Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
VIRUS-LIKE PARTICLE COMPOSITIONS AND VACCINES AGAINST EPSTEIN-BARR VIRUS INFECTION AND DISEASE
Document Type and Number:
WIPO Patent Application WO/2016/149384
Kind Code:
A1
Abstract:
The present iaveatioa relates to prophylactic and/or therapeutic vaccines thatpoatairj Newcastle disease Virus (NDV) vinis-like particles (VLPs) comprising one or mere Epstein- Ban Virus (EBV) antigens, in one embodiment, the invention provides a recombinant vims-iske particle (V'UP) comprising, i is operable combination, a) Neweasile disease virn^ iNDVj matrix (M) protein, and b) one or more Epstein-Barr Virus (BBV ) antigens. The im'eniion's prophylactic and/or therapeutic vacelrses are useful for preventing asd/or treatmg. infection with EBY aixi/or disease associated Epstein-Barr Virus, such as cancer.

Inventors:
OGEMBO JAVIER GORDON (US)
MORRISON TRUDY (US)
Application Number:
PCT/US2016/022663
Publication Date:
September 22, 2016
Filing Date:
March 16, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV MASSACHUSETTS (US)
International Classes:
A61K39/12; A61P31/00; C12N7/04; C12N15/38; C12N15/45
Domestic Patent References:
WO2014018858A22014-01-30
Foreign References:
US20090252761A12009-10-08
US20140227305A12014-08-14
Other References:
OGEMBO, JG ET AL.: "A Chimeric EBV gp350/220-based VLP Replicates The Virion B- cell Attachment Mechanism And Elicits Long-Lasting Neutralizing Antibodies In Mice.", JOURNAL OF TRANSLATIONAL MEDICINE., vol. 13, no. 1, 6 February 2015 (2015-02-06), pages 1 - 14, XP055311928
SMITH, C ET AL.: "A New Approach For Cellular Immunotherapy Of Nasopharyngeal Carcinoma.", ONCOLMUUNOLOGY., vol. 1, no. 8, 2012, pages 1440 - 1442, XP055311929
Attorney, Agent or Firm:
CARROLL, Peter G. (LLP1440 Broadway, Suite 51, Oakland California, US)
Download PDF:
Claims:
CLAIMS

We Claim:

1. A recombinant virus-like particle (VLP) comprising, in operable combination,

a) Newcastle disease virus (NDV) matrix (M) protein,

b) NDV nucleocapsid (NP) protein, and

c) one or more tumor-associated EBV antigen,

wherein said one or more tumor-associated EBV antigen is inside said VLP.

2. The VLP of Claim 1 , wherein said tumor-associated EBV antigen is selected from the group consisting of EBNA1, tEBNAl and LMP2.

3. The VLP of Claim 1, wherein said tumor-associated EBV antigen comprises tEBNAl and LMP2.

4. The VLP of Claim 3, further comprising, in operable combination, one or more Epstein-Barr Virus (EBV) antigens, wherein at least one of said one or more antigens is selected from the group consisting of gB, gH, and gL. 5. The VLP of Claim 3, wherein said VLP further comprises, in operable combination, EBV gp350/220.

6. The VLP of Claim 3, wherein said VLP further comprises, in operable combination, one or more NDV proteins.

7. The VLP of Claim 6, wherein said one or more NDV proteins comprise NDV heptad repeat domain 2 (HR2) protein.

8. The VLP of Claim 7, wherein said one or more NDV proteins comprise NDV fusion (F) protein.

9. The VLP of Claim 8, wherein said one or more NDV proteins comprise NDV heamagglutinin- neuraminidase (HN) protein.

10. The VLP of Claim 1, further comprising, in operable combination, one or more human papillomavirus antigens. 11. The VLP of Claim 10, wherein said one or more human papillomavirus antigens comprises one or more of LI and L2.

12. A vaccine comprising the VLP of Claim 1 and a physiologically acceptable carrier. 13. An expression, vector encoding the recombinant VLP of Claim 1.

14. A method for immunizing a mammalian subject against cancer, comprising administering an immunologically effective amount of one or more vaccine of Claim 12 to a mammalian subject in need thereof to produce a treated subject, wherein said administering is under conditions to produce an immune response to one or more tumor-associated EB V antigen.

