saphire, 2009, Curr Opin Struct Biol 19:408-17; Aman, 2016, MBio, 7(2):e00346-16). The virus then delivers its content through this pore into the host cytoplasm. The IFL consists of a two-strand beta sheet and a connecting loop that wrap arounds GP1. The Monoclonal antibodies KZ52 bind a species specific epitope at the base of the IFL (Lee, et al., 2008, Nature, 454 (7201): 177-182). While binding to this epitope by KZ52-like antibodies leads to potent inhibition of viral fusion, the epitope is highly specific to EBOV (Zaire) and KZ52 does not cross react with other ebolaviruses (Saphire, 2013, Immunotherapy, 5(11): 1221- 33). Thus development of therapeutic antibodies that inhibit the fusion of multiple ebolaviruses is highly desirable. Such antibodies would likely bind to the stem (the beta sheets β19 and β20 (Lee, et al., 2008, Nature, 454 (7201): 177-182)) or the tip of the IFL. We have previously reported that FVM02, a mAb that binds to the tip of the fusion loop but does not contact GP1, is unable to neutralize ebolaviruses. In contrast every neutralizing antibody that binds to the base of the GP trimer and neutralizes the virus contacts both GP1 and GP2, effectively bracing the two subunits (Saphire and Aman, 2016, Trends Microbiol, 24(9):684-686). This bracing effect most likely mechanically interferes with the structural rearrangements required for productive fusion (Saphire and Aman, 2016, Trends Microbiol, 24(9):684-686). The IFL closely interacts with GP1 particularly with the residues in the β3 strand (such as R64) as well as the N-terminal portion of the cathepsin cleavage loop (The loop consists of residues A189-Y214) suggesting that antibodies that contact both GP1 and GP2 residues in this region can brace the GP1 and GP2 and inhibit fusion. The antibody CA45 is a macaque-derived panfilovirus antibody that binds to the ebolavirus GP IFL across four different species of ebolavirus, EBOV, SUDV, RESTV, and BDBV (U.S. Provisional Application No. 62/406,598, filed October 11, 2016, which is incorporated by reference herein in its entirety).

[0010] Role of antibodies in protection against filovirus hemorrhagic fever: While both T and B cell responses are reported to play a role in protective immune responses to filoviruses (Warfield, et al., 2005, J Immunol, 175 (2): 1184-1191), a series of recent reports indicate that antibody alone can provide significant protection. Dye et al showed that purified convalescent IgG from macaques can protect NHPs against challenge with MARV and EBOV when administered as late as 48h post exposure (Dye, et al., 2012, Proc Natl Acad Sci USA, 109(13):5034-9). Olinger et al reported significant protection from EBOV challenge in NHPs treated with a cocktail of three monoclonal antibodies (mAbs) to GP administered 24h and 48h post exposure (Olinger, et al., 2012, Proc Natl Acad Sci USA, 109(44): 18030-18035). Similar results were also reported in two other studies (Qiu, et al., 2013, Sci Transl Med, 5 (207):207ral43; Qiu, et al, 2013, J Virol, 87(13):7754-7757). A recent study shows that a combination of three monoclonal antibodies, called ZMAPP™, can protect monkeys when administered five days after exposure to EBOV, at a time when the disease is fully manifest and the viremia is at its peak (Qiu, et al., 2014, Nature, 514(7520):47-53). Collectively these data demonstrate the ability of the humoral response to control filovirus infection. While ZMapp™ is strictly specific for EBOV, recent reports show that development of antibodies with broad neutralizing and protective property is feasible (WO2016/069627 and Keck, et al, 2015, J Virol, 90:279-291; WO2015/200522 A2 and Holtsberg, et al, 2015, J Virol, 90:266 -278; Howell et al 2016, Cell Reports, 15, 1514-1526).

SUMMARY

[0011] This disclosure provides an immunogen that includes a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, where the filovirus GP includes the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, where the base domain can include one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence, and where the one or more amino acid substitutions can affect the conformation of a cross- reactive epitope in the head domain, thereby increasing immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains. In certain aspects, the wild-type filovirus is a wild-type Ebola virus (EBOV), and the amino acid substitution(s) in the GP base domain can include an amino acid substitution: at a position corresponding to C53, F183, N521, A562, L569, or L573 of the wild-type Ebola virus (EBOV) GP, or a combination thereof; at a position corresponding to F159, P513, L515, or T565 of the wild-type EBOV GP, or a combination thereof; at a position corresponding to R164, LI 84, 1185, H516, or G546 of the wild-type EBOV GP, or a combination thereof; at a position corresponding to L51, G179, Q508, C511, Y517, R559, or C601 of the wild-type EBOV GP, or a combination thereof; at a position corresponding to 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, or 1610 of the wild-type EBOV GP, or a combination thereof; at a position corresponding to L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of the wild-type EBOV GP, or a combination thereof; or at a position corresponding to C53, F183, N521, A562, L569, L573, F159, P513, L515, T565, R164, L184, 1185, H516, G546, L51, G179, Q508, C511, Y517, R559, C601, 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, 1610, L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of the wild-type EBOV GP, or a combination thereof. In certain aspects, the wild-type Ebola virus is EBOV strain Mayinga-76. In certain aspects, the other filovirus species or strain against which cross-reactivity is broadened to include can be EBOV, SUDV, BDBV, RESTV, TAFV, MARV, any strain thereof, or a combination thereof.

The disclosure further provides an immunogen that includes a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, wherein the base domain comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence, and wherein the one or more amino acid substitutions can affect the conformation of a species specific epitope in the base domain, thereby masking an immunodominant epitope in the base domain. In certain aspects the wild-type filovirus is a wild-type Ebola virus (EBOV), and the amino acid substitution(s) in the GP base domain can include an amino acid substitution at a position corresponding to E103, F159, E178, F183, C511, L515A, W518, N550, D552, G553, C556, or R559 of the wild-type EBOV GP, or a combination thereof. In certain aspects, the one or more amino acid substitutions can also increase immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadens the cross-reactive immunogenicity of the immunogen against other filovirus species or strains. In certain aspects, the wild-type filovirus is a wild-type Ebola virus (EBOV), and the amino acid substitution(s) in the GP base domain can include an amino acid substitution: at a position corresponding to F159, F183, C511, L515, or R559 of the wild-type EBOV GP, or a combination thereof; e.g., at a position corresponding to F 183 of the wild-type EBOV GP. In certain aspects, the wild-type Ebola virus is EBOV strain Mayinga-76. In certain aspects, the other filovirus species or strain against which cross- reactivity is broadened to include can be EBOV, SUDV, BDBV, RES TV, TAFV, MARV, any strain thereof, or a combination thereof.

[0013] In various aspects, an immunogen as provided above can further include the glycan cap of the filovirus GP1 subunit or an immunogenic fragment thereof.

[0014] In certain aspects the disclosure further provides an immunogen that includes a filovirus spike glycoprotein (GP) or immunogenic fragment thereof that includes the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or an immunogenic fragment thereof, where the glycan cap can include one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP, and where the one or more amino acid substitutions can reduce glycosylation of the glycoprotein, thereby increasing immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains, e.g., EBOV, SUDV, BDBV, RESTV, TAFV, MARV, any strain thereof, or a combination thereof. In certain aspects, the wild-type filovirus is a wild-type Ebola virus (EBOV), and wherein the amino acid substitution(s) in the glycan cap can be an amino acid substitution at a position corresponding to N238, T240, N257, T259, N268, or T270 of the wild-type EBOV GP, or a combination thereof. In certain aspects the amino acid substitution in the glycan cap can include at least two single amino acid substitutions at positions corresponding to N238 and/or T240, N257 and/or T259, or N268 and/or T270 of the wild-type EBOV GP, wherein the substitutions disrupt at least two glycosylation sites. In certain aspects, the wild-type Ebola virus is EBOV strain Mayinga-76. [0015] In certain aspects the disclosure further provides an immunogen that includes a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, where the filovirus GP includes the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or immunogenic fragment thereof, where the glycan cap includes one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence within the disordered loop connecting the β17 to β18 strands, and where the one or more amino acid substitutions can affect the conformation of a cross-reactive epitope in the glycan cap, thereby increasing the immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadening the cross-reactive immunogenicity of the immunogen against other filovirus species or strains e.g., EBOV, SUDV, BDBV, RES TV, TAFV, MARV, any strain thereof, or a combination thereof. In certain aspects, the wild-type filovirus is a wild-type Ebola virus (EBOV), and the amino acid substitution(s) in the glycan cap includes an amino acid substitution: at a position corresponding to F290, W291, or E292 of the wild-type EBOV GP, or a combination thereof, e.g., at a position corresponding to E292 of the wild-type EBOV GP. In certain aspects the wild-type filovirus is a wild-type Ebola virus (EBOV) that includes a glycan cap, and the glycan cap comprises: (i) an amino acid substitution at a position corresponding N238, T240, N257, T259, N268, or T270 of the wild-type EBOV GP, or a combination thereof; and (ii) an amino acid substitution at a position corresponding to F290, W291, or E292 of the wild-type EBOV GP, or a combination thereof. In certain aspects, the wild-type Ebola virus is EBOV strain Mayinga-76.

[0016] In certain aspects of any of the provided immunogens, the wild-type amino acid is substituted with an alanine (A) residue, except where the wild-type amino acid is A. Where the wild-type amino acid is A it can be substituted with a serine (S) residue. In certain aspects of any of the provided immunogens, the wild-type amino acid is substituted with a glycine (G) residue, except where the wild-type amino acid is G.

[0017] In certain aspects of any of the provided immunogens, the immunogen lacks the mucin like domain (MLD) of the filovirus GP1 subunit.

[0018] In certain aspects the disclosure further provides an immunogen that includes a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, where the filovirus GP includes the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the GP internal fusion loop domain or an immunogenic fragment thereof, wherein the GP comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence, and wherein the one or more amino acid substitutions increases immunogenicity of the immunogen against the corresponding wild-type filovirus GP internal fusion loop domain, and/or broadens the cross-reactive immunogenicity of the immunogen against other filovirus species or strains, e.g., EBOV, SUDV, BDBV, RESTV, TAFV, MARV, any strain thereof, or a combination thereof. In certain aspects, the wild-type filovirus is a wild-type Ebola virus (EBOV), and the GP amino acid substitution(s) include an amino acid substitution at a position corresponding to N40, T42, D192, F193, F194, or Q595 of the wild-type Ebola virus (EBOV) GP, or a combination thereof, e.g., at a position corresponding to N40, D192, F193, or F194 of the wild-type Ebola virus (EBOV) GP, or a combination thereof. In certain embodiments, the immunogen further comprising any one or more GP amino acid substitutions disclosed elsewhere herein. In certain aspects, the wild- type Ebola virus is EBOV strain Mayinga-76. In certain aspects: the wild-type amino acid is substituted with an alanine (A) residue, except where the wild-type amino acid is A; the wild-type amino acid is A and the wild-type amino acid is substituted with a serine (S) residue; and/or he wild-type amino acid is substituted with a glycine (G) residue, except where the wild-type amino acid is G. In certain aspects, the immunogen lacks the mucin like domain (MLD) of the filovirus GP1 subunit.

[0019] In certain aspects the disclosure further provides for an immunogen that includes a GP filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the immunogen is a cleaved GP (GPC L ) lacking the filovirus GP1 subunit MLD and glycan cap. In certain aspects, the GPC L comprises one or more GP amino acid substitutions of an immunogen of any one or more GP amino acid substitutions disclosed elsewhere herein. In certain aspects, the wild-type Ebola virus is EBOV strain Mayinga-76. In certain aspects: the wild-type amino acid is substituted with an alanine (A) residue, except where the wild- type amino acid is A; the wild-type amino acid is A and the wild-type amino acid is substituted with a serine (S) residue; and/or the wild-type amino acid is substituted with a glycine (G) residue, except where the wild-type amino acid is G.

[0020] The disclosure further provides a method of increasing immunogenicity of and/or of broadening the cross-reactive immunogenicity of an immunogen that includes a filovirus GP or immunogenic fragment thereof, where the method includes using an immunogen provided herein and/or making substitutions as described for the immunogens provided herein. [0021] The disclosure further provides a method of masking an immunodominant epitope in a immunogen that includes a filovirus GP or a fragment thereof where the method includes making appropriate substitutions as described for the immunogens provided herein.

[0022] The disclosure further provides a composition comprising an immunogen as provided herein, and an adjuvant. In certain aspects, the immunogen is incorporated into a filovirus virus-like particle (VLP). In certain aspects, the filovirus VLP can further include a filovirus a filovirus VP40, a filovirus nucleoprotein ( P), and/or filovirus VP24.

[0023] The disclosure further provides an isolated polynucleotide that includes a nucleic acid encoding an immunogen or fragment thereof as provided herein, or a subunit thereof. Further provided is a vector that includes the provided polynucleotide and a host cell that includes the provided polynucleotide or the provided vector.

[0024] The disclosure further provides a method of making an immunogen as provided herein, where the method includes culturing the provided host cell and recovering the immunogen.

BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES

[0025] Figure 1: Figure 1 shows the trimeric structure of EBOV GP lacking the mucin like domain (MLD). The MLD is modeled on the structure as large gray balls. The different regions of the GP, the glycan cap, the core GPl in the head domain, the base consisting of the GP2 and the N-terminus of GPl . The base binder KZ52 structure is also shown. The positions of the cross-reactive/cross neutralizing antibodies are marked as small black circles on the head domain of the trimeric GP, as opposed to the epitopes for KZ52, 2G4, and 4G7 that are located within the base and marked with white circles. The epitope for the fusion loop binding antibody CA45 is shown as a star.

[0026] Figure 2A: Figure 2A shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with EBOV specific antibody KZ52 (A).

[0027] Figure 2B: Figure 2B shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with pan-ebolavirus antibody FVM04 (B).

[0028] Figure 2C: Figure 2C shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with pan-ebolavirus antibody m8C4 (C). [0029] Figure 2D: Figure 2D shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with pan-ebolavirus antibody FVM09 (D).

[0030] Figure 2E: Figure 2E shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with pan-ebolavirus antibody FVM17 (E).

[0031] Figure 2F: Figure 2F shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with pan-ebolavirus antibody FVM20 (F).

[0032] Figure 2G: Figure 2G shows binding of antibodies to the EBOV GP library. Shown are the binding values, relative to binding with wild-type GP, of all EBOV GP single alanine substitution library mutants with pan-ebolavirus antibody CA45 (G).

[0033] Figure 3A: Figure 3A shows epitope mapping of FVM04 mAb. (A) The shotgun mutagenesis library of EBOV GP was tested for immunoreactivity with mAb FVM04. Clones were initially identified to be critical for mAb FVM04 binding with reactivity of <30% relative to that of wild-type EBOV GP yet >65% reactivity for a control MAb, and were verified using algorithms described elsewhere (U.S. patent application 61/938,894, and Davidson and Doranz, 2014 Immunology 143 : 13-20).

[0034] Figure 3B: Figure 3B shows epitope mapping of FVM04 mAb. (B) Mutation of three individual residues reduced FVM04 binding (white bars) but had little effect on the binding of other mAbs FVM02 and FVM09; hatched and gray bars, respectively). Bars represent the mean and range of at least two replicate data points.

[0035] Figure 3C: Figure 3C shows epitope mapping of FVM04 mAb. (C) Sequence homology between filoviruses within the RBS crest region containing putative FVM04 epitope. Identical sequences among ebolavirus species and between ebolavirus and marburgvirus are shown in bold. The FVM04 binding site is boxed.

[0036] Figure 4: Figure 4 shows that mutation of glycan cap glycosylation sites N238, N257, and N268 enhances binding of FVM04 to EBOV GP. Relative bindings of FVM04 and KZ52 to the mutants affecting the four glycan cap glycosylation sites are shown as percent of binding of the same antibody to wild-type (wt) GP expressed on 293T cells and determined by flow cytometry.

[0037] Figure 5A: Figure 5A shows that residues that modify binding of mAbs FVM09 (A).

Alanine scan mutagenesis analysis of binding of pan-ebolavirus mAbs FVM09. Mutations of GP residues with significant reduction or increase in binding of the two antibodies to the respective GP mutant are shown.

[0038] Figure 5B: Figure 5B shows that residues that modify binding of m8C4 (B). Alanine scan mutagenesis analysis of binding of pan-ebolavirus mAbs m8C4. Mutations of GP residues with significant reduction or increase in binding of the two antibodies to the respective GP mutant are shown.

[0039] Figure 6A: Figure 6A shows the neutralization of VSV pseudotyped with EBOV

GPwt (wild-type) in comparison to EBOVGP-AAA (F290A/W291A/E292A) mediated by m8C4 (A).

[0040] Figure 6B: Figure 6B shows the neutralization of VSV pseudotyped with EBOV GPwt (wild-type) in comparison to EBOVGP-AAA (F290A/W291A/E292A) mediated by KZ52 (B).

[0041] Figure 7A: Figure 7A shows the binding of specific mutants of EBOV GP to pan- ebolavirus antibody CA45. Mutation of GP residues R64, Y517, G546, and N550 drastically reduced CA45 binding, alanine substitution of N40, D192, F193, and F194, but not SI 95, increased GP binding to CA45.

[0042] Figure 7B: Figure 7B shows the position of the CA45 epitope residues and enhancing residues on the structure of an EBOV GP monomer. GPl is shown in light gray, GP2 in dark grey, the epitope residues in black spheres and the enhancing residues in white spheres.

[0043] Figure 8: Figure 8 shows the breadth of antibody response elicited in mice by VSV- GPAmuc wild type or the indicated mutants. VSV-G was used as negative control. Y-axis shows EC 50 dilution titer against the different viruses listed in the graph legend.

DETAILED DESCRIPTION

Definitions

[0044] The term "a" or "an" entity refers to one or more of that entity; for example, "a polypeptide subunit" is understood to represent one or more polypeptide subunits. As such, the terms "a" (or "an"), "one or more," and "at least one" can be used interchangeably herein.

[0045] Furthermore, "and/or" where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, the term "and/or" as used in a phrase such as "A and/or B" herein is intended to include "A and B," "A or B," "A" (alone), and "B" (alone). Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).

[0046] It is understood that wherever aspects are described herein with the language "comprising," otherwise analogous aspects described in terms of "consisting of and/or "consisting essentially of are also provided.

[0047] All methods described herein can be performed in any suitable order unless otherwise indicated herein. No language or terminology in this specification should be construed as indicating any non-claimed element as essential or critical.