15. The method of Claim 14, wherein said cancer comprises an Epstein-Barr Virus (EBV) -associated cancer. 16. The method of Claim 14, wherein said EBV-associated cancer comprises cervical cancer.

17. The method of Claim 14, wherein said immune response comprises T lymphocytes that specifically bind to said one or more tumor-associated EBV antigen. 18. The method of Claim 17, wherein said immune response lacks antibody that specifically binds to said one or more tumor-associated EBV antigen.

19. The method of Claim 17, wherein said T lymphocytes are selected from CD4+ lymphocytes and CD8+ lymphocytes.

20. The method of Claim 14, wherein said method further comprises administering a recombinant VLP that contains, in operable combination,

a) Newcastle disease virus (NDV) matrix (M) protein, and

b) EBV gp350/220.

21. The method of Claim 14, wherein said method further comprises one or more of

a) detecting said immune response to said one or more tumor-associated EBV antigen, and b) detecting a reduction in one or more symptoms of said cancer in said treated subject.

22. The method of Claim 14, wherein said administering is before manifestation of one or more symptoms of said cancer.

23. The method of Claim 14, wherein said administering is after manifestation of one or more symptoms of said cancer.

Description:
VIRUS-LIKE PARTICLE COMPOSITIONS AND VACCINES AGAINST

EPSTEIN-BARR VIRUS INFECTION AND DISEASE

This application claims priority to co-pending U.S. provisional Application Serial No.

62/134,785, filed on March 18, 2015, which is herein incorporated by reference in its entirety.

FIELD OF THE INVENTION

The present invention relates to prophylactic and/or therapeutic vaccines that contain

Newcastle disease virus (NDV) virus-like particles (VLPs) comprising one or more Epstein-Barr Virus (EBV) antigens. The invention's prophylactic and/or therapeutic vaccmes are useful for preventing and/or treating infection with EBV and/or disease associated Epstein-Barr Virus, such as cancer.

BACKGROUND OF THE INVENTION

Epstein-Barr virus (EBV), an oncogenic gammahcrpesvirus, causes acute infectious mononucleosis (AIM) and is linked to the development of several human malignancies. Approaches for EBV vaccine development are limited due in part to the oncogenic potential of the EBV genome and lack of animal models to test vaccine candidates. The EBV envelope glycoprotein, gp350/220, has been proposed as a vaccine antigen. However, in small Phase Ι/Π clinical trials, vaccination with either vector constructs expressing gp350/220, or with the purified recombinant gp350 protein, did not prevent EBV infection although it did reduce the incidence of acute infectious mononucleosis (AIM) in young adults. Importantly, recombinant EBVAgp350/220 can infect both epithelial and primary B cells in vitro. While previous studies indicate that immunity to gp350/220 can limit infection, the poor success of using gp350/220 as a single vaccine antigen calls for innovative approaches utilizing multiple EBV proteins.

At least 4 EBVgp350/220 vaccine candidates have been tested in "clinical trials" such as

Vaccinia vector expressing gp350/220 (Gu et al., 1995 (Phase I- Chinese population, EBV naive 1-3 years old children), and Recombinant gp350 in CHO cells (Non-splicing variant) (3 dose regimen adjuvanted with AS04) (Jackman et al. 1999; Moutchen et al, 2007. (Phase l/l I) Safety and

Immunogcnicity in aged 18-37 years old EBV naive Belgians; Sokal et al., 2007. Phase I randomized, double-blind placebo control in aged 16-25 years EBV naive Belgians; Rees et al., 2009. Phase I chronic kidney disease kids awaiting organ transplants (UK)). However, none of these vaccine candidates achieved complete blockage of EBV infection. Notably, EBNA1, LMP2 and gp350/220 antigens have been developed and independently tested in various clinical trials as vaccine candidates against EBV infection and EBV+ cells with promising results.

Candidate therapeutic vaccines in clinical trials include M VA-vector expressing EBNA-1 and LMP1 or LMP2 (Taylor et al., 2004 construction of the MVA vector expressing EBNA1 and or

LMP2; Hui et al., 2013- EBNA1-LMP2 (Phase I targeting NPC patients in China); Taylor et al., 2014 EBNA1-LMP2 (A Phase I Trial in UK Patients with EBV-Positive Cancer), as well as Adoptive transfer PBMCs for treatment of PTLDs and NPCs (Louis, et al., 2009, 2010, Heslop et al. 1996 T cells adoptive transfer, and Chia et al., 2012 Phase 1 targeting NPC patients in China. Dendritic cells are transduced with adenovirus vector expressing ΔLMP1-LMP2). A recent phase I cluneal trial of recombinant modified vaccinia Ankara (MVA) vector encoding deletion of Gly-Ala regions from the EBNA 1 sequence fused to LMP2 as a vaccine candidate elicited a robust EBV-spccific CD4+ and CD8+ T cell response in humans. However, the strategy used to deliver these two important EBV antigens, known for their oncogenic potential, may pose major health risks, particularly in

immunosuppressed individuals. Furthermore, these vaccine candidates cannot generate neutralizing antibodies to eliminate reactivation or new EBV infections. There is also a risk of vaccine tolerance since the protein is constantly produced.

Thus, there is an urgent need for EBV vaccines that are safe, prevent EBV infection and/or limit EBV disease symptoms.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1. Schematic diagram showing construction and assembly of NDV NP as a carrier of EGFP, truncated EBNA1, LMP2 (full length) and/or tEBNAl -LMP2 incorporated into either gp350/220 or gH/gL VLPs. (A) Construction of NP-EGFP chimera plasmid. (B) Diagram of full- length NDV-F (top), full-length wild type NDV (middle) and chimeric gp350/220 NDV F (bottom). (C) Schematic illustration of cDNAs pCAGGS -NDV-M, -NP-EGFP and gp350/220-F (chimera) all cotransfected into CHO cells for VLPs assembly and release. (D) Construction of NP-tEBNAl, LMP2 and/or tEBNAl -LMP2. (E) Schematic illustration of cDNAs pCAGGS NDV-M, - NP-tEBNA1 , LMP2 and/or tEBNAl -LMP2 and gH-F/gL-HN (chimeras) cotransfected into CHO cells for VLPs assembly and release.

Figure 2. Assembly and characterization of EBVgp350/220 VLPs incorporated with NDV-NP- EGFP. (A) Expression of EGFP in CHO cells transfected with pCAGGS-NDV-NP-EGFP detected by microscopy. (B) Immunolot detection of gp350/220 in VLPs purified from supernatant of transfected

CHO cells mAb-72Al (anti gp350/220). Lysates were separated in 4-12% gel. Purified EBV from

B95-8 cells was used as positive control.

Figure 3. Immunoblot analysis of EBV-EBNA1, NDV-NP and gp350/220 incorporated into VLPs. (A) Detection of NDV NP-EBNA1 from purified VLPs from supernatant of CHO transfected cells using polyclonal anti-NDV Ab. (B) Detection of EBNA1 in gp350/220 VLPs using specific Ab to EBNA1 DNA binding domain. (C) Detection of gp350/220 incorporated into VLPs (D) anti-EBNAl Gly-Arg rich repeat domain.4-12% gel was used.

Figure 4-47. Refer to each drawing for its description.

Figure 48. EBV entry pathways.

Figure 49. Challenges of DNA based vaccines.

Figures 50-53. Refer to each drawing for its description.

Figures 54. Terminal protein LMP2A [Human herpesvirus 4] (SEQ ID NO:01) NCBI

Reference Sequence: YP 401631.1.

Figure 55. Glycoprotein gp110 precursor [Human herpesvirus 4] (SEQ ID NO:02) NCBI Reference Sequence: YP_401713.1.

Figure 56. gH, Glycoprotein gp85 precursor [Human herpesvirus 4] (SEQ ID NO:03) NCBI Reference Sequence: YP_401700.1.

Figure 57. gL (BKRF2) [Human herpesvirus 4 type 2] (SEQ ID NO:04) NCBI Reference Sequence: YP_001 129472.1.

Figure 58. LI [Human papillomavirus type 16] (SEQ ID NO:05) GenBank: AAD33259.1.

Figure 59. L2 [Human papillomavirus type 16] (SEQ ID NO:06) GenBank: AAD33258.1.

Figure 60. L2 protein [Human papillomavirus type 18] (SEQ ID NO:07) GenBank:

AGG40790.1.