[0048] The headings provided herein are not limitations of the various aspects or aspects of the disclosure, which can be had by reference to the specification as a whole.

[0049] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure is related. For example, the Concise Dictionary of Biomedicine and Molecular Biology, Juo, Pei-Show, 2nd ed., 2002, CRC Press; The Dictionary of Cell and Molecular Biology, 3rd ed., 1999, Academic Press; and the Oxford Dictionary Of Biochemistry And Molecular Biology, Revised, 2000, Oxford University Press, provide one of skill with a general dictionary of many of the terms used in this disclosure.

[0050] Units, prefixes, and symbols are denoted in their Systeme International de Unites (SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Unless otherwise indicated, amino acid sequences are written left to right in amino to carboxy orientation. The headings provided herein are not limitations of the various aspects or aspects of the disclosure, which can be had by reference to the specification as a whole. Accordingly, the terms defined immediately below are more fully defined by reference to the specification in its entirety.

[0051] As used herein, the term "non-naturally occurring" substance, composition, entity, and/or any combination of substances, compositions, or entities, or any grammatical variants thereof, is a conditional term that explicitly excludes, but only excludes, those forms of the substance, composition, entity, and/or any combination of substances, compositions, or entities that are well-understood by persons of ordinary skill in the art as being "naturally-occurring," or that are, or could be at any time, determined or interpreted by a judge or an administrative or judicial body to be, "naturally-occurring." [0052] As used herein, the term "polypeptide" is intended to encompass a singular "polypeptide" as well as plural "polypeptides," and refers to a molecule composed of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds). The term "polypeptide" refers to any chain or chains of two or more amino acids, and does not refer to a specific length of the product. Thus, peptides, dipeptides, tripeptides, oligopeptides, "protein," "amino acid chain," or any other term used to refer to a chain or chains of two or more amino acids are included within the definition of "polypeptide," and the term "polypeptide" can be used instead of, or interchangeably with any of these terms. The term "polypeptide" is also intended to refer to the products of post-expression modifications of the polypeptide, including without limitation glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-standard amino acids. A polypeptide can be derived from a natural biological source or produced by recombinant technology, but is not necessarily translated from a designated nucleic acid sequence. It can be generated in any manner, including by chemical synthesis.

[0053] A "protein" as used herein can refer to a single polypeptide, i.e., a single amino acid chain as defined above, but can also refer to two or more polypeptides that are associated, e.g., by disulfide bonds, hydrogen bonds, or hydrophobic interactions, to produce a multimeric protein. As used herein, the term "glycoprotein" refers to a protein coupled to at least one carbohydrate moiety that is attached to the protein via an oxygen-containing or a nitrogen-containing side chain of an amino acid residue, e.g., a serine residue or an asparagine residue.

[0054] By an "isolated" polypeptide or a fragment, variant, or derivative thereof is intended a polypeptide that is not in its natural milieu. No particular level of purification is required. For example, an isolated polypeptide can be removed from its native or natural environment. Recombinantly produced polypeptides and proteins expressed in host cells are considered isolated as disclosed herein, as are recombinant polypeptides that have been separated, fractionated, or partially or substantially purified by any suitable technique.

[0055] As used herein, the term "non-naturally occurring" polypeptide, or any grammatical variants thereof, is a conditional term that explicitly excludes, but only excludes, those forms of the polypeptide that are well-understood by persons of ordinary skill in the art as being "naturally-occurring," or that are, or could be at any time, determined or interpreted by a judge or an administrative or judicial body to be, "naturally-occurring." [0056] Other polypeptides disclosed herein are fragments, derivatives, analogs, or variants of the foregoing polypeptides, and any combination thereof. The terms "fragment," "variant," "derivative" and "analog" when referring to polypeptide subunit or multimeric protein as disclosed herein can include any polypeptide or protein that retain at least some of the activities of the complete polypeptide or protein (for example retain at least some of the antibody-binding properties), but which is structurally different. Fragments of polypeptides include, for example, proteolytic fragments, as well as deletion fragments. Variants include fragments as described above, and also polypeptides with altered amino acid sequences due to amino acid substitutions, deletions, or insertions. Variants can occur spontaneously or be intentionally constructed. Intentionally constructed variants can be produced using art- known mutagenesis techniques. Variant polypeptides can comprise conservative or non- conservative amino acid substitutions, deletions or additions. Derivatives are polypeptides that have been altered so as to exhibit additional features not found on the native polypeptide. Examples include fusion proteins. Variant polypeptides can also be referred to herein as "polypeptide analogs. " As used herein a "derivative" refers to a subject polypeptide having one or more amino acids chemically derivatized by reaction of a functional side group. Also included as "derivatives" are those peptides that contain one or more standard or synthetic amino acid derivatives of the twenty standard amino acids. For example, 4-hydroxyproline can be substituted for proline; 5-hydroxylysine can be substituted for lysine; 3-methylhistidine can be substituted for histidine; homoserine can be substituted for serine; and ornithine can be substituted for lysine.

[0057] As used herein, a "single amino acid substitution" means replacing an amino acid residue in a polypeptide sequence with a different amino acid residue (such as replacing the native residue in a wild-type sequence with a non-native amino acid), unless otherwise specified. Also encompassed by the disclosure are a "single amino acid deletion" and/or a "single amino acid insertion."

[0058] A "conservative amino acid substitution" is one in which one amino acid is replaced with another amino acid having a similar side chain. Families of amino acids having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is a conservative substitution. Methods of identifying nucleotide and amino acid conservative substitutions which do not eliminate protein activity are well-known in the art (see, e.g., Brummell et al, Biochem. 32: 1180-1 187 (1993); Kobayashi et al, Protein Eng. 12(10):879-884 (1999); and Burks et al, Proc. Natl. Acad. Sci. USA 94:.412-417 (1997)).

[0059] Disclosed herein are certain antibodies, or antigen-binding fragments, variants, or derivatives thereof. Unless specifically referring to full-sized antibodies such as naturally- occurring antibodies, the term "antibody" encompasses full-sized antibodies as well as antigen-binding fragments, variants, analogs, or derivatives of such antibodies, e.g., naturally-occurring antibody or immunoglobulin molecules or engineered antibody molecules or fragments that bind antigen in a manner similar to antibody molecules.

[0060] As described further herein, an antibody or fragment thereof can comprise one or more "binding domains." As used herein, a "binding domain" or "antigen binding domain" is a two- or three-dimensional structure, e.g., a polypeptide structure that cans specifically bind a given antigenic determinant, e.g., the region formed by the heavy and light chain variable regions of an antibody or fragment thereof.

[0061] The terms "antibody" and "immunoglobulin" can be used interchangeably herein. An antibody (or a fragment, variant, or derivative thereof as disclosed herein comprises at least the variable domain of a heavy chain and at least the variable domains of a heavy chain and a light chain. Basic immunoglobulin structures in vertebrate systems are relatively well understood. See, e.g., Harlow et al, Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988).

[0062] Antibodies or antigen-binding fragments, variants, or derivatives thereof include, but are not limited to, polyclonal, monoclonal, human, humanized, or chimeric antibodies, single chain antibodies, epitope-binding fragments, e.g., Fab, Fab' and F(ab') 2 , Fd, Fvs, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv), fragments comprising either a VL or VH domain, fragments produced by a Fab expression library. ScFv molecules are known in the art and are described, e.g., in US patent 5,892,019. Immunoglobulin or antibody molecules encompassed by this disclosure can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule. [0063] By "specifically binds," it is meant that an antibody or fragment, variant, or derivative thereof binds to an epitope via its antigen binding domain, and that the binding entails some complementarity between the antigen binding domain and the epitope. According to this definition, an antibody or fragment thereof is said to "specifically bind" to an epitope when it binds to that epitope, via its antigen-binding domain more readily than it would bind to a random, unrelated epitope. The term "specificity" is used herein to qualify the relative affinity by which a certain antibody or fragment thereof binds to a certain epitope. For example, antibody or fragment thereof "A" can be deemed to have a higher specificity for a given epitope than antibody or fragment thereof "B" or antibody or fragment thereof "A" can be said to bind to epitope "C" with a higher specificity than it has for related epitope "D."

[0064] As used herein, the term "affinity" refers to a measure of the strength of the binding of an individual epitope with the CDR of an immunoglobulin molecule. See, e.g., Harlow et al, Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988) at pages 27-28. As used herein, the term "avidity" refers to the overall stability of the complex between a population of immunoglobulins and an antigen, that is, the functional combining strength of an immunoglobulin mixture with the antigen. See, e.g., Harlow at pages 29-34. Avidity is related to both the affinity of individual immunoglobulin molecules in the population with specific epitopes, and also the valencies of the immunoglobulins and the antigen. For example, the interaction between a bivalent monoclonal antibody and an antigen with a highly repeating epitope structure, such as a polymer, would be one of high avidity. An interaction between a between a bivalent monoclonal antibody with a receptor present at a high density on a cell surface would also be of high avidity.

[0065] The term "polynucleotide" is intended to encompass a singular nucleic acid as well as plural nucleic acids, and refers to an isolated nucleic acid molecule or construct, e.g., messenger RNA (mRNA) or plasmid DNA (pDNA). A polynucleotide can comprise a conventional phosphodiester bond or a non-conventional bond {e.g., an amide bond, such as found in peptide nucleic acids (PNA)). The term "nucleic acid" refers to any one or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide. By "isolated" nucleic acid or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment. For example, a recombinant polynucleotide encoding a polypeptide subunit contained in a vector is considered isolated as disclosed herein. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of polynucleotides. Isolated polynucleotides or nucleic acids further include such molecules produced synthetically. In addition, polynucleotide or a nucleic acid can be or can include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator.

[0066] As used herein, a "non-naturally occurring" polynucleotide, or any grammatical variants thereof, is a conditional definition that explicitly excludes, but only excludes, those forms of the polynucleotide that are well-understood by persons of ordinary skill in the art as being "naturally-occurring," or that are, or that could be at any time, determined or interpreted by a judge or an administrative or judicial body to be, "naturally-occurring."

[0067] As used herein, a "coding region" is a portion of nucleic acid comprising codons translated into amino acids. Although a "stop codon" (TAG, TGA, or TAA) is not translated into an amino acid, it can be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, and the like, are not part of a coding region. Two or more coding regions can be present in a single polynucleotide construct, e.g., on a single vector, or in separate polynucleotide constructs, e.g., on separate (different) vectors. Furthermore, any vector can contain a single coding region, or can comprise two or more coding regions, e.g., a single vector can separately encode an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region. In addition, a vector, polynucleotide, or nucleic acid can encode heterologous coding regions, either fused or unfused to a nucleic acid encoding a polypeptide subunit or fusion protein as provided herein. Heterologous coding regions include without limitation specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain.

[0068] In certain embodiments, the polynucleotide or nucleic acid is DNA. In the case of DNA, a polynucleotide comprising a nucleic acid that encodes a polypeptide normally can include a promoter and/or other transcription or translation control elements operably associated with one or more coding regions. An operable association or linkage can be when a coding region for a gene product, e.g., a polypeptide, can be associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s). Two DNA fragments (such as a polypeptide coding region and a promoter associated therewith) can be "operably associated" or "operably linked" if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not interfere with the ability of the expression regulatory sequences to direct the expression of the gene product or interfere with the ability of the DNA template to be transcribed. Thus, a promoter region would be operably associated with a nucleic acid encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid. The promoter can be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells. Other transcription control elements, besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription. Suitable promoters and other transcription control regions are disclosed herein.

[0069] A variety of transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions that function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (the immediate early promoter, in conjunction with intron-A), simian virus 40 (the early promoter), and retroviruses (such as Rous sarcoma virus). Other transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit β-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as lymphokine-inducible promoters (e.g., promoters inducible by interferons or interleukins).

[0070] Similarly, a variety of translation control elements are known to those of ordinary skill in the art. These include, but are not limited to ribosome binding sites, translation initiation and termination codons, and elements derived from picornaviruses (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence).

[0071] In other embodiments, a polynucleotide can be RNA, for example, in the form of messenger RNA (mRNA).

[0072] Polynucleotide and nucleic acid coding regions can be associated with additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide as disclosed herein. According to the signal hypothesis, proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated. Those of ordinary skill in the art are aware that polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the complete or "full length" polypeptide to produce a secreted or "mature" form of the polypeptide. In certain embodiments, the native signal peptide is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it. Alternatively, a heterologous mammalian signal peptide, or a functional derivative thereof, can be used. For example, the wild-type leader sequence can be substituted with the leader sequence of human tissue plasminogen activator (TP A) or mouse B-glucuronidase.

[0073] A "vector" is nucleic acid molecule as introduced into a host cell, thereby producing a transformed host cell. A vector can include nucleic acid sequences that permit it to replicate in a host cell, such as an origin of replication. A vector can also include one or more selectable marker gene and other genetic elements known in the art. Illustrative types of vectors include plasmids, phages, viruses and retroviruses.

[0074] A "transformed" cell, or a "host" cell, is a cell into which a nucleic acid molecule has been introduced by molecular biology techniques. As used herein, the term transformation encompasses those techniques by which a nucleic acid molecule can be introduced into such a cell, including transfection with viral vectors, transformation with plasmid vectors, and introduction of naked DNA by electroporation, lipofection, and particle gun acceleration. A transformed cell or a host cell can be a bacterial cell or a eukaryotic cell.

[0075] The term "expression" as used herein refers to a process by which a gene produces a biochemical, for example, a polypeptide. The process includes any manifestation of the functional presence of the gene within the cell including, without limitation, gene knockdown as well as both transient expression and stable expression. It includes without limitation transcription of the gene into messenger RNA (mRNA), and the translation of such mRNA into polypeptide(s). If the final desired product is a biochemical, expression includes the creation of that biochemical and any precursors. Expression of a gene produces a "gene product." As used herein, a gene product can be either a nucleic acid, e.g., a messenger RNA produced by transcription of a gene, or a polypeptide that is translated from a transcript. Gene products described herein further include nucleic acids with post transcriptional modifications, e.g., polyadenylation, or polypeptides with post translational modifications, e.g., methylation, glycosylation, the addition of lipids, association with other protein subunits, proteolytic cleavage, and the like. [0076] As used herein the terms "treat," "treatment," or "treatment of (e.g., in the phrase "treating a subject") refers to reducing the potential for disease pathology, reducing the occurrence of disease symptoms, e.g., to an extent that the subject has a longer survival rate or reduced discomfort. For example, treating can refer to the ability of a therapy when administered to a subject, to reduce disease symptoms, signs, or causes. Treating also refers to mitigating or decreasing at least one clinical symptom and/or inhibition or delay in the progression of the condition and/or prevention or delay of the onset of a disease or illness. The term "protection" and related grammatical terms, when used in the context of the ability of a therapeutic agent to affect the course of an infectious disease refers to any protective effect observed in comparison to a control agent. For example if two groups of animals are challenged with an infectious agent, e.g., a lethal dose of EBOV, and one group of animals is administered the therapeutic agent while the other group is administered a control, if a statistically significant number of animals in the therapeutic group survive relative to the number of survivors in the control group, a protective effect is observed. "Protection" can be, but does not have to be, 100%.

[0077] By "subject" or "individual" or "animal" or "patient" or "mammal," is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired. Mammalian subjects include humans, domestic animals, farm animals, sports animals, and zoo animals, including, e.g., humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, bears, and so on.

[0078] The term "pharmaceutical composition" refers to a preparation that is in such form as to permit the biological activity of the active ingredient to be effective, and that contains no additional components that are unacceptably toxic to a subject to which the composition would be administered. Such composition can be sterile.

[0079] By a "filovirus" is meant a virus belonging to the family Filoviridae . Exemplary filoviruses are Lloviu virus (LLOV), Bundibugyo virus (BDBV), Reston virus (RESTV), Sudan virus (SUDV), Tai Forest virus (TAFV), Ebola virus (EBOV), Marburg virus (MARV), and Ravn virus (RAW). In certain embodiments, a filovirus is selected from the group consisting of EBOV, SUDV, BDBV, RESTV, TAFV, MARV, and a combination thereof.

[0080] The virions of filoviruses contain seven proteins which include a surface glycoprotein (GP), a nucleoprotein ( P), an RNA-dependent RNA polymerase (L), and four virion structural proteins (VP24, VP30, VP35, and VP40). [0081] The term "epitope," as used herein, refers to portions of a polypeptide (or other biological molecule such as a carbohydrate) having antigenic or immunogenic activity in an animal, for example a mammal, for example, a human. An "immunogen" or an "immunogenic epitope," as used herein, is defined as a portion of a protein that elicits an immune response in an animal, as determined by any method known in the art. The term "antigen," or "antigenic epitope," as used herein, is defined as a portion of a protein to which an antibody or T-cell receptor can immunospecifically bind its antigen as determined by any method well known in the art. Immunospecific binding excludes non-specific binding but does not exclude cross-reactivity with other antigens. Whereas all immunogenic epitopes are antigenic, antigenic epitopes need not be immunogenic. An "epitopic fragment" of a polypeptide antigen is a portion of the antigen that contains an epitope. An "epitopic fragment" can, but need not, contain amino acid sequence in addition to one or more epitopes.

[0082] By "vaccine," is meant a composition comprising an immunogen which, when inoculated into a mammal, has the effect of stimulating a cellular immune response comprising a T-cell response and/or a humoral immune response comprising a B-cell response generally resulting in antibody production. The T cell response can be a cytotoxic T-cell response directed against an organism that expresses the antigen. However, the induction of a T-cell response comprising other types of T cells by an immunogen disclosed herein is also contemplated. A B-cell response results in the production of antibody that binds to the antigen. The vaccine can serve to elicit an immune response in the mammal which serves to protect the mammal against a disease. The terms "vaccine," "immunogen," "immunogenic polypeptide," and the like may be used interchangeably. The term "vaccine" does not in any way connote that the composition is capable of fully preventing a disease in a vaccinated subject, or providing any specific or general level of protection against an infectious agent. In certain aspects a "vaccine" can be ineffective in certain subjects while inducing an immune response in other subjects.

[0083] By "subunit vaccine" is meant a vaccine produced from a specific protein subunit of a virus and thus having less risk of adverse reactions than whole virus vaccines. A subunit vaccine can be, for example, a soluble immunogenic polypeptide, or immunogenic fragment thereof, or the polypeptide or fragment (or two or more polypeptides) that can self-assemble as a multimer. The term "peptide vaccine" or "subunit vaccine" can also refer to a composition comprising one or more polypeptide subunits, which when administered to an animal are useful in stimulating an immune response against filovirus infection.

[0084] By "virus-like particle" or "VLP" is meant a non-infectious particle that resembles an infectious virus, e.g., in immune surveillance, but which lacks the genetic material to create new virus particles. VLPs can be used as immunogens or vaccines as described elsewhere herein. VLPs can include any combination of virus components provided that the VLP lacks the genetic machinery to self-replicate. In certain aspects, VLPs can be made by allowing self-assembly of a matrix or scaffold-like protein, e.g., a viral capsid protein, and can further include a viral envelope.

[0085] The term "viral vector" refers to a recombinant virus that can express a heterologous immunogen of interest. For example, a filovirus glycoprotein as described herein can be expressed in a recombinant virus, e.g., a vesicular stomatitis virus (VSV), a pox virus (e.g., a vaccinia virus), an adenovirus, a herpesvirus, or other recombinant virus, using techniques well established in the art.

[0086] As used herein, "cross-reactive" means the ability of an immunogen, e.g., an immunogenic polypeptide as provided herein, to elicit an immune response against two or more filovirus species or strains. For example, an immunogen with cross-reactive immunogenicity induces the production of antibodies capable of specifically binding to, recognizing, and/or neutralizing two or more heterologous filovirus species or strains.

[0087] As used herein, an "increase," "increased," "increasing," and the like of immunogenicity means that an immunogen as provided herein can provide an improved immune response against its original target (e.g., the wild-type filovirus from which the immunogen was derived) than the corresponding wild-type immunogen. For example, the immunogen can induce a more effective immune response (increased protection, increased potency, increased virus killing, increased induction of antibodies, increased T-cell immunity, etc.) than the corresponding wild-type immunogen. Likewise, an immunogen as provided herein can exhibit increased immunogenicity against a cross-reactive target (e.g., another filovirus species or strain).

[0088] As used herein, to "broaden, "broadened, "broadening," and the like of cross-reactive immunogenicity means that the immunogen as provided herein has a broader scope of cross-reactivity than the corresponding wild-type immunogen. For example, an immunogen as provided herein can induce an immune response against EBOV and SUDV strains, where the corresponding wild-type immunogen only induced an immune response against EBOV. [0089] As used herein, "species specific" or "strain specific" when used in reference to an immunogen, e.g., an immunogenic polypeptide as provided herein, means that an immune response elicited by the immunogen is predominantly directed towards a single filovirus species (or a single filovirus strain) in comparison to multiple filovirus species or strains.

[0090] As used herein, "cross-protective" means the ability of an immunogen, e.g., an immunogenic polypeptide as provided herein, to elicit an immune response that provides at least some level of protection (e.g., preventing disease symptoms, reducing disease symptoms, or preventing death) against infection by two or more heterologous filovirus species or strains.

[0091] "Inducing specific immunity" as used herein refers to providing to a population or an individual the ability to direct an immune response against specific pathogens (e.g., filoviruses).

[0092] "Conferring protective immunity" as used herein refers to providing to a population or an individual the ability to generate an immune response to protect against a disease (e.g., hemorrhagic fever) caused by a pathogen (e.g., Ebola virus) such that the clinical manifestations, pathology, or symptoms of disease in a host are minimized, reduced, or prevented as compared to a non-treated host, or such that the rate at which infection, or clinical manifestations, pathology, or symptoms of disease appear within a population are reduced, as compared to a non-treated population.

[0093] In certain embodiments, an immunogen, e.g., an immunogenic polypeptide as provided herein, can be useful for inducing immunity against one, two, three, four, five or more filovirus species or strains. Such immunity can protect/treat an individual or a population with filovirus infection. The term "immune response" refers to any response by the immune system of an individual to an immune response inducing composition or other immunogenic compound. Exemplary immune responses include, but not limited to cellular as well as local and systemic humoral immunity, such as CTL responses, including antigen-specific induction of CD8+ CTLs, helper T-cell responses, including T-cell proliferative responses and cytokine release, and B-cell responses including, e.g., an antibody producing response. The term "inducing an immune response" or "eliciting an immune response" refers to administration of an immune response inducing composition or other immunogenic compound or a nucleic acid encoding the immune response inducing composition or other immunogenic compound, wherein an immune response is affected, i.e., stimulated, initiated or induced. [0094] As used herein, the term "a filovirus-mediated disease" encompasses a condition which is a direct result of filovirus infection; and a condition which is an indirect result, e.g., a sequela, of a filovirus infection. An exemplary filovirus disease is hemorrhagic fever. Symptoms of hemorrhagic fever include, but are not limited to, fever, internal hemorrhaging, edema, organ failure, headache, malaise, myalgia, nausea, vomiting, bleeding of needle puncture sites, hematemesis, melena, petechiae, ecchymosis, maculopapular rash, disseminated intravascular coagulation, shock, jaundice, conjunctivitis, diarrhea, pharyngitis, convulsions, delirium, coma, oligura, epistaxis, and death.

[0095] As used herein, the term "adjuvant" is intended to encompass a substance or vehicle that non-specifically enhances the immune response to an antigen or immunogen. Adjuvants can include a suspension of minerals (such as alum, aluminum hydroxide, or phosphate) on which antigen is adsorbed; or water-in-oil emulsion in which antigen solution is emulsified in mineral oil (for example, Freund's incomplete adjuvant), sometimes with the inclusion of killed mycobacteria (Freund's complete adjuvant) to further enhance immunogenicity.

[0096] As used herein the term "engineered" includes manipulation of nucleic acid or polypeptide molecules by synthetic means (e.g. by recombinant techniques, in vitro peptide synthesis, by enzymatic or chemical coupling of peptides or some combination of these techniques).

[0097] As used here, CA45-like broadly neutralizing antibodies are antibodies that bind to the same or similar GP epitope as the CA45 antibody (U.S. Provisional Application No. 62/406,598), but are not limited to antibodies merely sharing similar sequence to the CA45 antibody.

[0098] As referred to herein, all residue numbers are based on Ebola virus GP (EBOV GP).

Unless otherwise specified herein, reference to EBOV GP indicates wild-type protein amino acid sequences (e.g., SEQ ID NOs 1, 3, and 5). When specified, the EBOV GP can be a mucin-deleted version (delta (Δ) mucin) (e.g., SEQ ID NOs 2, 4, and 6).

Cross-reactive monoclonal antibodies

[0099] The most divergent region of the filovirus GP molecule is the mucin like domain (MLD). The MLD can be deleted (delta (Δ) mucin) to focus the immune response to the more conserved regions of the GP. Using immunization of mice and macaques with a prime boost strategy involving filovirus virus like particles (VLPs) and soluble glycoproteins lacking the MLD, several cross-reactive antibodies have been generated (Keck, et al., 2016, J Virol, 90:279 -291; Holtsberg, et al, 2016, J Virol, 90:266 -278). Analysis of the binding pattern of these cross-reactive monoclonal antibodies showed that nearly all of them bind to the top of the GP trimer (Keck, et al, 2016, J Virol, 90:279 -291; Holtsberg, et al, 2016, J Virol, 90:266 -278), in contrast to the strain specific neutralizing antibodies KZ52 and 2G4 that bind to the base of the chalice (Lee, et al, 2008, Nature, 454 (7201): 177-182; Murin, et al, 2014, Proc Natl Acad Sci USA, 111(48): 17182-7).

Structure-based Pan Filovirus Vaccine Design

[0100] Provided herein are methods of incorporating substitutions of specified residues into a filovirus GP in order to increase immunogenicity and/or broaden the cross-reactivity against other filovirus species or strain. Also provided herein are mutant filovirus GPs comprising such substitutions. In certain embodiments, the filovirus GP is an EBOV GP. In certain embodiments, the EBOV is EBOV strain Mayinga-76. In certain embodiments, the other filovirus species or strain comprises EBOV, SUDV, BDBV, RESTV, TAFV, MARV, any strain thereof, or a combination thereof.

[0101] The substitutions can be incorporated into the filovirus GP in a variety of vaccine platforms, including, but not limited to a recombinant protein or subunit vaccine, a viruslike particle (VLP), a recombinant viral vector, e.g., a vesicular stomatitis virus (VSV) vector, an adenovirus vector, a poxvirus vector, e.g., a modified vaccine Ankara (MVA) vector, a rabies virus vector, or a parainfluenza virus vector.

[0102] In certain embodiments, the substituted amino acid is any amino acid other than the wild-type amino acid at that position. In certain embodiments, the substitution is with a conserved amino acid and in other embodiments, the substitution is with a non-conserved amino acid. The substitution can be guided by structural analysis. In certain embodiments, mutations can be designed to be an alanine (A) substitution where the wild-type amino acid is other than A or serine (S) where the wild-type amino acid is A.

[0103] Provided herein are immunogens, e.g., immunogenic polypeptides, comprising a substituted filovirus spike glycoprotein (GP) or an immunogenic fragment thereof where the filovirus GP comprises the GP head domain (or an immunogenic fragment thereof) and the GP base domain (or an immunogenic fragment thereof). As used throughout, reference to an immunogen or a polypeptide, oligopeptide, and the like also implies an immunogenic fragment thereof, unless otherwise specified. Unless otherwise specified, an immunogen comprising a filovirus spike glycoprotein (GP) lacks the mucin like domain (MLD) of the filovirus GP1 subunit.

[0104] In certain embodiments, a filovirus GP is substituted in its GP base domain with one or more single amino acid substitutions relative to the corresponding wild-type filovirus amino acid sequence. The substitution(s) can affect the conformation of an epitope (e.g., increase the exposure of the epitope) elsewhere in the GP, such as a cross-reactive epitope in the GP head domain. In certain embodiments, the conformational change can increase the immunogenicity of the immunogen against one or more filovirus species or strains relative to the corresponding wild-type filovirus sequence. For example, in certain embodiments, a substitution in the GP base domain of EBOV GP can elicit an antibody response that is increased, e.g., increased potency, increased protection, etc., to the corresponding EBOV GP or to GPs of other EBOV strains or other filovirus species. In certain aspects, the substitution in the GP base can increase binding to the substituted EBOV GP of one or more cross-reactive top binding antibodies relative to their binding to wild-type EBOV GP. Exemplary antibodies include, but are not limited to FVM04, m8C4, FVM09, FVM17, FVM20, and h4B8. In certain embodiments, the conformational change can broaden the cross-reactive immunogenicity of the immunogen against additional, e.g., against two or more filovirus species or strains relative to the corresponding wild-type filovirus. In certain embodiments, the conformational change can both increase immunogenicity and broaden the cross-reactive immunogenicity of the immunogen against two or more filovirus species or strains relative to the corresponding wild-type filovirus.

[0105] As will be appreciated by those in the art, analogous regions from different viruses may not directly coincide by amino acid number. To the extent amino acid location is provided herein, it is understood that the analogous regions from different filoviruses can readily be identified by sequence alignment and comparison. For example, as referred to throughout, an amino acid substitution at a position corresponding to an amino acid position of a wild- type Ebola virus (EBOV) GP, wherein the filovirus GP (or immunogenic fragment thereof) is other than that wild-type EBOV GP, can be determined by aligning the wild-type EBOV GP amino acid sequence with another filovirus GP amino acid sequence (for example, see Figure 3C) to identify amino acid positions corresponding to the wild-type EBOV GP sequence. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, for example, by a local homology algorithm (Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by a global alignment algorithm (Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), by search for similarity methods (Pearson & Lipman, Proc. Natl. Acad. Sci. U.S.A. 55:2444 (1988); Altschul et al, Nucl. Acids Res. 25:3389-402 (1997), by computerized implementations of these algorithms (e.g., GAP, BESTFIT, FASTA, and BLAST in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), typically using the default settings, or by manual alignment and visual inspection (see, e.g., Current Protocols in Molecular Biology, Ausubel et al. (eds.), 1994).

[0106] Certain embodiments provide for an immunogen, e.g., an immunogenic polypeptide, comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof and wherein the base domain comprises one or more single amino acid substitutions relative to the corresponding wild- type filovirus GP amino acid sequence. The one or more amino acid substitutions are characterized by their ability to affect the conformation of a cross-reactive epitope in the head domain. This change in confirmation can increase immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broaden the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

[0107] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to C53, F183, N521, A562, L569, or L573 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution at a position corresponding to C53, F183, N521, A562, L569, or L573 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0108] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise a substitution at a position corresponding to F183, N521, A562, L569, or L573 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to F183, N521, A562, L569, or L573 of a wild- type Ebola virus (EBOV) GP, or a combination thereof.

[0109] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to F159, P513, L515, or T565 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to F159, P513, L515, or T565 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0110] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to R164, L184, 1185, H516, or G546 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to R164, L184, 1185, H516, or G546 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0111] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to L51, G179, Q508, C511, Y517, R559, or C601 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to L51, G179, Q508, C511, Y517, R559, or C601 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0112] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, or 1610 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to 133, P34, 138, V48, V52, L68, E103, A182, R498, R501, N514, W531, P533, E545, C556, L561, S583, or 1610 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0113] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of a wild- type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to L43, V45, R54, L57, L63, L66, E71, Y99, L161, L165, P187, N506, P509, K510, W518, A525, L558, Q560, E564, T566, Q570, 1603, L604, G605, C608, C609, D614, T616, 1623, or W648 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. [0114] In certain embodiments, a substitution can affect the conformation of an epitope in the GP base domain, such as the masking or silencing of an immunodominant epitope. In certain embodiments, the immunodominant epitope is a species specific epitope. For example, mutations that result in reduction of KZ52 binding to EBOV GP to less than 20% are highlighted bold and double underlined in Table 2.

[0115] Certain embodiments provide for an immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof and the GP base domain or an immunogenic fragment thereof, and wherein the base domain comprises one or more single amino acid substitutions relative to the wild-type filovirus GP amino acid sequence. The one or more amino acid substitutions are characterized by their ability to affect the conformation of a species specific epitope in the base domain, thereby masking the an immunodominant epitope in the base domain.

[0116] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to E103, F159, E178, F183, C511, L515A, W518, N550, D552, G553, C556, or R559 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to E103, F159, E178, F183, C511, L515A, W518, N550, D552, G553, C556, or R559 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0117] Masking of certain epitopes can focus the immune response on certain other epitopes, such as cross-reactive epitopes that would generally not be immunodominant. Further, mutations of specific GP base residues can be generated that simultaneously silence a base epitope and increase binding of GP to all or some of the cross-reactive antibodies listed in Table 1 (see Examples below). Examples of residues that can silence the species specific base epitope (reactive to KZ52) and increase cross-reactive epitopes are listed in Table 1. Thus, certain embodiments provide for an immunogen wherein an amino acid substitution masks an immunodominant epitope in the base domain and also increase immunogenicity of the immunogen against the corresponding wild-type filovirus GP, and/or broadens the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

[0118] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to F159, F183, C511, L515, or R559 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to F 159, F 183, C51 1, L515, or R559 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0119] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to F159, F183, L515, or R559 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to F159, F 183, L515, or R559 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0120] In certain embodiments, the amino acid substitution is in the GP base domain and can comprise an amino acid substitution at a position corresponding to F 183 of a wild-type Ebola virus (EBOV) GP. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the GP base domain at a position corresponding to F183 of a wild-type Ebola virus (EBOV) GP.

[0121] Unless otherwise specified, an immunogen comprising a filovirus spike glycoprotein (GP) can comprise the glycoprotein glycan cap or a fragment thereof (e.g., an immunogenic fragment thereof), but does not necessarily comprises a portion of the glycan cap.

[0122] The glycosylation state of proteins can affect immunogenicity. Thus, certain embodiments provide for one or more amino acid substitutions that alter the glycosylation of a filovirus GP. In certain embodiments, the amino acid substitution reduces glycosylation. GP1 contains a highly glycosylated regions termed the "glycan cap" (wild- type EBOV residues 227-313). In certain embodiments, the amino acid substitution reducing glycosylation is in the glycan cap region. The GP1 glycan cap comprises at least four N-glycosylation sites. For example, it was observed that substitutions in three (N238, N257, N268) out of these four glycosylation sites increased the binding of the cross-reactive antibody FVM04. Residues at certain positions play a role in directing glycosylation (recognition sites). In particular, threonine (T) residues near the glycosylation site can be part of a glycosylation recognition site. For example, when threonine residues two amino acids C-terminal to certain glycosylation sites (T240, T259, and T270) were mutated, increased binding of the cross-reactive antibody FVM04 was also observed. Thus, certain embodiments provide for an amino acid substitution at a position corresponding to N238, T240, N257, T259, N268, or T270 of a wild-type EBOV GP, or a combination thereof. [0123] Certain embodiments provide for an immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or an immunogenic fragment thereof, wherein the glycan cap comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP. The one or more amino acid substitutions are characterized by their ability to reduce glycosylation of the glycoprotein. This can increase immunogenicity of the immunogen, and/or broaden the cross-reactive immunogenicity of the immunogen against other filovirus species or strains.

[0124] In certain embodiments, the amino acid substitution is in the glycan cap and can comprise an amino acid substitution at a position corresponding to N238, T240, N257, T259, N268, or T270 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the glycan cap at a position corresponding to N238, T240, N257, T259, N268, or T270 of a wild-type Ebola virus (EBOV) GP, or a combination thereof.

[0125] In certain embodiments, at least two glycosylation sites are disrupted by substituting the Asparagine (N) of the N-glycosylation site and/or the upstream threonine (T) of the recognition site for N-glycosylation. In certain embodiments, amino acid substitutions occur in the glycan cap comprising at least two substitutions selected from a group consisting of amino acid substitutions at a position corresponding to N238 and/or T240, at a position corresponding to N257 and/or T259, and at a position corresponding to N268 and/or T270 of a wild-type Ebola virus (EBOV) GP. Thus, in certain embodiments, an immunogen is provided comprising at least two amino acid substitutions in the glycan cap selected from a group consisting of amino acid substitutions at a position corresponding to N238 and/or T240, at a position corresponding to N257 and/or T259, and at a position corresponding to N268 and/or T270 of a wild-type Ebola virus (EBOV) GP. In certain embodiments, the amino acid substitutions disrupt at least two glycosylation sites. For purposes of this disclosure the pairings of N238/T240, N257/T259, and N268/T270 can be considered glycosylation sites.