Figure 61. EBNA-1 protein [Human herpesvirus 4] (SEQ ID NO:08) NCBI Reference

Sequence: YP_401677.1.

Figure 62. EBNA-1_326-641 LMP2 NP in NDVNP in pCAGGS.SEQ Translation

Figure 63. EBV LMP2 in NDV NP in pCAGGS.SEQ Translation

Figure 64. EBV-EBNA-1 in NDV NP in pCAGGS.seq Translation

Figure 65. pCAGGS HN-EBVgL EGFP in pCAGGS.SEQ Translation

Figure 66. pUC57 EBV gH-NDV-F-TM-CT in pUC57-Amp.scq Translation

Figure 67. pUC57 EBV-gL WT in pUC57-Amp.seq Translation Figure 68. pUC57-EBVgB-NDV-F in pUC57-Amp.seq Translation

Figure 69. pUC57-EBV-gH-WT in pUC57-Amp.SEQ Translation

Figure 70. puC57-EBV-gL-NDV-HN in pUC57-Amp.SEQ Translation

Figure 71. EBV-pUC57-gB WT in pUC57-Amp.seq Translation

Figure 72. A diagram showing the transfection process used in the production of chimeric

EBVgp350/220(-/+)HR2 F VLPs. Ogembo et aL, J. Trans. Med. 2015; 13:50. Pantua et aL, J. Virol. 2006; 11062-73.

Figure 73. Schematic of EBVgp350/220 F chimeric protein. A diagram of full length wild type NDV-F (top), full length EBVgp350/220- WT (center), and chimeric EBVgp350/220(-/+)HR2 F contruct (bottom) (not to scale). C-terminal amino acid sequences comprising the gp350/220 ectodomain (ED) and N-terminal sequences from NDV-F HR2 at the point of fusion are indicated. The single line represents amino acid sequences deleted in frame in the gp220 isoform. Both isoforms contain the N-terminal B-cell attachment epitope. Ogembo et al., J. Trans. Med. 2015; 13:50.

Figure 74 Expression of EBVgp350/220-WT and EBVgp350/220-F was measured in three transfected cell lines (CHO, ELL-0, and 293A) compared to the vector control. One million cells from each cell line was transfected with 1μg of either pCAGGS-EBVgp350/220 WT, pCAGGS- EBVgp350/220-HR2 F, pCAGGS-EBVgp350/220+HR2 F or pCAGGS alone (vector control). At 72h post-transfection, cells were stained with anti-gp350/220 mAb-72Al followed by AF488-coup1ed goat anti-mouse IgG (H+L) and analyzed by flow cytometry.

Figure 75. Characterization of EBV-gp350/220-F VLPs. Cell supematants from independent

EBV VLP preparations were harvested daily between 24-96 h, concentrated, and purified by sucrose- gradient centrifugation followed by particle lysis and immunoblot analysis. (A) Supematants produced in CHO cells were pooled and purified. Immunoblot indicating the presence of EBVgp350/220 ED, and NDV-F C-terminal peptides using mAb-72Al anti-gp350/220 (left panel), polyclonal anti-HR2 (middle panel), and polyclonal anti-NDV (right panel), respectively. Each blot included lysates of purified EBV and ND, which served as controls. (B) Electron micrograph of negatively stained sucrose gradient purified EBVgp350/220-F VLPs prepared in CHO cells compared with native EBV using immunogold-coupled goat anti-mouse IgG binds the surface of a chimeric EBVgp350/220-F VLP and EBV (control). Image shows the structure and size of the chimeric VLP compared with EBV in the absence of antibody (top), and in the presence of mAb-72Al anti-gp350/220 (middle), as well as a chimeric KSHV-derived VLP or KSHV. (C) Different combinations of pCAGGS plasmids encoding F, M, NP, gp350/220 and gp350/220-F as indicated were co-transfected into 293T cells. Material released into the supernatant was pelleted and analyzed by immunoblot using anti-gp350/220 and anti-HR2 antibodies. Lane I : NDV-VLP (NP, M, F), lane 2: NP, M, lane 3: NP, M, gp350/220 WT, lane 4: EBV

VLP (NP, M, EBVgp350/220-F), lane 5: M, EBVgp350/220-F, lane 6: pCAGGS . Bands of the expected molecular weight were detected by both antibodies in lane 4 alone, indicating a chimeric

EBV VLP specifically assembled and was released. (D) Silver stain of increasing amounts of purified chimeric VLPs released from ELL-0 cells compared with NDV. The position of EBVgp350/220-F protein, NDV-NP and -M are indicated by arrows. Molecular weight markers are indicated at left.

Figure 76. Purified FBVgp350/220 F VLPs bind CD21 and CD35. Raji, Nalm6, and K562 cells lines transfected with either CD21 or CD35 were characterized and prepared as described (Ogembo et al., Cell Reports 2013; 371-385). Untransfected NALM6 cells were used as a control. The cells were stained with primary mAbs to either CD35 or CD21 depending on which complement receptor they had been transfected with followed by AF594-coupled goat Fab'2 anti-mouse IgG (red). Next, attachment of EBVgp350/220-F VLPs was detected directly with AF488-coupled anti- gp350/220 (mAb-2L10) (green) that recognizes an epitope distal to the attachment site. Cell content was documented by nuclear staining with DAPI. Sequential confocal images showed that the chimeric VLP binds to CD21 or CD35 bearing cells whereas no binding to receptor negative Nalm6 cells was seen. Visualization of 3D merged images confirmed extensive co-localization (yellow) of the chimeric VLPs with both CD21 and CD35. Ogembo et al., J. Trans. Med. 2015; 13:50.

Figure 77. Long-term IgG anti-gp350/220 antibodies are generated in mice immunized with EBVgp350/220-F VLPs, UV-EBV and soluble recombinant gp350/220 ED. Groups of five mice were immunized intraperitoneally with either EBVgp350/220(+)HR2 VLPs (green), EBVgp350/220(-)HR2 VLPs (black), inactivated UV-EBV (red), soluble recombinant gp350/220 ED (purple) or TNE, which served as vehicle control (blue). Each immunogen contained equivalent amounts of gp350/220, and both primary and booster vaccinations contained equivalent immunogen. Booster immunizations were performed on days 43, 172, 183 and 218 as indicated on the time line (bottom). All immunizations were performed in the absence of adjuvant. Anti-gp350/220 IgG titers were determined for 8 time points during the immunization schedule using ELISA. Ogembo et al., J. Trans. Med. 2015; 13:50.

Figure 78. Neutralization of EBV (EGFP-EBV) infection of Raji cells by pre-incubarion with antibodies generated in response to EBVgp350/220-F VLP immunization. (A) EBV infection assay. Because EBV does not plaque, the infectivity of EGFP-EBV from a frozen stock was directly quantitated by cytometry. Five microliters of stock virus in the absence of serum yielded ~50% infection (green fluorescence) 73 h after infection of Raji and was selected for neutralization experiments. (B) EBV neutralization assay. Pooled terminal bleed sera from groups of five BALB/c mice immunized with EBVgp350/220- F VLP, UV-EBV or soluble recombinant gp350/220 ED were pre-incubated with EGFP-EBV to assess neutralization (Methods). Infected cells were incubated at

37°C for 72 h at which time EGFP positive Raji cells were enumerated by cytometry. X-axis indicates neutralizing antibody source. Y- axis displays percent of EGFP+ Raji cells post-infection. Terminal sera or mAb controls were pre-incubated 1 :1 with EGFP-EBV resulting in further virus dilution such that ~25"o of Raji cells were maximally infected (fluoresced green) in the presence of the non-blocking mAb-2L10 or TNE. Pre-incubation with mAb-72A1 (neutralizing) served as the positive control.

Results are expressed as mean ± standard deviations (SD). Horizontal black lines terminating in short vertical lines compare sets of neutralization experiments with p values indicated above the line.

Ogembo e/tf/.-J. Trans. Med. 2015; 13:50.

Figure 79. Schematic of construction and assembly of NDV NP as a carrier of EGFP into gp350/220/HR2 F VLPs. Schematic of (A) NP-EGFP chimera plasmid construction; (B) gp350/220- HR2 F plasmid construction showing Hill-length NDV-F (top), full-length wild type gp350/220 with splicing sites (middle) and chimeric gp350/220 NDV F HR2 (bottom); and (C) co-transfection of plasmids into CHO cells for assembly and release of VLPs.

Figure 80 Characterization of EBVgp350/220 VLPs incorporated with NDV-NP-ECFP.