[0126] Two monoclonal pan-ebolavirus antibodies, FVM09 (Keck, et al., 2016, J Virol, 90:279 -291; PCT US15/57627 which are incorporated herein by reference) and m8C4 (Holtsberg, et al, 2016, J Virol, 90:266 -278; PCT US15/37493 which are incorporated herein by reference) have been previously reported. Alanine scanning mutagenesis analysis revealed that EBOV GP residues W288, F290, and W291 and to a lesser extent E278 were involved in binding of FVM09 to EBOV GP (Figure 5A). In contrast, mutation of the adjacent E292 to alanine resulted in over two fold increase in binding of FVM09 over binding to wild-type EBOV GP (Figure 5A). All these residues are located within a disordered loop in EBOV GP structure connecting the β17 to β18 strands in the glycan cap (Lee, et al, 2008, Nature, 454 (7201): 177-182). Binding of m8C4 to EBOV GP was dependent primarily on R136 (within the inner chalice) as well as Q251, F252 (within the glycan cap adjacent to the β17-β18 loop where FVM09 epitope is located (Figure 5B). To a lesser extent, E106A and E258A mutations also reduced m8C4 binding (Figure 5B). In contrast, mutation of F290, W291, and E292 to alanine resulted in 3-4 fold increase in m8C4 binding to the respective mutant compared to wild-type GP (Figure 5B). These data indicate that knocking out FVM09 binding site (F290, W291) results in better exposure of m8C4 epitope. Mutation of E292 enhances the binding of both FVM09 and m8C4 suggesting that incorporation of a mutation in this site can enhance cross-reactive immunogenicity.

[0127] Certain embodiments provide for an immunogen comprising a filovirus spike glycoprotein (GP) or immunogenic fragment thereof, wherein the filovirus GP comprises the GP head domain or an immunogenic fragment thereof, the GP base domain or an immunogenic fragment thereof, and the glycan cap or immunogenic fragment thereof, wherein the glycan cap comprises one or more single amino acid substitutions relative to the corresponding wild-type filovirus GP amino acid sequence within the disordered loop connecting the β17 to β18 strands. The one or more amino acid substitutions are characterized by their ability to affect the conformation of a cross-reactive epitope in the glycan cap. This can increase immunogenicity of the immunogen and/or broaden cross- reactive immunogenicity of the immunogen against other filovirus species or strains.

[0128] In certain embodiments, the amino acid substitution is within the disordered loop connecting the β17 to βΐ 8 strands and can comprise an amino acid substitution at a position corresponding to F290, W291, or E292 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution within the disordered loop connecting the β17 to β18 strands at a position corresponding to F290, W291, or E292 of a wild-type Ebola virus (EBOV) GP, or a combination thereof. [0129] In certain embodiments, the amino acid substitution is in the disordered loop connecting the β17 to βΐ 8 strands and can comprise an amino acid substitution at a position corresponding to E292 of a wild-type Ebola virus (EBOV) GP. Thus, in certain embodiments, an immunogen is provided comprising an amino acid substitution in the disordered loop connecting the β17 to β18 strands at a position corresponding to E292 of a wild-type Ebola virus (EBOV) GP.

[0130] In certain embodiments, the amino acid substitution can comprise a combination of at least one single amino acid substitution in the glycan cap that disrupts N-glycosylation of the glycoprotein (e.g., at a position corresponding to N238, T240, N257, T259, N268, or T270 of wild-type EBOV GP, or a combination thereof) and at least one single amino acid substitution within the disordered loop connecting the β17 to β18 strands (e.g., at a position corresponding to F290, W291, or E292 of wild-type EBOV GP, or a combination thereof).

[0131] Certain embodiments provide for the mutation of GP1 and GP2 residues, in the region surrounding the GP fusion loop, including, but not limited to the epitope recognized by monoclonal antibody CA45, that increase the binding efficacy of CA45-like antibodies (Figure 7A). Bulky residues in this region surround the CA45 epitope (Figure 7B) and possibly restrict access of antibodies to the epitope. Such mutations can be incorporated into Ebola GP -based vaccines to enhance the ability of an immunogen to elicit CA45-like broadly neutralizing antibodies in the host. (U.S. Provisional Application No. 62/406,598).

[0132] Thus, certain embodiments provide for at least one single amino acid substitution at a position corresponding to N40, D192, F 193, and/or F194 of wild-type EBOV GP, or a combination thereof. In certain embodiments, the residues are substituted with an amino acid residue with no side chain or a small sidechain, such as glycine, alanine, or serine. In certain embodiments, the substitution is to alanine. Mutations that can enhance the binding of CA45-like antibodies include one or more of N40A, D192A, F193A, and/or F194A (Figure 7 A).

[0133] Certain embodiments provide for a combination of one or more mutations that enhance the elicitation and binding of CA45-like antibodies to the fusion loop region, with any other mutation described herein to simultaneously enhance reactivity of the antigen to both the apex binders (e.g., FVM04) and fusion loop binders (e.g., CA45).

[0134] Filoviruses enter the endosomes by micropinocytosis and after entry into endosomes GPs are cleaved by cathepsin proteases as an essential step in infectivity, reducing GP1 to a -18 kDa product (referred to here as GPCL) that is associated with the whole GP2, but lacks the entire glycan cap and the MLD (Chandran, et al., 2005, Science, 308 (5728): 1643-1645; Kaletsky, et al., 2007, J Virol, 81 (24): 13378-13384; Schornberg, et al., 2006, J Virol, 80 (8):4174-4178). The cleavage occurs between residues R200 and E201 in the cathepsin cleavage loops (Hood, et al., 2010, J. Virol, 84: 2972-2982). It was discovered that several cross-neutralizing antibodies including FVM04 and CA45 bind with higher affinity to the GPC L than to full length GP

[0135] In certain embodiments, GPC L can be used as a modified vaccine for inducing broadly neutralizing protective responses. Cleaved GP can be produced by a number of methods including: cleavage of purified GP by cathepsin B, cathepsin L, or thermolysin; and recombinant generation of GPC L by replacing the glycan cap and MLD in GP by a flexible linker of 5-50 amino acids. Other mutations that affect the binding of the antigens to cross- neutralizing antibodies described herein may also be incorporated in such recombinant GPC L - Further, recombinant GPC L can be incorporated in various vaccine formats including: recombinant purified protein; virus-like particles; vesicular stomatitis virus (VSV) vector expressing Ebola GPC L instead of VSV-G; an adenovirus vector carrying GPC L ; a parainfluenza virus (PIV) vector carrying GPC L ; and any other viral vector GPC L

[0136] Also provided herein are methods of increasing the immunogenicity and/or broadening the cross-reactive immunogenicity of an immunogen comprising a filovirus GP or an immunogenic fragment thereof, and the like, and methods of masking an immunodominant epitope in the same. The methods comprise making any one or more of the single amino acid substitutions disclosed herein in a filovirus GP.

[0137] Certain embodiments provide for a composition comprising an immunogen or a fragment thereof described anywhere herein and an adjuvant.

[0138] Certain embodiments provide for a composition comprising an immunogen or a fragment thereof described anywhere herein as part of a filovirus virus-like particle (VLP) (Warfield and Aman, 2011, J Infect Dis, 204(Suppl 3):S1053-1059). The expression and subsequent oligomerization of the matrix protein VP40 has been described as sufficient to drive the formation of filamentous, enveloped VLPs that are released from cells (Bavari et al., 2002, J Exp Med, 195:593-602; Swenson et al., 2004, FEMS Immunol Med Microbiol, 40:27-31; Harty et al., 2000, Proc Natl Acad Sci U S A, 97: 13871-6; Han et al., 2003, J Virol, 77: 1793-800; Licata et al., 2004, J Virol, 78:7344-51; Jasenosky and Kawaoka, 2004, Virus Res, 106: 181-8; Jasenosky et al., 2001, J Virol, 75:5205-14). Efficiency of the production of VLPs driven by sole expression of VP40, however, is low and VLP formation and release can be increased by the presence of additional viral proteins, for example, GP, NP, and VP24 (Swenson DL et al, 2004, FEMS Immunol Med Microbiol, 40:27-31; Warfield et al, 2007, J Infect Dis, 196(Suppl 2):S421-9; Kallstrom et al, 2005, J Virol Methods, 127: 1-9; Licata et al, 2004, J Virol, 78:7344-51).

[0139] In certain embodiments, a VLP as provided herein comprises a modified filovirus GP as provided herein and a filovirus VP40. In certain embodiments, the VLP comprises a filovirus GP and a filovirus nucleoprotein (NP). In certain embodiments, the VLP comprises a filovirus GP and a filovirus VP24. In certain embodiments, the VLP comprises a filovirus GP and a filovirus VP40 and further comprises a filovirus nucleoprotein (NP) and/or a filovirus VP24.

[0140] Certain embodiments provide for an isolated polynucleotide comprising a nucleic acid encoding an immunogen, e.g., an immunogenic polypeptide, or a fragment thereof disclosed anywhere herein, or a subunit thereof. Certain embodiments provide for a vector comprising such a polynucleotide. Certain embodiments provide for a composition comprising such a polynucleotide or vector.

[0141] Polynucleotides encoding polypeptides disclosed herein can be cloned using DNA amplification methods, such as the polymerase chain method (PCR) (see e.g., Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor, N.Y.; Berger & Kimmel (1987) Methods in Enzymology. Vol. 152). Thus, for example, a nucleic acid molecule encoding a filovirus GP can be PCR amplified using a sense primer containing one restriction site and an antisense primer containing another restriction site. This will produce a nucleic acid encoding the desired sequence or subsequence having terminal restriction sites. This nucleic acid can then be ligated into a vector having appropriate corresponding restriction sites. Suitable PCR primers can be chosen by one of ordinary skill in the art based on the sequence to be expressed. Appropriate restriction sites can also be added by ligation or site-directed mutagenesis (see Gillman & Smith Gene 8: 81-97(1979); Roberts et al. Nature 328: 731-4 (1987)).

[0142] Immunogens disclosed herein are typically expressed using an expression vector.

Expression vectors can be either self-replicating extrachromosomal vectors or vectors that integrate into a host genome. Generally, expression vectors include transcriptional and translational regulatory nucleic acid sequences operably linked to the nucleic acid encoding the target protein. The term "control sequences" refers to DNA sequences involved in the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers. One of ordinary skill in the art will recognize that a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example: DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Operably linked DNA sequences can be contiguous or non-contiguous. Methods for linking DNA sequences are well-known in the art and include use of the polymerase chain reaction and ligation. The transcriptional and translational regulatory nucleic acid will generally be appropriate to the host cell used to express the target protein.

[0143] Numerous types of expression vectors, and regulatory sequences are known in the art for a variety of host cells. Methods for expressing polypeptides are also well known (e.g., Sambrook et al. (1989) Molecular Cloning, A Laboratory Manual, 2nd ed., vol. 1-3, Cold Spring Harbor Laboratory; Berger and Kimmel (1987) Guide to Molecular Cloning Techniques, Methods in Enzymology, vol. 152, Academic Press, Inc., San Diego, Calif; Ausubel et al. (1995) Current Protocols in Molecular Biology, John Wiley & Sons, Inc., NY). Certain illustrative, non-limiting, examples of vectors include plasmids pET, pALTER, pCMV, pBlueScript, pcDNA3, pHCMV/Zeo, pCR3.1, pEFl/His, pIND/GS, pRc/HCMV2, pSV40/Zeo2, pTRACER-HCMV, pUB6/V5-His, pVAXl, and pZeoSV2 (available from Invitrogen, San Diego, CA), and plasmid pCI (available from Promega, Madison, WI).

[0144] Transcriptional and translational regulatory sequences can include, but are not limited to, promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences. Promoter sequences can encode either constitutive or inducible promoters. Promoters can be either naturally occurring promoters or hybrid promoters. Hybrid promoters, which combine elements of more than one promoter, are also known in the art.

[0145] An expression vector can comprise additional elements. For example, the expression vector can have two replication systems, thus allowing it to be maintained in two organisms, for example in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification. Furthermore, for integrating expression vectors, the expression vector contains at least one sequence homologous to a sequence in the host cell genome, and preferably two homologous sequences that flank the expression construct. The integrating vector can be directed to a specific locus in the host cell by selecting the appropriate homologous sequence for inclusion in the vector.

[0146] Certain embodiments provide for a host cell comprising a polynucleotide or vector comprising a nucleic acid encoding an immunogen disclosed herein or a fragment thereof. Certain embodiments provide a method of making an immunogen or fragment thereof disclosed anywhere herein, the method comprising culturing a host cell comprising a polynucleotide or vector described herein and recovering and/or isolating the immunogen.

[0147] Immunogens comprising filovirus GP can be produced by culturing a host cell transformed with an expression vector containing a nucleic acid encoding a filovirus GP, under the appropriate conditions to induce or cause expression of the immunogen. The conditions appropriate for protein expression will vary with the choice of the expression vector and the host cell. Also, the coding sequences can be optimized for expression in the selected host cells.

[0148] Host cells include yeast, bacteria, archaebacteria, fungi, insect and animal cells, including mammalian cells such as, but are not limited to, Drosophila melanogaster cells, Saccharomyces cerevisiae and other yeasts, E. coli, Bacillus subtilis, Sf9 cells, CI 29 cells, 293 cells, Neurospora, BHK, CHO, COS, HeLa cells, and Hep G2 cells.

[0149] In some embodiments, the immunogens are expressed in mammalian host cells.

Exemplary mammalian host cell lines include, but are not limited to, 293, CHO (Chinese Hamster Ovary), DG44 and DUXBl 1 (Chinese Hamster Ovary lines, DHFR minus), HELA (human cervical carcinoma), CVI (monkey kidney line), COS (a derivative of CVI with SV40 T antigen), VERY, BHK (baby hamster kidney), MDCK, WI38, R1610 (Chinese hamster fibroblast) BALBC/3T3 (mouse fibroblast), HAK (hamster kidney line), SP2/0 (mouse myeloma), P3x63-Ag3.653 (mouse myeloma), BFA-lclBPT (bovine endothelial cells), RAJI (human lymphocyte) and 293 (human kidney). Host cell lines are typically available from commercial services, the American Tissue Culture Collection or from published literature.

[0150] Promoters from viral genes are frequently used in mammalian expression systems, because the viral genes are often highly expressed and have a broad host range. Examples include the SV40 early promoter, mouse mammary tumor virus LTR promoter, adenovirus major late promoter, herpes simplex virus promoter, and the CMV promoter. Typically, transcription termination and polyadenylation sequences recognized by mammalian cells are regulatory regions located 3' to the translation stop codon and thus, together with the promoter elements, flank the coding sequence. Examples of transcription terminator and polyadenylation signals include those derived from SV40.

[0151] Methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used. Suitable techniques include, but are not limited to, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, viral infection, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.

[0152] In some embodiments, the immunogens are expressed in bacterial systems. Bacterial expression systems are well known in the art. Promoters from bacteriophage can also be used and are known in the art. In addition, synthetic promoters and hybrid promoters are also useful; for example, the tac promoter is a hybrid of the tip and lac promoter sequences. Furthermore, a bacterial promoter can include naturally occurring promoters of nonbacterial origin that have the ability to bind bacterial RNA polymerase and initiate transcription. In addition to a functioning promoter sequence, an efficient ribosome binding site is desirable. The expression vector can also include a signal peptide sequence that provides for secretion of the target protein in bacteria. A polypeptide is either secreted into the growth media (gram-positive bacteria) or into the periplasmic space, located between the inner and outer membrane of the cell (gram-negative bacteria). The expression vector can also include an epitope tag providing for affinity purification of the target protein. The bacterial expression vector can also include a selectable marker gene to allow for the selection of bacterial strains that have been transformed. Suitable selection genes include genes that render the bacteria resistant to drugs such as ampicillin, chloramphenicol, erythromycin, kanamycin, neomycin and tetracycline. Selectable markers also include biosynthetic genes, such as those in the histidine, tryptophan, and leucine biosynthetic pathways. These components are assembled into expression vectors. Expression vectors for bacteria are well known in the art, and include vectors for Bacillus subtilis, E. coli, Streptococcus cremoris, and Streptococcus lividans, among others. The bacterial expression vectors are transformed into bacterial host cells using techniques well known in the art, such as calcium chloride treatment, electroporation, and others. [0153] The immunogens disclosed herein can also be produced in insect cells. Expression vectors for the transformation of insect cells, and in particular, baculovirus-based expression vectors, are well known in the art. The filovirus GP can also be produced in yeast cells. Yeast expression systems are well known in the art, and include expression vectors for Saccharomyces cerevisiae, Candida albicans and C. maltosa, Hansenula polymorpha, Kluyveromyces fragilis and K. lactis, Pichia guillerimondii and P. pastoris, Schizosaccharomyces pombe, and Yarrowia lipolytica.

[0154] The immunogens disclosed herein can be produced in a cell-free expression system in vitro using an expression vector containing nucleic acid encoding the immunogen, under the appropriate conditions to induce or cause expression of the immunogen in vitro. Cell-free in vitro expression systems are well known in the art.

[0155] The immunogens can also be made as fusion proteins, using techniques that are well known in the art. For example, a filovirus GP can be made as a fusion protein to increase expression, to increase serum half-life, or to link it with a tag polypeptide that provides an epitope to which an anti-tag antibody can selectively bind. Exemplary tags or fusion partners include the myc epitope, the immunoglobulin Fc domain, and 6-histidine. The epitope tag is generally placed at the amino- or carboxyl-terminus of the target protein. The presence of such epitope-tagged forms of a target protein can be detected using an antibody against the tag polypeptide. Thus, the epitope tag enables the target proteins to be readily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag.

[0156] The immunogens can be purified or isolated after expression in a host cell. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. The term "purified" denotes that a protein gives rise to essentially one band in an electrophoretic gel. For example, it means that the protein is at least 85% pure, such as at least 95% pure, such as at least 99% pure. The term "isolated polypeptides" also includes polypeptides in situ within recombinant host cells, since at least one component of the polypeptide natural environment will not be present.

[0157] The immunogens can be isolated or purified in a variety of ways known to those of ordinary skill in the art, depending on what other components are present in the sample. Standard purification methods include electrophoretic, molecular, immunological and chromatographic techniques, including ion exchange, hydrophobic, affinity, lectin-affinity and reverse-phase HPLC chromatography, and chromatofocusing. For example, the target protein can be purified using an affinity column. Ultrafiltration and diafiltration techniques, in conjunction with protein concentration, are also useful. Suitable purification techniques are standard in the art (see generally R. Scopes (1982) Protein Purification, Springer- Verlag, N.Y.; Deutcher (1990) Methods in Enzymology vol. 182: Guide to Protein Purification, Academic Press, Inc. N.Y.). The degree of purification will vary depending on the use of the polypeptide. In some instances no purification is necessary.

[0158] In certain embodiments, an immunogen described herein can comprise a heterologous amino acid sequence or one or more other moieties not normally associated with a filovirus polypeptide {e.g., an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, pharmaceutical agent, a lymphokine, a heterologous antibody or fragment thereof, a detectable label, polyethylene glycol (PEG), and a combination of two or more of any said agents). In further embodiments, an immunogen can comprise a detectable label selected from the group consisting of an enzyme, a fluorescent label, a chemiluminescent label, a bioluminescent label, a radioactive label, or a combination of two or more of any said detectable labels.

[0159] Certain embodiments provide for composition comprising an immunogen and methods for preparing them. In certain embodiments, a composition in prepared by suspending and packaging one or more immunogens with a suitable pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include sterile water or a sterile physiological salt solution, e.g., phosphate buffered saline (PBS). Pharmaceutically acceptable carriers also encompass any of the standard pharmaceutical carriers, such as those suitable for parenteral administration, such as by intramuscular, intravenous, intradermal, intraperitoneal, and subcutaneous routes. Examples of such formulations include aqueous and non-aqueous, isotonic sterile injection solutions, which contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.

[0160] Certain embodiments comprise an immunogen and an adjuvant. Adjuvants are used to enhance an immune response to an immunogen. Adjuvants can be obtained from any of a number of sources including from natural sources, recombinant sources, and/or be chemically synthesized, etc. Suitable exemplary adjuvants include, among others, liposomes, alum, monophosphoryl lipid A, immune-stimulating complexes (ISCOMS), LPS analogs including 3-O-deacylated monophosphoryl lipid A (Ribi Immunochem Research, Inc.; Hamilton, Mont.), mineral oil and water, aluminum hydroxide, Amphigen, Avirdine, L121/squalene, muramyl peptides, and saponins, such as Quil A, and any biologically active factor, such as cytokine, an interleukin, a chemokine, a ligands, and optimally combinations thereof. Certain of these biologically active factors can be expressed in vivo, e.g., via a plasmid or viral vector. For example, such an adjuvant can be administered with a priming DNA vaccine encoding an antigen to enhance the antigen-specific immune response compared with the immune response generated upon priming with a DNA vaccine encoding the antigen only.

[0161] Certain embodiments provide for DNA vaccines comprising a nucleic acid molecule encoding one or more immunogens disclosed herein. Thus, some embodiments provide an immunogenic composition comprising a nucleic acid molecule encoding an immunogen and a pharmaceutically acceptable carrier. Methods for preparing and administering a DNA vaccine expressing heterologous polypeptides are known in the art and have been previously described (see, e.g., Doolan & Hoffman, Int. J. Parasitol. 37:753-762, (2001)).

[0162] In certain embodiments, a vaccine composition is a viral vaccine comprising a viral vector encoding one or more immunogens disclosed herein. Exemplary viral vectors for use in the vaccine compositions, but are not limited to, vaccinia viral vectors (such as vectors based on modified vaccinia virus Ankara or avian pox viruses), adenoviral vectors, VSV vectors, herpesvirus vectors, rabies virus vectors, and yellow fever viral vectors {see, e.g., Tao et al, J. Exp. Med. 207:201-209 (2005)). Methods for preparing and administering viral vaccines expressing filovirus proteins are known in the art. A vaccine composition as provided herein can also be a naked DNA vaccine.

[0163] In another aspect, the immunogens and the sequences encoding them can be used to induce an immune response to a filovirus in a subject. Suitably, the immunogens or coding sequences are delivered to the cells in an effective amount and in a manner which presents them favorably for induction of an antibody response, a cellular immune response, or both. Any of the immunogens described herein can be utilized. The effective amount and method of administration of a particular therapeutic or prophylactic treatment can vary based on the individual patient and the stage of the disease, as well as other factors known to those of skill in the art. Therapeutic efficacy and toxicity of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED 50 (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population). The dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD 50 /ED 50 . The data obtained from cell culture assays and animal studies is used in formulating a range of dosages for human use. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.

[0164] In certain embodiments, an immunogen is delivered to the subject in protein form by a non-viral delivery vehicle. For example, liposomes, micelles, gels, multiple antigen complexes can be prepared utilizing the chimeric glycoproteins described herein. A suitable immunogenic amount of the immunogen can be readily determined. Optionally, the immunogen can be conjugated with a polyethylene glycol conjugate according to published techniques. See, e.g., U.S. Pat. No. 6,399,385 and references cited therein.

[0165] In certain embodiments, a viral vector is utilized for delivery of an immunogen to a subject. Any suitable viral system can be used including, e.g., adenoviruses, poxviruses, and the like.

[0166] An immunogen can be administered by any suitable method of administration known in the art including, but not limited to, intradermally, subcutaneously, intramuscularly, intraperitoneally, orally, ocularly (e.g., as an eye spray), and intravenously. Vaccines are conventionally administered parenterally, by injection, for example, either subcutaneously or intramuscularly. Additional formulations that are suitable for other modes of administration include suppositories and, in some cases, oral formulations, or nasal sprays. For suppositories, traditional binders and carriers can include, for example, polyalkalene glycols or triglycerides. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like.

[0167] Certain embodiments provide for detecting anti-filovirus GP antibodies in a sample. In some embodiments, the sample is suspected of containing a filovirus infection. The method includes contacting the sample suspected of containing an antibody which binds an epitope of the immunogen described herein, allowing the antibody to bind to the immunogen to form an immunological complex, detecting the formation of the immunological complex and correlating the presence or absence of the immunological complex with the presence or absence of anti-filovirus antibodies in the sample. The sample can be biological, environmental or a food sample.

[0168] As used herein, "detecting the formation of the immunological complex" is intended to include discovery of the presence or absence of anti-filovirus antibodies in a sample. The presence or absence of anti- filovirus antibodies can be detected using an immunoassay. A number of immunoassays used to detect and/or quantitate antigens are well known to those of ordinary skill in the art. See Harlow and Lane, Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory, New York 1988 555-612). Such immunoassays include antibody capture assays, antigen capture assays, and two-antibody sandwich assays. These assays are commonly used in the art. In an antibody capture assay, the antigen is attached to solid support, and labeled antibody is allowed to bind. After washing, the assay is quantitated by measuring the amount of antibody retained on the solid support. A variation of this assay is a competitive ELISA wherein the antigen is bound to the solid support and two solutions containing antibodies which bind the antigen, for example, serum from an filovirus vaccinee and a known anti-filovirus glycoprotein monoclonal antibody, are allowed to compete for binding of the chimeric protein. The amount of monoclonal bound is then measured, and a determination is made as to whether the serum contains anti-filovirus glycoprotein antibodies. This competitive ELISA can be used to indicate immunity to known protective epitopes in a vaccinee following vaccination.

[0169] In an antigen capture assay, the antibody is attached to a solid support, and labeled antigen is allowed to bind. The unbound proteins are removed by washing, and the assay is quantitated by measuring the amount of antigen that is bound. In a two-antibody sandwich assay, one antibody is bound to a solid support, and the antigen is allowed to bind to this first antibody. The assay is quantitated by measuring the amount of a labeled second antibody that can bind to the antigen.

[0170] These immunoassays typically rely on labeled antigens, antibodies, or secondary reagents for detection. These proteins can be labeled with radioactive compounds, enzymes, biotin, or fluorochromes. Of these, radioactive labeling can be used for almost all types of assays and with most variations. Enzyme-conjugated labels are particularly useful when radioactivity must be avoided or when quick results are needed. Biotin-coupled reagents usually are detected with labeled streptavidin. Streptavidin binds tightly and quickly to biotin and can be labeled with radioisotopes or enzymes. Fluorochromes, although requiring expensive equipment for their use, provide a very sensitive method of detection. Antibodies useful in these assays include monoclonal antibodies, polyclonal antibodies, and affinity purified polyclonal antibodies. The binding of these labels to antibodies or fragments thereof can be accomplished using standard techniques commonly known to those of ordinary skill in the art. Typical techniques are described by Kennedy, J. H., et al, Clin. Chim. Acta 70: 1-31 (1976), and Schurs, A. H. W. M., et al. Clin. C 'him Acta 57: 1-40 (1977). Coupling techniques mentioned in the latter are the glutaraldehyde method, the periodate method, the dimaleimide method, and others, all of which are incorporated by reference herein.

[0171] Provided herein are methods for monitoring the immune status of a subject vaccinated against infection or disease caused by filovirus, comprising (a) contacting a biological sample comprising antibodies from a subject with one or more immunogens disclosed herein under conditions enabling the formation of antigen/antibody complexes between the polypeptides and the antibodies, and (b) detecting the formation of antigen/antibody complexes.

[0172] In the diagnostic and monitoring methods described above, the biological sample can be further contacted with one or several antigenic peptides originating from other filovirus antigens.

[0173] In some embodiments, the diagnostic and screening agents and assays are nucleic acid- based. Exemplary diagnostic and screening agents for use in nucleic acid-based assays include nucleic acid probes complementary to nucleic acid molecules encoding filovirus polypeptides of the disclosure. Nucleic-acid based diagnostic and screening assays are well known in the art. Exemplary diagnostic and screening assays to be used in this aspect of the disclosure are described in Scherf et al., U.S. Pat. No. 6,855,323, herein incorporated by reference.

[0174] In another aspect a kit for detecting filovirus in a biological sample is provided. The kit includes a container holding one or more immunogens and instructions for using them for the purpose of identifying the presence of anti-filovirus antibodies to form an immunological complex and detecting the formation of the immunological complex such that the presence or absence of the immunological complex correlates with presence or absence of filovirus in the sample. Examples of containers include multiwell plates which allow simultaneous detection of filovirus in multiple samples. Examples

Example 1 :

[0175] ELISA analyses were used to measure antibody binding to different isolated domains of EBOV GP (Keck, et al, 2016, J Virol, 90:279 -291; Holtsberg, et al, 2016, J Virol, 90:266 -278), and an alanine-scanning mutagenesis approach to determine the epitopes of several cross-reactive antibodies. Comprehensive high-throughput alanine scanning mutagenesis was carried out on an expression construct for EBOV GP (EBOV strain Mayinga-76; UniProt accession number Q05320) (Davidson et al., 2015, J Virol, 89(21): 10982-9). Residues 33-676 of full-length EBOV GP were mutagenized to alanine (with alanine residues changed to serine) to create a library of clones, each representing an individual point mutant. GP residues 1-32, which constitute the GP signal peptide, were not mutagenized. The resulting EBOV GP alanine-scan library covered 99.5% of target residues (641 of 644).

[0176] The immunoreactivity of each mAb with expressed wild-type EBOV GP was first optimized by determining reactivity with fixed or unfixed cells over a range of MAb concentrations to identify optimal signal-to-background ratios (>5: 1) and to ensure that signals were within the linear range of detection. The EBOV GP mutation library, arrayed in 384-well microplates, was transfected into HEK-293T cells and allowed to express for 22 hours. Cells were fixed in 4% paraformaldehyde in PBS plus calcium and magnesium, or left unfixed, and were then incubated with an anti-EBOV mAb diluted in 10% normal goat serum (NGS) (Sigma-Aldrich, St. Louis, MO). The cells were incubated with anti-EBOV antibody for 1 hour at room temperature, followed by a 30 minute incubation with Alexa Fluor 488-conjugated secondary antibody (Jackson ImmunoResearch Laboratories, Westgrove, PA) in 10% NGS. Cells were washed twice with PBS without calcium or magnesium and resuspended in Cellstripper (Cellgro, Manassas, VA) plus 0.1% BSA (Sigma-Aldrich, St. Louis, MO). Cellular fluorescence was detected using the Intellicyt high throughput flow cytometer (Intellicyt, Albuquerque, NM). Background fluorescence was determined by fluorescence measurement of vector-transfected control cells. MAb reactivities against each mutant EBOV GP clone were calculated relative to wild-type EBOV GP reactivity by subtracting the signal from mock-transfected controls and normalizing to the signal from wild-type GP -transfected controls. [0177] Mutated residues within critical clones were identified as critical to the mAb epitope if they did not support reactivity of the test mAb but did support reactivity of other control EBOV mAbs. This counter-screen strategy facilitates the exclusion of GP mutants that are locally rnisfolded or that have an expression defect. The detailed algorithms used to interpret shotgun mutagenesis data are described elsewhere (Davidson and Doranz, 2014, which is incorporated herein in its entirety).

[0178] Using this approach, residues critical for the GP binding of pan-ebolavirus mAbs FVM04, FVM09, FVM17, FVM20, m8C4, 4B8, and CA45 were determined.

[0179] A vesicular stomatitis virus (VSV) pseudotyped with EBOV GPwt (wild-type) or a mutant of EBOV GP in which residues F290, W291, and E292 are mutated to alanine (VSV-EBOV GP-AAA) was created. The generation of the pseudotyped viruses was performed as described previously (Whitt MA, 2010, J Virol Methods 169, 365-374). The pseudotyped virus expressed luciferase upon infection of the cells (Whitt MA, 2010, J Virol Methods 169, 365-374). Vero cells were infected with VSV-EBOV GPwt or VSV-EBOV GP-AAA and neutralization of the two viruses by different concentrations of m8C4 was determined. As shown in Figure 6, while m8C4 poorly neutralized the VSV pseudotype expressing wild-type EBOV GP, the pseudotype with the EBOV GP-AAA was potently neutralized by m8C4. The neutralizing activity of m8C4 was over 300 fold higher against VSV-EBOV GP-AAA as compared with VSV-EBOV GPwt (EC50 of 0.035 μg/ml for mutant vs. >10 μ^να\ for wt (Figure 6). These data further indicate that mutating these residues within an EBOV GP vaccine could significantly enhance the neutralizing response to EBOV and likely other ebolavirus species as the epitope of m8C4 is conserved among ebolaviruses.

[0180] A mutation at positions E292 to any other amino acid (to be determined empirically) can be incorporated in a GP -based Ebola vaccine. Such mutant is expected to enhance the generation of FVM09 and m8C4 type antibodies by the immune system. Such mutation can be further combined with specific mutations in the base of the GP or glycosylation silencing mutants of the glycan cap described above.

[0181] Any combination of E292 mutation with F290 or W291 mutation can (double and triple mutants) incorporated into a vaccine. These mutations are expected to primarily enhance neutralizing responses to the m8C4 epitope. Such mutations can be further combined with specific mutations in the base of the GP or glycosylation silencing mutants of the glycan cap described above. [0182] It has been determined that specific mutations in the base of the GP trimer can have a global impact on the exposure of cross-reactive and cross-neutralizing epitopes in the GP head domain. Previously described EBOV neutralizing antibodies KZ52, 2G4, and 4G7 (Lee, et al., 2008, Nature, 454 (7201): 177-182; Murin, et al, 2014, Proc Natl Acad Sci USA, 111(48): 17182-7) as well as SUDV-specific neutralizing mAb 16F6 (Dias, et al, 2011, Nat Struct Mol Bio, 18(12): 1424-7) bind at the base of the GP trimer to overlapping epitopes consisting of residues from both GP2 and the base domain of GP1 (As shown in Figure 1 for KZ52). In contrast, the cross-reactive and cross-neutralizing pan-ebolavirus mAbs (Keck, et al, 2016, J Virol, 90:279 -291; Holtsberg, et al, 2016, J Virol, 90:266 -278) (PCT/US 15/57627 and PCT/US 15/37493, both of which are incorporated herein in their entireties) appear to bind from above to either the glycan cap or the GP1 core (GP head domain) (Tables 1A, IB, and 1C).

[0183] Tables 1A, IB, and 1C summarize the characterization and efficacy of pan-ebolavirus antibodies. ELISA reactivity and neutralization capacity is shown. The binding regions were determined by ELISA using various domains of EBOV GP as antigen (Keck, et al., 2016, J Virol, 90:279 -291; Holtsberg, et al, 2016, J Virol, 90:266 -278). Binding residues were determined using the alanine scanning mutagenesis as described herein. Efficacy was tested in mice as described (Keck, et al, 2016, J Virol, 90:279 -291; Holtsberg, et al, 2016, J Virol, 90:266 -278). For EBOV guinea pig studies animals were infected with guinea pig adapted EBOV (GPA-EBOV) (Cross et al, 2015, J Infect Dis. 212 Suppl 2:S305-15) followed by injection of FVM04 (5 mg/animal) either on day 1 and survival monitored for 28 days. This study showed 33% protection. To test efficacy in guinea pigs against SUDV, animals were infected with guinea pig adapted Sudan virus (GPA-SUDV) (Wong et al, 2015, J Virol, 90(l):392-9). In one group FVM04 was injected at one day post infection and in another group animals were given FV04 at 3 days post infection. 100% protection was observed in both studies. CA45 testing in both EBOV and SUDV guinea pig models was performed by injecting one dose of antibody at 3 days post challenge at a dose of 5 mg. NT: not tested.

Table 1A.

ELISA reactivity Neutralization

Table IB.

Table 1C.

Efficacy

Guinea Guinea

Mouse Mouse Pig Pig mAb EBOV Sudan EBOV SUDV

FVM04 100% 80% 33% 100%

FVM09 70% NT NT NT

FVM17 NT NT NT NT

FVM20 60% NT NT NT

4B8 100% NT NT NT m8C4 50% 70% NT NT CA45 100% 90% 50% 100% [0184] Alanine scanning experiments revealed differences between GP binding patterns. While the binding of the GP base binder KZ52 to all individual mutants remained below 150% of binding value with wild-type (wt) GP (Figure 2A), several single alanine substitutions of GP had an enhancing effect (as high as 200-300%) of the binding to wt GP) on the binding of cross-neutralizing antibody FVM04 (Figure 2B). Out of 217 amino acids forming the base, alanine mutation of 23 residues increased FVM04 binding to GP by more than two fold (Figure 2B and Table 2).

[0185] Table 2 shows the relative binding of individual single point alanine mutations of the residues within the base of EBOV GP to KZ52 (base binder) as compared to GP1 top binders m8C4, FVM09, FVM20, FVM17, and 4B8, as well as the fusion loop binder CA45. The binding values are shown for each mutant and each antibody, given as a percentage relative to the binding of each antibody to wild-type (wt) GP. Where the binding is elevated to 150-200%) compared to wt GP the values are shown in bold font. For enhancements of more than 200%> of wild-type the values are shown in bold italicized font on black background. Binding values representing mutations with reduction in binding of KZ52 to 20%) or less compared to wt GP are double underlined and bold.