(A) Expression of EGFP in CHO cells transfected with pCAGGS-NDV-NP-EGFP detected by microscopy. (B) Immunoblot of VLPs purified from the supernatant of transfected CHO cells using mAb-72Al (anti-gp350/220) as the detection antibody. Lysates were separated on a 4-12% gel. Purified EBV from B95-8-EGFP cells was used as a positive control.

Figure 81. EBVgp350/220-HR2 F EGFP VLPs binds CD21 on Raji cells. Raji cells expressing complement receptor type 2 (CD21) were incubated with purified EBV, EBVgp350/220 F, EBVgp350/220-HR2 F, or EBVgp350/220-HR2 F NP-EGFP VLPs. Attachment of EBVgp350/220 VLPs was next detected directly with AF488-coupled anti-gp350/220 (mAb-2L10) that recognizes an epitope distal to the attachment site. The results were measured by flow cytometry. Figure 82. Characterization of EBVgp350/220-EBNAl VLPs. Purified EBVgp350/220- tEBNAl VLPs were lysed in non-reducing Laemmli buffer and run on 4-20% SDS-agarose gel and proteins were detected via immunoblot. (A) mAb-72Alanti-gp350/220, (B) mAb anti DNA binding domain EBV-EBNA1 (a gift from Dr. F. Grasser, Institut fur Virologie, Germany), (C) polyclonal anti- NDV, and (D) anti-EBNAl Gly-Arg regions Abs were used to detect respective proteins.

Figure 83. Construction and schematic illustration of EBVgH/gL-NP-tEBNAl VLP production and immunoblot detection of EBVgp350/220-NP-tEBNAl VLPs. Construction and schematic of: (A) EBV gH-F, (B) EBV gL-HN, and (C) NP-tEBNAl plasmids for VLP production. (D) EBVgp350/220-tEBNAl VLPs were produced as outlined in Figure 73, Purified EBVgp350/220- tEBNAl VLPs were lysed on non-reducing Laemmli buffer and run on 4-20% SDS-agarose gel followed by silver stain and immunoblot. Polyclonal anti-NDV (top panel), mAb anti DNA binding domain EBV-EBNA1 (a gift from Dr. F. Grasser, Institut fur Virologie, Germany, second panel), mAb-72Alanti-gp350/220 (third panel) and anti-EBNAl Gly-Arg regions (bottom panel) Abs were used to detect respective proteins.

Figure 84. Construction and schematic illustration of EBVgB-NP-LMP2 VLP or EBVgB- NP-IEBNA1-LMP2 VLP production. Schematic illustration of (A) NDV-F, EBVgB, EBVgB-F, NP- LMP2 and NO-tEBNAl -LMP2 plasmids for VLP production. (B) Schematic of cDNAs pCAGGS- NDV-M, -NP-tEBNAl -LMP2 or NP-LMP2, and -gB-F chimera co-transfected into CHO cells for VLP production.

Figure 85. Chimeric EBVgB-NDV F is well expressed on the surface of CHO cells. High expression of EBVgB-WT, EBVgB His and EBVgB-F on the surface of CHO cells. 10 6 cells were transfected with ^g of EBVgB-WT, EBVgB His and EBVgB-F or pCAGGS alone (vector control). At 72 h post-transfection, cells were stained with mAb anti-gB (clone CL55) followed by AF488- coupled goat anti-mouse IgG (H+L) and analyzed by flow cytometry.

Figure 86. Characterization of EBVgB-F INP-LMP2 and EBVgB- F NP-EBNA1 proteins were incorporated in VLPs. Characterization of EBVgp350/220-EBNAl VLPs. Purified VLPs were lysed on non-reducing Laemmli buffer and run on 4-20% SDS-agarose gel followed by immunoblot. (A) mAb-CL55 and BA23 anti-B (top and middle panels), polyclonal anti-NDV bottom panel), (B)mAb anti EBV-LMP2 (top panel), and polyclonal anti-NDV (bottom panel) Abs were used to detect respective proteins. (C) Silver stain of increasing amounts of purified chimeric VLPs released from CHO cells compared with NDV. The position of EBVgB-F LMP2 protein, NDV-NP and -M are indicated by arrows. Molecular weight markers are indicated at left. Figure 87. Experimental layout and immunization of BALB/c mice with VLPs carrying EBV glycoproteins. BALB/c mice were immunized with 10μg of EBVgH/gL-EBNAl VLP, EBVgB- LMP2 VLP, or UV-inactivated EBV and boosted twice at days 26 and 56 without adjuvants. TNE served as a negative control for immunogen. At day 161 mice were sacrificed Figure 88. Splenocytes from mice immunized with EBVgH/gL-EBNAl VLPs and EBVgB-