Table 2.

FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

I33A 68 208 94 168 168 143 302 129

P34A 84 231 150 188 182 139 323 116

L35A 121 123 105 153 140 102 149 109

G36A 86 146 103 118 138 130 93 71

V37A 86 125 115 147 109 125 174 91

138 A 70 251 160 195 186 133 277 118

H39A 100 149 144 108 126 117 133 131

N40A 148 153 132 125 148 93 123 178

S41A 96 121 71 143 119 81 104 122

T42A 127 138 120 139 118 107 103 153

L43A 71 172 152 167 196 151 405 54

Q44A 83 118 66 130 127 85 110 88

V45A 88 102 95 149 117 100 253 147

S46A 95 104 77 84 111 79 85 66

D47A 98 106 92 92 76 83 99 64

V48A 93 143 168 173 229 128 420 74 FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

D49A 115 114 111 144 114 98 49 69

K50A 103 134 138 128 166 109 97 91

L51A 98 201 184 172 218 161 480 88

V52A 105 169 163 201 192 175 366 137

C53A 50 256 219 273 251 224 578 122

R54A 108 153 122 150 166 111 309 93

D55A 97 118 109 136 124 123 78 83

K56A 80 132 86 132 132 117 86 114

L57A 95 163 129 135 134 159 254 59

S58A 111 138 136 97 131 100 125 88

S59A 116 105 129 159 81 94 97 99

T60A 81 120 98 138 120 93 64 100

N61A 118 130 107 125 118 123 127 103

Q62A 79 135 70 165 106 105 115 88

L63A 120 162 140 194 173 179 484 112

R64A 75 126 95 113 104 79 19 2

S65A 96 138 116 125 160 114 132 93

V66A 99 159 73 159 78 114 354 98

L68A 83 213 155 187 177 155 902 60

N69A 106 141 175 198 140 161 148 108

K95A 92 159 173 148 154 92 173 106

V96A 116 109 96 92 82 79 96 98

V97A 92 115 108 145 143 128 108 92

N98A 117 130 98 149 110 110 98 111

Y99A 43 169 139 164 217 168 139 39

El 00 A 116 135 98 129 129 105 98 123

AIOIS 96 118 110 162 139 114 110 117

G102A 52 148 183 178 189 166 183 76

E103A 129 124 158 204 228 124 1

W104A 84 124 85 102 122 94 85 72

F159A 4 249 224 189 235 203 762 8

F160A 61 108 82 143 141 150 111 65

L161A 92 153 101 163 159 138 512 75

Y162A 58 40 68 117 132 112 26 49

D163A 124 110 104 148 131 122 82 114

R164A 97 193 134 232 239 223 610 73

L165A 103 158 136 182 146 126 247 128

A166S 87 104 139 196 172 157 135 95 FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

SI 67 A 92 127 96 118 108 92 45 85

T168A 100 103 165 130 177 134 148 118

A177S 96 110 122 125 135 104 26 90

E178A 66 54 111 127 119 109 9

G179A 65 193 114 229 203 176 822 56

V180A 95 133 72 122 106 79 46 115

V181A 86 230 124 228 314 271 916 64

A182S 110 95 219 174 143 109 343 92

F183A JJ 306 273 275 282 264 1212 23

LI 84 A 111 195 213 184 217 241 546 89

I185A 113 231 264 282 194 141 643 142

L186A 109 107 165 160 122 122 189 60

PI 87 A 129 156 123 168 121 137 325 60

Q188A 99 156 119 173 118 88 93 121

A189S 92 143 113 148 88 112 81 90

K190A 108 151 119 116 115 132 187 33

K191A 114 117 96 198 127 103 130 110

D192A 122 134 130 161 85 90 95 170

F193A 118 129 96 148 110 110 121 182

F194A 96 167 173 156 163 85 178 224

S195A 105 106 78 109 105 69 89 113

R498A 65 213 121 141 193 177 295 56

T499A 106 153 142 135 110 99 100 94

R500A 77 178 140 80 116 111 114 65

R501A 61 224 150 129 140 131 302 51

E502A 99 145 129 117 83 92 68 92

A503S 97 150 99 88 118 95 111 82

I504A 73 99 179 142 113 138 94 95

V505A 76 108 128 159 131 92 132 103

N506A 93 142 299 165 149 136 89 105

A507S 89 144 122 137 116 86 75 141

Q508A 36 141 427 216 204 144 177 83

P509A 85 118 210 142 155 162 145 89

K510A 89 174 192 173 250 185 146 79

C511A 1 174 227 179 234 181 891 7

N512A 35 279 343 247 319 240 876 43

P513A 82 175 226 242 234 208 599 84

N514A 74 144 219 153 166 164 245 81 FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

L515A 241 254 196 274 230 799 4

H516A 89 203 257 185 275 189 716 68

Y517A 70 188 221 176 226 163 659 4

W518A 142 150 129 138 133 378 6

T519A 104 100 127 117 107 120 73 140

T520A 107 93 90 110 76 110 89 77

Q521A 115 99 163 89 103 88 60 128

D522A 64 105 129 96 119 94 76 126

E523A 93 139 170 145 125 118 48 122

G524A 93 144 159 156 106 115 62 93

A525S 85 91 212 101 111 70 56 95

A526S 94 111 104 128 114 87 43 90

I527A 98 142 130 137 114 109 62 107

G528A 109 123 169 116 76 122 131 93

L529A 98 126 125 132 128 111 51 94

A530S 95 140 136 131 92 115 43 103

W531A 87 214 218 143 128 118 66 144

I532A 100 135 103 123 106 106 20 95

P533A 97 180 218 160 97 143 225 117

Y534A 72 120 107 117 94 95 23 140

F535A 82 112 171 112 71 95 33 107

G536A 112 119 144 129 141 99 43 82

P537A 106 137 167 142 122 138 127 84

A538S 114 122 108 105 87 94 52 107

A539S 78 95 138 87 115 74 38 77

E540A 91 111 146 116 95 76 83 99

G541A 30 42 62 37 45 50 1 44

I542A 72 114 132 116 95 89 24 98

Y543A 76 142 165 167 162 135 183 82

I544A 73 106 122 121 118 97 29 84

E545A 92 175 294 152 199 163 300 39

G546A 71 189 240 224 215 180 636 11

L547A 80 106 155 131 134 126 53 88

M548A 116 124 102 146 121 117 65 61

H549A 78 130 127 130 80 118 105 73

N550A 167 85 144 123 116 131 22

Q551A 100 91 103 131 84 86 42 114

D552A 12 115 153 142 101 149 76 119 FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

G553A Ik 137 160 173 143 131 148 71

L554A 88 157 99 137 102 87 1 12 134

C556A 174 231 171 146 143 858 6

G557A 124 131 81 130 1 10 91 122 103

L558A 77 188 142 158 152 175 395 84

R559A 2 190 273 175 232 181 834 7

Q560A 72 145 111 211 137 162 186 79

L561A 40 278 196 178 224 140 139 1 13

A562S 30 273 323 206 291 231 950 33

N563A 96 139 132 193 138 130 186 72

E564A 93 145 61 145 142 105 200 102

T565A 26 214 202 189 258 200 720 79

T566A 133 156 175 186 137 121 546 131

Q567A 99 122 130 156 156 93 39 100

A568S 107 1 12 143 150 123 88 44 101

L569A 59 252 209 246 277 217 574 61

Q570A 101 173 165 168 1 10 161 269 109

L571A 83 134 171 134 147 123 64 88

F572A 102 121 141 144 120 1 17 54 90

L573A 86 251 251 239 205 298 793 126

R574A 87 131 165 1 11 1 11 96 58 86

A575S 96 1 1 1 159 1 13 1 13 108 27 70

T576A 121 159 161 129 133 1 15 137 90

T577A 104 141 135 166 106 106 107 84

E578A 1 10 1 19 130 107 104 1 15 1 16 80

L579A 97 148 91 1 17 89 106 52 53

R580A 101 167 97 120 123 101 34 86

T581A 93 135 130 124 141 101 197 135

F582A 98 121 121 1 12 1 15 137 56 105

S583A 126 129 203 131 145 98 206 1 16

I584A 107 153 144 144 148 129 102 121

L585A 81 155 152 131 108 98 69 90

N586A 108 134 172 127 109 109 93 100

R587A 97 124 109 103 102 97 50 85

K588A 105 167 140 125 106 122 74 75

A589S 96 157 145 106 73 121 39 1 15

I590A 88 122 1 13 1 15 1 18 98 30 90

D591A 96 123 120 1 14 89 1 12 142 135 FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

F592A 108 126 113 115 120 62 66 77

L593A 100 107 127 96 96 103 67 80

L594A 88 187 156 127 115 107 68 72

Q595A 92 117 100 105 129 96 123 151

R596A 106 97 157 149 93 101 125 102

W597A 97 118 170 134 116 91 40 79

G598A 72 111 146 144 135 144 137 95

G599A 86 136 170 132 137 136 60 98

T600A 99 163 155 143 148 109 88 98

C601A 78 210 269 174 268 127 669 105

H602A 106 130 127 127 116 82 105 94

1603 A 102 128 224 137 130 147 167 97

L604A 94 135 194 133 131 132 204 124

G605A 88 131 157 150 152 163 226 104

P606A 110 114 132 146 140 108 197 78

D607A 95 107 77 131 129 79 114 90

C608A 43 181 155 174 127 137 741 56

C609A 22 205 144 149 178 191 868 27

I610A 83 183 200 137 181 168 588 74

E611A 87 123 132 93 136 107 186 98

P612A 89 137 147 98 128 70 147 95

H613A 103 178 154 146 98 176 41 89

D614A 89 142 141 159 158 138 361 147

W615A 85 135 143 161 151 132 162 102

T616A 126 136 115 105 123 151 278 105

K617A 97 132 109 105 107 101 90 85

N618A 101 125 147 112 118 131 141 132

I619A 108 111 91 109 92 82 50 123

T620A 117 139 180 176 197 97 151 152

D621A 116 126 114 117 121 77 87 99

K622A 99 89 139 129 127 83 119 125

1623 A 93 99 226 133 124 112 21 106

D624A 104 127 124 145 154 128 194 90

Q625A 90 117 128 95 85 111 64 103

I626A 83 150 179 104 101 83 29 124

I627A 98 134 121 130 116 113 50 122

H628A 84 143 141 142 66 115 74 94

D629A 102 129 143 100 57 96 94 4 FVM04 m8C4 FVM09 FVM17 FVM20 h4B8 CA45

Mutation KZ52

Binding Binding Biding Binding Binding Binding Binding

F630A 76 121 151 131 110 90 18 105

V631A 96 122 123 116 115 94 55 105

D632A 104 131 98 138 102 160 187 97

K633A 98 138 166 143 150 141 146 94

T634A 82 168 114 125 150 118 100 89

L635A 87 101 116 92 101 87 87 123

P636A 86 130 127 114 130 133 95 73

D637A 88 100 94 124 81 111 73 104

Q638A 113 132 141 157 125 145 65 84

G639A 86 115 151 124 123 107 91 97

D640A 105 113 151 116 148 102 101 82

N641A 84 104 129 121 109 93 85 78

D642A 101 106 121 94 99 113 86 99

N643A 106 106 121 143 117 115 91 83

W644A 107 105 166 129 86 88 77 114

W645A 99 130 177 95 118 119 68 126

T646A 102 151 126 143 130 122 86 107

G647A 92 137 132 149 97 110 77 84

W648A 66 107 224 116 98 73 61 95

R649A 85 112 161 139 90 96 66 101

Q650A 120 136 168 117 109 103 104 110

[0186] These mutants included both C53 and C609 that form a disulfide bond between GP1 and GP2. The majority of these residues are hydrophobic, highly networked, and not surface exposed (Table 3). In addition to the 23 base residues, mutation of R498 and R501, within the furin cleavage site separating GP1 and GP2, also led to an over two fold increase in FVM04 binding (Figure 2B and Table 3).

[0187] Table 3: Single alanine substitution within the EBOV GP base that increase FVM04 binding by more than 200% as compared to wild type EBOV GP. Percent binding is also shown for the same mutants to m8C4, FVM09, FVM20, and 4B8. All values greater than 200%) are double underlined. Values are average of 2 or 3 experiments ± standard error defined as half the difference between maxim and minimum for each replicate.

Table 3

Mutant Position in GP structure % binding compared to wt GP

FVM04 m8C4 FVM09 FVM20 4B8

I33A Buried 208 ±16 94 ±30 168 ±10 143 ±15 302 ±5 Mutant Position in GP structure % binding compared to wt GP

P34A Buried 231 ±40 150 ±40 188 ±68 139 ±12 323 ±20

I38A Buried 251 ±45 160 ±21 195 ±4 133 ±1 277 ±18

L51A Surface exposed 201 ±8 184 ±46 172 ±6 161 ±22 480 ±46

C53A Buried; Disulfide bond with 256 ±18 219 ±9 273 ±14 224 ±33 578 ±30

C609

L68A Buried 213 ±28 155 ±30 187 ±12 155 ±5 902 ±5

E71A Surface exposed 238 ±7. 245 ±18 264 ±5 269 ±7 780 ±73

F159A Buried 249 ±11 224 ±2 189 ±15 203 ±4 762 ±28

V181A Buried 230 ±5 124 ±35 228 ±7 271 ±14 916 ±40

F183A Buried 306 ±45 273 ±3 275 ±18 264 ±7.0 1212 ±80

I185A Buried 231 ±15 264 ±0 282 ±2 141 ±5 643 ±237

R498A Furin cleavage site 213 ±39 121 ±0 141 ±3 177 ±27 295 ±74

R501A Furin cleavage site 224 ±12 150 ±25 129 ±10 131 ±35 302 ±53

N512A Surface exposed 279 ±9 343 ±65 247 ±19 240 ±32 876 ±208

L515A Buried 241 ±27 254 ±9 196 ±15 230 ±3 799 ±39

H516A Surface exposed 203 ±11 257 ±13 185 ±2 189 ±54 716 ±9

W531A Surface exposed 214 ±0 218 ±28 143 ±4 118 ±7 66 ±8

L561A Buried 278 ±21 196 ±2 178 ±23 140 ±43 139 ±26

A562S Buried 273 ±23 323 ±22 206 ±22 231 ±9 950 ±132

T565A Buried 214 ±54 202 ±49 189 ±20 200 ±25 720 ±235

L569A Buried 252 ±19 209 ±34 246 ±20 217 ±37 574 ±10

L573A Buried 251 ±17 251 ±1 239 ±17 298 ±11 793 ±195

C601A Not resolved in crystal 210 ±13 269 ±6 174 ±0 127 ±9 669 ±194 structure

C609A Not resolved in crystal 205 ±27 144 ±1 149 ±2 191 ±5 868 ±127 structure; Disulfide bond with

C53

[0188] These data demonstrate that minor structural modifications within the GP base can have a significant impact on the accessibility of the FVM04 epitope and possibly other cross- reactive and neutralizing epitopes within the conserved regions of the GP1 head domain. They indicate that the exposure of cross-reactive epitopes can be modulated by mutations in the GP base and could have major implications for development of pan-ebolavirus vaccines.

[0189] In addition, many of these single residue alanine substitutions that increased binding to FVM04 also increased binding of GP to other antibodies that bind to the top of GP trimer, which have reported previously (Keck, et al., 2016, J Virol, 90:279 -291 ; Holtsberg, et al., 2016, J Virol, 90:266 -278). As shown in Table 2, a number of mutations were identified that increase the binding of most or all of these top binders. These data indicate that certain GP base mutations have a global effect on the exposure of cross-reactive epitopes. [0190] Another pan-ebolavirus antibody, CA45, was discovered to bind to a phylogenetically conserved epitope within the internal fusion loop of ebolaviruses (U.S. Provisional Patent No. 62/406,598). Using alanine scanning mutagenesis the specific contact sites for CA45 in EBOV GP were shown to be R64 in the N-terminal base domain of GP1 and Y517, G546, and N550 in GP2 (Table 1). The alanine scanning mutagenesis data for CA45 also identified several GP residues that enhance the exposure and binding of CA45 (Figure 2G). These residues are shown in Table 2.

Example 3 :

[0191] It was observed that substitutions in three out four glycosylation sites within the glycan cap increased the binding of the cross-reactive antibody FVM04. Shotgun mutagenesis epitope mapping identified EBOV GP residues K115, D117, and G118 as critical for FVM04 binding (Figure 3). Alanine substitutions at these residues reduced FVM04 binding to 29%, 1%, and 2% of wild-type respectively, suggesting that these residues constitute contact sites for FVM04, with Dl 17 and Gl 18 having the greatest energetic contribution to FVM04 binding (Figures 3A and 3B). In contrast, binding of two other pan-ebolavirus antibodies FVM02 and FVM09 (Keck, et al, 2016, J Virol, 90:279 -291) were not affected by these mutations (Figure 3B). The putative epitope of FVM04 is positioned in a previously described region with a crest and trough morphology (Hashiguchi, et al., 2015, Cell, 160, 904-912; Bornholdt et al, 2016, MBio, In Press; Wang et al, 2016, Cell, 164(1- 2):258-68) within the receptor binding site and constitutes the tip of the hydrophilic crest, which interacts with a loop from the endosomal filovirus receptor NPCl (31). The three putative FVM04 contact residues of GP are 100% conserved across all ebolaviruses (Figure 3C).

[0192] Individual alanine mutation of 3 out of four glycosylation sites (N238, N257, N268) on EBOV glycan cap led to an increase in FVM04 binding (Figure 4). When the threonine residues two amino acids C-terminal to these glycosylation sites were mutated, the same enhancing effect was observed. These threonines are part of the recognition site for N- glycosylation, further suggesting that the observed effect is due to loss of glycosylation rather than other conformational effects. Thus, the glycosylation sites in the glycan cap of a filovirus GP can be mutated individually or in combination to increase exposure of FVM04 site. Such mutants are expected to induce other cross-neutralizing antibodies. Tar residues include: N238, T240, N257, T259, N268, and T270.

Example: 4

[0193] It was observed that several cross-neutralizing GP antibodies bind better to GPC L that to full length GP. In ELISA, CA45 bound to GP CL with an EC50 of 0.06 nM compared to an EC50 of 1.4 nM for binding to full length GP ectodomain (GPATM), representing a 24-fold enhanced binding to GPC L (Table 4). Biolayer interferometry showed that CA45, while binding to GPC L GPATM with the same rate, exhibits over 1000 fold slower dissociation rate. As a result CA45 binds GPC L with a dissociation constant (KD) of <6 pM while CA45 KD for GPATM is 10.6 nM (Table 4) representing >1700 fold increased binding affinity.

[0194] Table 4: Binding kinetics, determined by Biolayer Interferometry, and ELISA binding EC50 values for binding of CA45 to full length G ectodomain (GPATM) compared to binding to thermolysin-cleaved GP (GPC L )- K on : association rate, K 0ff : dissociation rate, K D : dissocitation constant. EC50: concentration of the antibody that leads to 50% maximal binding to GP.

Table 4

Example 5: Mutations in the base of GP that increase binding of antibodies such as

CA45 to the fusion loop region of GP

[0195] To test if base mutations widen the breadth of antibody response, several base mutations identified in Table 2 were incorporated into a VSV virus in which the VSV G protein was replaced by EBOV GPAmuc protein produced mutant viruses. VSV-GPAmuc and the mutants were used to immunize mice at 100 pfu by i.p. injection. Immune sera were collected after 28 days and tested for reactivity to GP from EBOV, SUDV, BDBV, and RESTV. As shown in Figure 8, the base mutants W531A and L561A induced significantly elevated heterologous response to SUDV, BDBV, and RESTV (as much as 30 fold higher than GPdeltamuc alone). P34A also increased the response to BDBV and RESTV but dampened the anti-SUDV titers. In contrast P573A mutation did not impact the breadth of the antibody response. A mutant in which three of the GC glycans were mutated showed a 10-fold higher heterologous response to SUDV and BDBV (Figure 8). Taken together, these data support the hypothesis that mutating specific residues within the trimer base can enhance the magnitude of the heterologous response.

Example 6: Exemplary Anti-Filovirus Glycoprotein Antibodies [0196] This Example lists examples of anti-filovirus glycoprotein antibodies.

Antibodies binding to the receptor binding site (RBS) of ebolavirus:

[0197] FVM04: FVM04 is a macaque-human chimeric monoclonal antibody that binds to an exposed epitope within the RBS of multiple ebolaviruses including the virulent strains Ebola virus (EBOV), Sudan virus (SUDV), and Bundibugyo virus (BDBV). See PCT Application No. PCT/US15/57627. In order to define the epitope recognized by FVM04 we employed a comprehensive alanine scanning approach, where FVM04 binding was evaluated against a 'shotgun mutagenesis' mutation library of EBOV GP with 641 of 644 target residues individually mutated. Human HEK-293T cells were transfected with the entire mutation library in a 384-well array format (one clone per well) and assessed for reactivity to FVM04 using high-throughput flow cytometry. The method for shotgun mutagenesis is described in patent application 61/938,894 and (Davidson, E., and Doranz, B.J., 2014, Immunology, 143, 13-20). The shotgun mutagenesis revealed that FVM04 contact sites included Kl 15, Dl 17, and Gl 18. This is located within a hydrophilic region of the RBS known as the Crest ((Hashiguchi, et al, 2015, Cell, 160, 904-912; Wang et al, 2016, Cell, 164(l-2):258-68). This epitope is conserved across all ebolaviruses. FVM04 is also weakly reactive to marburgvirus glycoprotein (PCT/US15/57627; Keck, et al., 2015, J Virol, 90:279 -291 ("Keck et al.")).

[0198] FVM04 neutralizes EBOV and SUDV (PCT/US 15/57627; Keck et al.), as well as BDBV and protects mice and guinea pigs against lethal EBOV and SUDV infection (see below).

Antibodies that bind to the glycan cap:

[0199] 13C6FR1 : Monoclonal antibody 13C6 was developed using mouse hybridoma technology and was shown to protect mice from lethal challenge with Ebola virus (Wilson et al, 2000, Science, 287(5458): 1664-6). 13C6 binds on the top of EBOV GP glycan cap (Murin, et al, 2014, Proc Natl Acad Sci USA, 111(48): 17182-7) with contact residues being T270 and K272 (Davidson et al, 2015, J Virol., 89(21): 10982-9). 13C6FR1, a variant of 13C6 (U.S. Patent No. 7,335,356), is a component of the ZMAPP™ therapeutic cocktail (Qiu, et al, 2014, Nature, 514(7520):47-53). Effective neutralization of EBOV by 13C6 requires the presence of complement (Wilson et al, 2000, Science, 287(5458): 1664-6).

[0200] FVM09: FVM09 binds with high affinity to a linear epitope within the disordered loop connecting the β strands 17 and 18 in the glycan cap region of EBOV GP (PCT/US15/57627; Keck et al). Using overlapping peptide mapping we mapped the epitope for FVM09 to amino acids 286-290 (GEWAF) of EBOV GP, and this epitope is 100% conserved among all ebolaviruses (PCT/US 15/57627; Keck et al ).

[0201] FVM09 alone does not neutralize or provide protection in vivo against EBOV, but in combination with several other antibodies it enhances their neutralizing and protective potency as described in (Keck et al) and below.

[0202] ADI-15731: The ADI-15731 mAb was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt, et al, 2016, Science, pii:aad5788. [Epub ahead of print] "Bornholdt et al "). Negative stain electron microscopy reconstructions of the ADI- 15731 fab bound to EBOV GP showed that ADI-15731 bound directly to the GP1 glycan cap structure in a manner reflective of 13C6 (Murin, et al, 2014, Proc Natl Acad Sci USA, 111(48): 17182-7 and Bornholdt et al). However unlike 13C6, ADI-15731 binds to EBOV GP, BDBV GP and SUDV GP. ADI-15731 only effectively neutralizes vesicular stomatitis virus (VSV) pseudovirions displaying either EBOV GP or BDBV GP (as determined by the rVSV GP-GFP assay described in Example 1).

Antibodies that simultaneously bind to glycan cap and core of GP1

[0203] m8C4: Mouse monoclonal antibody m8C4 cross neutralizes EBOV and SUDV and provides partial protection against both viruses in mice (PCT Publication No. WO2015/200522; Holtsberg, et al, 2015, J Virol, 90:266 -278). Efficacy of m8C4 was enhanced when used in combination with FVM09 (PCT Publication No. WO2015/200522; PCT/US 15/57627; Keck et al). In order to define the epitope recognized by m8C4 we employed a comprehensive alanine scanning approach, where m8C4 binding was evaluated against a 'shotgun mutagenesis' mutation library of EBOV GP with 641 of 644 target residues individually mutated. Human HEK-293T cells were transfected with the entire mutation library in a 384-well array format (one clone per well) and assessed for reactivity to m8C4 using high-throughput flow cytometry. The method for shotgun mutagenesis is described in patent application 61/938,894 and (Davidson, E., and Doranz, B.J., 2014, Immunology, 143, 13-20). Shotgun mutagenesis epitope mapping identified EBOV GP residues R136, Q251, and F252 as critical for m8C4 binding. Of these residues, Q251 and F252 are located within the glycan cap, while R136 is located within the core GP1 head domain (Figure 2). m8C4 bridges the core GP1 region with the glycan cap, and the epitope is conserved across ebolaviruses.

[0204] ADI-15750: ADI-15750 was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-15750 can compete for binding with 13C6 and binds with high affinity to a quaternary epitope in the EBOV GP1 structure also present on EBOV soluble GP (sGP). ADI-15750 demonstrated neutralization activity against VSV pseudoviruses displaying either EBOV GP or SUDV GP with IC50 values of 8.80 nM and 32.30 nM, respectively (as determined by the rVSV GP-GFP assay described in Example 1).

[0205] ADI-15968: ADI-15968 was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-15968 can compete for binding with 13C6 and binds with high affinity to a quaternary epitope in the EBOV GP1 structure also present on EBOV sGP. ADI-15968 demonstrated neutralization activity against VSV pseudoviruses displaying either EBOV GP or SUDV GP with IC50 values of 11.74 nM and 47.30 nM respectively (as determined by the rVSV GP-GFP assay described in Example 1).

Antibodies that bind to GP1/GP2 base epitope (Base binders)

[0206] 2G4 and 4G7: The most studied EBOV neutralizing epitope is in a region at the base of the trimeric GP that involves contact sites within GP1 and GP2. Antibodies such as KZ52 (Maruyama, et al, J. Virol. 1999;73 :6024-6030), as well as two of ZMAPP™ components 2G4 and 4G7 bind to this region (Lee, et al, 2008, Nature, 454 (7201): 177-182; Murin, et al, 2014, Proc Natl Acad Sci USA, 111(48): 17182-7). The epitopes for 2G4 and 4G7 are largely overlapping Davidson et al, 2015, J Virol., 89(21): 10982-9) but the angle of binding for these two antibodies is different.

[0207] 2G4 and 4G7 were shown to provide significant protection in mice and guinea pig models of EBOV infection (Qiu, et al, 2012, PLoS Negl Trop Dis, 6: 1575). Both of these antibodies, along with 13C6FR1, are components of the ZMAPP™ antibody cocktail (Qiu, et al, 2014, Nature, 514(7520):47-53). 2G4 and 4G7 are specific to EBOV and do not cross react with other filovirus glycoproteins.

[0208] ADI-15734: ADI-15734 was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-15734 binds specifically to the EBOV GP and directly competes with KZ52. ADI-15734 neutralizes EBOV (as determined by the rVSV GP-GFP assay described in Example 1) and provides significant levels of protection in the EBOV murine model.

Antibodies that bind the fusion loop

[0209] FVM02 (also called FVM02p): FVM02 is a macaque-derived panfilovirus antibody that binds to the tip of the internal fusion loop (IFL) of all ebolaviruses and marburgvirus (IBT PCT/US 15/57627; Keck et al ). FVM02 provides partial protection against EBOV and MARV and potentiates the efficacy of FVM09 against EBOV in mouse models (IBT PCT/US 15/57627; Keck et al).

[0210] CA45: CA45 is a macaque-derived panfilovirus antibody that binds to the ebolavirus GP IFL across four different species of ebolavirus, EBOV, SUDV, RESTV, and BDBV (U.S. Provisional Application No. 62/406,598).

Antibodies that bind between the tip of the fusion loop and the base GP1/GP2 epitope:

[0211] ADI-15742: The ADI-15742 mAb was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-15742 is an ultra-potent pan- ebolavirus neutralizing antibody demonstrating sub-nanomolar IC50 values against VSV pseudovirions displaying GP from the following species: EBOV, BDBV, SUDV, RESTV and TAFV (as determined by the rVSV GP-GFP assay described in Example 1). ADI-15742 also provides complete protection against either EBOV or SUDV in their respective murine models.

[0212] ADI-15878: The ADI-15878 mAb was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak and is a clonal relative of ADI-15742 (Bornholdt et al). ADI-15878 is also an ultra-potent pan-ebolavirus neutralizing antibody demonstrating sub- nanomolar IC50 values against VSV pseudovirions displaying GP from the following species: EBOV, BDBV, SUDV, RESTV and TAFV (as determined by the rVSV GP-GFP assay described in Example 1). ADI-15878 showed significant levels of protection against EBOV and complete protection against SUDV in their respective murine models. [0213] ADI-15946: The ADI-15946 mAb was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-15946 is a potent pan-ebolavirus neutralizing antibody demonstrating sub-nanomolar IC50 values against VSV pseudovirions displaying GP from the following species: EBOV, BDBV, and SUDV (as determined by the rVSV GP-GFP assay described in Example 1).

Antibodies that bind to the viral membrane proximal (stalk) region of filovirus glycoprotein:

[0214] ADI- 16061: AD I- 16061 is a human mAb was derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-16061 binds to the heptad repeat 2 helices in the stalk region of EBOV GP, BDBV GP, and SUDV GP. However, ADI-16061 only effectively neutralizes VSV pseudovirions displaying GP from EBOV and BDBV with IC50 values 0.21 nM and 0.59 nM, respectively (as determined by the rVSV GP-GFP assay described in Example 1). Further ADI-16061 provided significant levels of protection from EBOV in the murine infection model post infection.

[0215] ADI-15974, ADI-15956, and ADI-15758: ADI-15974, ADI-15956, and ADI-15758 are clonally related human mAbs derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). These mAbs bind to the heptad repeat 2 helices in the stalk region of EBOV GP and BDBV GP. All three mAb effectively neutralize VSV pseudovirions displaying GP from EBOV and BDBV with sub-nanomolar IC50 values. In PRNT assays ADI-15974 ADI-15956, and ADI-15758 potently neutralize EBOV (as determined by the rVSV GP-GFP assay described in Example 1) and provided significant levels of protection from EBOV in the murine infection model post infection.

[0216] ADI-15848: ADI-15848 is a human mAb derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-15848 binds to the heptad repeat 2 helices in the stalk region of EBOV GP and BDBV GP. ADI-15848 effectively neutralizes VSV pseudovirions displaying GP from EBOV and BDBV with IC50 values 0.50 nM and 0.77 nM, respectively (as determined by the rVSV GP-GFP assay described in Example 1). In plaque reduction neutralization (PRNT) assays ADI-15848 potently neutralized EBOV and provided significant levels of protection from EBOV in the murine infection model post infection.

[0217] ADI-16021: ADI-16021 is a human mAb derived from a human survivor (subject 45) of the 2014 Ebola virus outbreak (Bornholdt et al). ADI-16021 binds to the heptad repeat 2 helices in the stalk region of EBOV GP, BDBV GP and SUDV GP. The breadth and scope of the present disclosure should not be limited by any of the above-described exemplary embodiments, but should be defined only in accordance with the following claims and their equivalents.

Ebolavirus GP sequences:

SEQ ID NO: 1 : Ebola vims (Mayinga strain) glycoprotein (Genbank Acc# AF086833):

MGVTGILQLPRDRFKRTSFFLWVIILFQRTFSIPLGVIHNSTLQVSDVDKLVCRDKL S

STNQLRSVGLNLEGNGVATDVPSATKRWGFRSGVPPKVVNYEAGEWAENCYNLEI

KKPDGSECLPAAPDGIRGFPRCRYVHKVSGTGPCAGDFAFHKEGAFFLYDRLASTV

IYRGTTFAEGVVAFLILPQAKKDFFSSHPLREPVNATEDPSSGYYSTTIRYQATGFG T

NETEYLFEVDNLTYVQLESRFTPQFLLQLNETIYTSGKRSNTTGKLIWKVNPEIDTT I

GEWAFWETKKNLTRKIRSEELSFTVVSNGAKNISGQSPARTSSDPGTNTTTEDHKF

ASENSSAMVQVHSQGREAAVSHLTTLATISTSPQSLTTKPGPDNSTHNTPVYKLDIS

EATQVEQHHRRTDNDSTASDTPSATTAAGPPKAENTNTSKSTDFLDPATTTSPQNH

SETAGNNNTHHQDTGEESASSGKLGLITNTIAGVAGLITGGRRTRREAIVNAQPKCN

PNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYIEGLMHNQDGLICGLRQLANETTQA

LQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGPDCCIEPHDWTKNITDKIDQ II

HDFVDKTLPDQGDNDNWWTGWRQWIPAGIGVTGVIIAVIALFCICKFVF

SEQ ID NO: 2: Delta mucin Ebola virus (Mayinga strain) glycoprotein:

MGVTGILQLPRDRFKRTSFFLWVIILFQRTFSIPLGVIHNSTLQVSDVDKLVCRDKLS

STNQLRSVGLNLEGNGVATDVPSATKRWGFRSGVPPKVVNYEAGEWAENCYNLEI

KKPDGSECLPAAPDGIRGFPRCRYVHKVSGTGPCAGDFAFHKEGAFFLYDRLASTV

IYRGTTFAEGVVAFLILPQAKKDFFSSHPLREPVNATEDPSSGYYSTTIRYQATGFG T

NETEYLFEVDNLTYVQLESRFTPQFLLQLNETIYTSGKRSNTTGKLIWKVNPEIDTT I

GEWAFWETKKNLTRKIRSEELSFTVVSHHQDTGEESASSGKLGLITNTIAGVAGLIT

GGRRTRREAIVNAQPKCNPNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYIEGLMHN

QDGLICGLRQLANETTQALQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGPD

CCIEPHDWTKNITDKIDQIIHDFVDKTLPDQGDNDNWWTGWRQWIPAGIGVTGVII

AVIALFCICKFVF

SEQ ID NO: 3 : Ebola virus (Kikwit strain) glycoprotein (Genbank Acc# KU182909):

MGVTGILQLPRDRFKRTSFFLWVIILFQRTFSIPLGVIHNSTLQVSDVDKLVCRDKL S

STNQLRSVGLNLEGNGVATDVPSATKRWGFRSGVPPKVVNYEAGEWAENCYNLEI

KKPDGSECLPAAPDGIRGFPRCRYVHKVSGTGPCAGDFAFHKEGAFFLYDRLASTV

IYRGTTFAEGVVAFLILPQAKKDFFSSHPLREPVNATEDPSSGYYSTTIRYQATGFG T ETEYLFEVD LTYVQLESRFTPQFLLQLNETIYTSGKRSNTTGKLIWKVNPEIDTTI

GEWAFWETKK LTRKIRSEELSFTAVS EAKNISGQSPARTSSDPGTNTTTEDHKF

ASENSSAMVQVHSQGREAAVSHLTTLATISTSPQPPTTKPGPDNSTHNTPVYKLDIS

EATQVEQHHRRTD DSTASDTPPATTAAGPLKAENTNTSKGTDLLDPATTTSPQ H

SETAGNNNTHHQDTGEESASSGKLGLITNTIAGVAGLITGGRRARREAIVNAQPKC P LHYWTTQDEGAAIGLAWIPYFGPAAEGIYIEGLMHNQDGLICGLRQLA ETTQ

ALQLFLRATTELRTFSIL RKAIDFLLQRWGGTCHILGPDCCIEPHDWTKNITDKIDQ

IIHDFVDKTLPDQGDNDNWWTGWRQWIPAGIGVTGVIIAVIALFCICKFVF

SEQ ID NO: 4: Delta mucin Ebola vims (Kikwit strain) glycoprotein:

MGVTGILQLPRDRFKRTSFFLWVIILFQRTFSIPLGVIHNSTLQVSDVDKLVCRDKLS

STNQLRSVGLNLEGNGVATDVPSATKRWGFRSGVPPKVVNYEAGEWAENCYNLEI

KKPDGSECLPAAPDGIRGFPRCRYVHKVSGTGPCAGDFAFHKEGAFFLYDRLASTV

IYRGTTFAEGVVAFLILPQAKKDFFSSHPLREPVNATEDPSSGYYSTTIRYQATGFG T

NETEYLFEVDNLTYVQLESRFTPQFLLQLNETIYTSGKRSNTTGKLIWKVNPEIDTT I

GEWAFWETKKNLTRKIRSEELSFTAVSHHQDTGEESASSGKLGLITNTIAGVAGLIT

GGRRARREArVNAQPKCNPNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYIEGLMHN

QDGLICGLRQLANETTQALQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGPD

CCIEPHDWTKNITDKIDQIIHDFVDKTLPDQGDNDNWWTGWRQWIPAGIGVTGVII

AVIALFCICKFVF

SEQ ID NO: 5: Ebola virus (Makona strain) glycoprotein (Genbank Acc# KT013256):

MGVTGILQLPRDRFKRTSFFLWVIILFQRTFSIPLGVIHNSTLQVSDVDKLVCRDKL S

STNQLRSVGLNLEGNGVATDVPSATKRWGFRSGVPPKVVNYEAGEWAENCYNLEI

KKPDGSECLPAAPDGIRGFPRCRYVHKVSGTGPCAGDFAFHKEGAFFLYDRLASTV

IYRGTTFAEGVVAFLILPQAKKDFFSSHPLREPVNATEDPSSGYYSTTIRYQATGFG T

NETEYLFEVDNLTYVQLESRFTPQFLLQLNETIYASGKRSNTTGKLIWKVNPEIDTT I

GEWAFWETKKNLTRKIRSEELSFTAVSNGPKNISGQSPARTSSDPETNTTNEDHKFM

ASENSSAMVQVHSQGRKAAVSHLTTLATISTSPQPPTTKTGPDNSTHNTPVYKLDIS

EATQVGQHHRRADNDSTASDTPPATTAAGPLKAENTNTSKSADSLDLATTTSPQNY

SETAGNNNTHHQDTGEESASSGKLGLITNTIAGVAGLITGGRRTRREVIVNAQPKCN

PNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYTEGLMHNQNGLICGLRQLANETTQ ALQLFLRATTELRTFSIL RKAIDFLLQRWGGTCHILGPDCCIEPHDWTKNITDKIDQ IIHDFVDKTLPDQGDNDNWWTGWRQWIPAGIGVTGVIIAVIALFCICKFVF

SEQ ID NO: 6: Delta mucin Ebola vims (Makona strain) glycoprotein:

MGVTGILQLPRDRFKRTSFFLWVIILFQRTFSIPLGVIHNSTLQVSDVDKLVCRDKLS

STNQLRSVGLNLEGNGVATDVPSATKRWGFRSGVPPKVVNYEAGEWAENCYNLEI

KKPDGSECLPAAPDGIRGFPRCRYVHKVSGTGPCAGDFAFHKEGAFFLYDRLASTV

IYRGTTFAEGVVAFLILPQAKKDFFSSHPLREPVNATEDPSSGYYSTTIRYQATGFG T

NETEYLFEVDNLTYVQLESRFTPQFLLQLNETIYASGKRSNTTGKLIWKVNPEIDTT I

GEWAFWETKKNLTRKIRSEELSFTAVSHHQDTGEESASSGKLGLITNTIAGVAGLIT

GGRRTRREVIVNAQPKCNPNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYTEGLMH

NQNGLICGLRQLANETTQALQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGP

DCCIEPHDWTKNITDKIDQIIHDFVDKTLPDQGDNDNWWTGWRQWIPAGIGVTGVI

IAVIALFCICKFVF

SEQ ID NO: 7: Sudan virus (Boniface strain) glycoprotein (Genbank# FJ968794 ):

MEGLSLLQLPRDKFRKSSFFVWVIILFQKAFSMPLGVVTNSTLEVTEIDQLVCKDHL

ASTDQLKSVGLNLEGSGVSTDIPSATKRWGFRSGVPPKVFSYEAGEWAENCYNLEI

KKPDGSECLPPPPDGVRGFPRCRYVHKAQGTGPCPGDYAFHKDGAFFLYDRLAST

VIYRGVNFAEGVIAFLILAKPKETFLQSPPIREAVNYTENTSSYYATSYLEYEIENF G

AQHSTTLFKINNNTFVLLDRPHTPQFLFQLNDTIHLHQQLSNTTGKLIWTLDANINA

DIGEWAFWENKKNLSEQLRGEELSFETLSLNETEDDDATSSRTTKGRISDRATRKYS

DLVPKDSPGMVSLHVPEGETTLPSQNSTEGRRVDVNTQETITETTATIIGTNGNNMQ

ISTIGTGLSSSQILSSSPTMAPSPETQTSTTYTPKLPVMTTEESTTPPRNSPGSTTE APT

LTTPENITTAVKTVLPQESTSNGLITSTVTGILGSLGLRKRSRRQVNTRATGKCNPN L

HYWTAQEQHNAAGIAWIPYFGPGAEGIYTEGLMHNQNALVCGLRQLANETTQAL

QLFLRATTELRTYTILNRKAIDFLLRRWGGTCRILGPDCCIEPHDWTKNITDKINQI IH

DFIDNPLPNQDNDDNWWTGWRQWIPAGIGITGIIIAIIALLCVCKLLC

SEQ ID NO: 8: Delta mucin Sudan virus (Boniface strain) glycoprotein:

MEGLSLLQLPRDKFRKSSFFVWVIILFQKAFSMPLGVVTNSTLEVTEIDQLVCKDHL ASTDQLKSVGLNLEGSGVSTDIPSATKRWGFRSGVPPKVFSYEAGEWAENCYNLEI KKPDGSECLPPPPDGVRGFPRCRYVHKAQGTGPCPGDYAFHKDGAFFLYDRLAST VIYRGVNFAEGVIAFLILAKPKETFLQSPPIREAVNYTENTSSYYATSYLEYEIENFG

AQHSTTLFKINNNTFVLLDRPHTPQFLFQL DTIHLHQQLSNTTGKLIWTLDANINA

DIGEWAFWE KKNLSEQLRGEELSFETLSTTAVKTVLPQESTSNGLITSTVTGILGSL

GLRKRSRRQVNTRATGKC PNLHYWTAQEQHNAAGIAWIPYFGPGAEGIYTEGLM

HNQNALVCGLRQLA ETTQALQLFLRATTELRTYTIL RKAIDFLLRRWGGTCRIL

GPDCCIEPHDWTKNITDKINQIIHDFID PLPNQD DDNWWTGWRQWIPAGIGITGII

IAIIALLCVCKLLC

SEQ ID NO: 9: Sudan virus (Gulu strain) glycoprotein (Genbank# AY729654):

MGGLSLLQLPRDKFRKS SFFVW VIILFQKAF SMPLGVVTNSTLEVTEIDQLVCKDHL

ASTDQLKSVGLNLEGSGVSTDIPSATKRWGFRSGVPPKVVSYEAGEWAENCYNLEI

KKPDGSECLPPPPDGVRGFPRCRYVHKAQGTGPCPGDYAFHKDGAFFLYDRLAST

VIYRGVNFAEGVIAFLILAKPKETFLQSPPIREAVNYTENTSSYYATSYLEYEIENF G

AQHSTTLFKIDNNTFVRLDRPHTPQFLFQLNDTIHLHQQLSNTTGRLIWTLDANINA

DIGEWAFWENKKNLSEQLRGEELSFEALSLNETEDDDAASSRITKGRISDRATRKYS

DLVPKNSPGMVPLHIPEGETTLPSQNSTEGRRVGVNTQETITETAATIIGTNGNHMQ I

STIGIRPSSSQIPSSSPTTAPSPEAQTPTTHTSGPSVMATEEPTTPPGSSPGPTTEA PTLT

TPENITTAVKTVLPQESTSNGLITSTVTGILGSLGLRKRSRRQTNTKATGKCNPNLH Y

WTAQEQHNAAGIAWIPYFGPGAEGIYTEGLMHNQNALVCGLRQLANETTQALQLF

LRATTELRTYTILNRKAIDFLLRRWGGTCRILGPDCCIEPHDWTKNITDKF QIIHDFI

DNPLPNQDNDDNWWTGWRQWIPAGIGITGIIIAIIALLCVCKLLC

SEQ ID NO: 10: Delta mucin Sudan virus (Gulu strain) glycoprotein:

MGGLSLLQLPRDKFRKS SFFVW VIILFQKAF SMPLGVVTNSTLEVTEIDQLVCKDHL

ASTDQLKSVGLNLEGSGVSTDIPSATKRWGFRSGVPPKVVSYEAGEWAENCYNLEI

KKPDGSECLPPPPDGVRGFPRCRYVHKAQGTGPCPGDYAFHKDGAFFLYDRLAST

VIYRGVNFAEGVIAFLILAKPKETFLQSPPIREAVNYTENTSSYYATSYLEYEIENF G

AQHSTTLFKIDNNTFVRLDRPHTPQFLFQLNDTIHLHQQLSNTTGRLIWTLDANINA

DIGEWAFWENKKNLSEQLRGEELSFEALSTTAVKTVLPQESTSNGLITSTVTGILGS L

GLRKRSRRQTNTKATGKCNPNLHYWTAQEQHNAAGIAWIPYFGPGAEGIYTEGLM

HNQNALVCGLRQLANETTQALQLFLRATTELRTYTILNRKAIDFLLRRWGGTCRIL

GPDCCIEPHDWTKNITDKINQIIHDFIDNPLPNQDNDDNWWTGWRQWIPAGIGITGI I

IAIIALLCVCKLLC SEQ ID NO: 11 : Bundibugyo virus (tc/UGA/2007/Bundibugyo-200706291) glycoprotein (Genbank# KU182911):

MVTSGILQLPRERFRKTSFFVWVIILFHKVFPIPLGVVHNNTLQVSDIDKLVCRDKLS

STSQLKSVGLNLEGNGVATDVPTATKRWGFRAGVPPKVVNYEAGEWAENCYNLD

n KADGSECLPEAPEGVRGFPRCRYVHKVSGTGPCPEGYAFHKEGAFFLYDRLASTI

IYRSTTFSEGVVAFLILPETKKDFFQSPPLHEPANMTTDPSSYYHTVTLNYVADNFG

TNMTNFLFQVDHLTYVQLEPRFTPQFLVQLNETIYTNGRRSNTTGTLIWKVNPTVD

TGVGEWAFWENKKNFTKTLSSEELSVIFVPRAQDPGSNQKTKVTPTSFANNQTSKN

HEDLVPEDPASVVQVRDLQRENTVPTPPPDTVPTTLIPDTMEEQTTSHYEPPNISRN

HQERNNT AFIPETL ANNPPDNTTP STPPQDGERT S SHTTP SPRP VPT STIUPTTRETHIP

TTMTTSHDTDSNRPNPIDISESTEPGPLTNTTRGAANLLTGSRRTRREITLRTQAKC N

PNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYTEGFMHNQNGLICGLRQLANETTQA

LQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGPDCCIEPHDWTKNITDKIDQ II

HDFIDKPLPDQTDNDNWWTGWRQWVPAGIGITGVIIAVIALLCICKFLL

SEQ ID NO: 12: Delta mucin Bundibugyo virus (tc/UGA/2007/Bundibugyo-200706291) glycoprotein:

MVTSGILQLPRERFRKTSFFVWVIILFHKVFPIPLGVVHNNTLQVSDIDKLVCRDKLS

STSQLKSVGLNLEGNGVATDVPTATKRWGFRAGVPPKVVNYEAGEWAENCYNLD

n KADGSECLPEAPEGVRGFPRCRYVHKVSGTGPCPEGYAFHKEGAFFLYDRLASTI

IYRSTTFSEGVVAFLILPETKKDFFQSPPLHEPANMTTDPSSYYHTVTLNYVADNFG

TNMTNFLFQVDHLTYVQLEPRFTPQFLVQLNETIYTNGRRSNTTGTLIWKVNPTVD

TGVGEWAFWENKKNFTKTLSSEELSVIFVPSNRPNPIDISESTEPGPLTNTTRGAAN L

LTGSRRTRREITLRTQAKCNPNLHYWTTQDEGAAIGLAWIPYFGPAAEGIYTEGFMH

NQNGLICGLRQLANETTQALQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGP

DCCIEPHDWTKNITDKIDQIIHDFIDKPLPDQTDNDNWWTGWRQWVPAGIGITGVII

AVIALLCICKFLL

SEQ ID NO: 13 : Reston virus (Pennsylvania strain) glycoprotein (Genbank# AF522874): MGSGYQLLQLPRERFRKTSFLVWVIILFQRAISMPLGIVTNSTLKATEIDQLVCRDKL SSTSQLKSVGLNLEGNGIATDVPSATKRWGFRSGVPPKVVSYEAGEWAENCYNLEI KKSDGSECLPLPPDGVRGFPRCRYVHKVQGTGPCPGDLAFHKNGAFFLYDRLASTV IYRGTTF AEGV V AFLIL SEPKKHF WK ATP AHEP VNTTDD S T S YYMTLTL S YEMSNF

GGNESNTLFKVDNHTYVQLDRPHTPQFLVQLNETLRRNNRLSNSTGRLTWTLDPKI

EPD VGEWAFWETKK F SQQLHGENLHFQIP STHTNNS SDQSP AGTVQGKIS YHPP A

NNSELVPTDSPPVVSVLTAGRTEEMSTQGLTNGETITGFTA PMTTTIAPSPTMTSE

VDNNVPSEQPNNTASIEDSPPSAS ETIYHSEMDPIQGSNNSAQSPQTKTTPAPTTSP

MTQDPQETANSSKPGTSPGSAAGPSQPGLTINTVSKVADSLSPTRKQKRSVRQNTA KC PDL Y YWT A VDEGA A VGL AWIP YF GP A AEGI YIEGVMHNQNGLIC GLRQL AN

ETTQALQLFLRATTELRTYSLLNRKAIDFLLQRWGGTCRILGPSCCIEPHDWTKNIT

DEINQIKHDFIDNPLPDHGDDLNLWTGWRQWIPAGIGIIGVIIAIIALLCICKILC

SEQ ID NO: 14: Delta mucin Reston vims (Pennsylvania strain) glycoprotein:

MGSGYQLLQLPRERFRKTSFLVWVIILFQRAISMPLGIVTNSTLKATEIDQLVCRDKL

SSTSQLKSVGLNLEGNGIATDVPSATKRWGFRSGVPPKVVSYEAGEWAENCYNLEI

KKSDGSECLPLPPDGVRGFPRCRYVHKVQGTGPCPGDLAFHKNGAFFLYDRLASTV

IYRGTTF AEGV V AFLIL SEPKKIIF WK ATP AtfEP VNTTDD S T S YYMTLTL S YEMSNF

GGNESNTLFKVDNHTYVQLDRPHTPQFLVQLNETLRRNNRLSNSTGRLTWTLDPKI

EPDVGEWAFWETKKNFSQQLHGENLHFQIPSKPGTSPGSAAGPSQPGLTINTVSKV

ADSLSPTRKQKRSVRQNTANKCNPDLYYWTAVDEGAAVGLAWIPYFGPAAEGIYI

EGVMHNQNGLICGLRQLANETTQALQLFLRATTELRTYSLLNRKAIDFLLQRWGGT

CRILGPSCCIEPHDWTKNITDEINQIKHDFIDNPLPDHGDDLNLWTGWRQWIPAGIG I

IGVIIAIIALLCICKILC

SEQ ID NO: 15: Tai Forrest virus (tc/CIV/1994/Pauleoula-CI strain) glycoprotein (Genbank# NC_014372):

MGASGILQLPRERFRKTSFFVWVIILFHKVFSIPLGVVHNNTLQVSDIDKFVCRDKLS

STSQLKSVGLNLEGNGVATDVPTATKRWGFRAGVPPKVVNCEAGEWAENCYNLA

n KVDGSECLPEAPEGVRDFPRCRYVHKVSGTGPCPGGLAFHKEGAFFLYDRLASTI

IYRGTTF AEGVIAFLILPKARKDFFQSPPLHEPANMTTDPSSYYHTTTINYVVDNFGT

NTTEFLFQVDHLTYVQLEARFTPQFLVLLNETIYSDNRRSNTTGKLIWKF PTVDTS

MGEWAFWENKKNFTKTLSSEELSFVPVPETQNQVLDTTATVSPPISAHNHAAEDHK

ELVSEDSTPVVQMQNIKGKDTMPTTVTGVPTTTPSPFPINARNTDHTKSFIGLEGPQ

EDHSTTQPAKTTSQPTNSTESTTLNPTSEPSSRGTGPSSPTVPNTTESHAELGKTTP TT

LPEQHTAASAIPRAVHPDELSGPGFLTNTIRGVTNLLTGSRRKRRDVTPNTQPKCNP LHYWTALDEGAAIGLAWIPYFGPAAEGIYTEGIMENQNGLICGLRQLA ETTQAL QLFLRATTELRTFSIL RKAIDFLLQRWGGTCHILGPDCCIEPQDWTKNITDKIDQIIH DFVDN LPNQ DGSNWWTGWKQWVPAGIGITGVIIAIIALLCICKFML

SEQ ID NO: 16: Delta mucin Tai Forrest virus (tc/CIV/1994/Pauleoula-CI strain) glycoprotein:

MGASGILQLPRERFRKTSFFVWVIILFHKVFSIPLGVVHNNTLQVSDIDKFVCRDKLS

STSQLKSVGLNLEGNGVATDVPTATKRWGFRAGVPPKVVNCEAGEWAENCYNLA

n KVDGSECLPEAPEGVRDFPRCRYVHKVSGTGPCPGGLAFHKEGAFFLYDRLASTI

IYRGTTFAEGVIAFLILPKARKDFFQSPPLHEPANMTTDPSSYYHTTTINYVVDNFG T

NTTEFLFQVDHLTYVQLEARFTPQFLVLLNETIYSDNRRSNTTGKLIWKF PTVDTS

MGEWAFWENKKNFTKTLSSEELSFVPVPSAIPRAVHPDELSGPGFLTNTIRGVTNLL

TGSRRKRRDVTPNTQPKCNPNLHYWTALDEGAAIGLAWIPYFGPAAEGIYTEGF E

NQNGLICGLRQLANETTQALQLFLRATTELRTFSILNRKAIDFLLQRWGGTCHILGP

DCCIEPQDWTKNITDKIDQIIHDFVDNNLPNQNDGSNWWTGWKQWVPAGIGITGVI

I AIIALLC ICKFML