LMP2 VLPs generated significantly higher IFN-y than mice immunized with UV-inactivated EBV and EBVgp350/220 VLPs. BALB/c mice were immunized with 10ug of EBVgH/gL-EBNA1 VLP, EBVgB-LMP2 VLP, or UV-inactivated EBV and boosted twice at days 29 and 54 without adjuvants. TNE served as a negative control for immunogen. At day 161 mice were sacrificed and 5 * 10 5 splenocytes were stimulated in vitro with the corresponding as well as control peptides. 1 μg/mL of synthetic peptides derived from or Promix EBV peptide

pool consisting of 26 peptides, each corresponding to a defined HLA class I-restricted T cell epitope from EBV were used in the assay. After overnight culturing, the supernatants were tested for IFN-γ release by ELISA. SUNFEKL ovalbumin was used as a negative control and concavalin A and IL1B were used as model antigens.

DEFINITIONS

To facilitate understanding of the invention, a number of terms are defined below.

The term "recombinant" molecule refers to a molecule that is produced using molecular biological techniques. Thus, "recombinant DNA molecule" refers to a DNA molecule that is comprised of segments of DNA joined together by means of molecular biological techniques. A "recombinant protein" or "recombinant polypeptide" as used herein refers to a protein molecule that is expressed using a recombinant DNA molecule. A "recombinant" virus-like particle (VLP) refers to a VLP that is expressed using a recombinant DNA molecule.

A "virus-like particle" and "VLP" interchangeably refer to a non-replicating, non-infectious particle shell that contains one or more virus proteins, lacks the viral RNA and/or DNA genome, and that approximately resembles live virus in external conformation. Methods for producing and characterizing recombinant VLPs containing Newcastle Disease Virus (NDV) proteins have been described (Pantua et al. (2006) J. Virol. 80:11062-11073; U.S. Pat. No. 7,951,384 issued to Morrison et al. on May 01, 2011; U.S. Pat. No. 8,974,797, issued to Morrison on Mar-10-2015, each of which is incorporated by reference). Further methods for producing NDV VLPs are disclosed herein. The term "inside" a VLP when made in reference to the location of a polypeptide sequence means that the polypeptide sequence is located on the inner surface of the virus-like particle, and is encapsulated by the virus-like particle such that the polypeptide sequence is not exposed on the outside surface of the virus-like particle. Preferably, though not necessarily, the polypeptide that is inside the VLP is not accessible to binding with antibodies that are present outside the VLP.

"Operable combination" and "operably linked" when in reference to the relationship between nucleic acid sequences and/or amino acid sequences refers to linking (i.e., fusing) the sequences in frame such that they perform their intended function. For example, operably linking a promoter sequence to a nucleotide sequence of interest refers to linking the promoter sequence and the nucleotide sequence of interest in a manner such that the promoter sequence is capable of directing the transcription of the nucleotide sequence of interest and/or the synthesis of a polypeptide encoded by the nucleotide sequence of interest.

The term "matrix protein", "membrane protein", or "M protein" as used herein, means any protein localized between the envelope and the nucleocapsid core and facilitates the organization and maintenance of the virion structure and budding processes. Exemplary ND V M protein sequences include those described in U.S. Fat. No. 7,951,384 issued to Morrison et al. on May 01, 2011; U.S. Pat. No. 8,974,797, issued to Morrison on Mar-10-2015, each of which is incorporated by reference.

The term "nucleocapsid protein" or "NP protein" as used herein, means any protein that associates with genomic RNA (i.e., for example, one molecule per hexamer) and protects the RNA from nuclease digestion. Exemplary NP protein sequences from NDV include those described in U.S. Pat. No. 7,951 ,384 issued to Morrison et al. on May 01, 2011 ; U.S. Pat. No. 8,974,797, issued to Morrison on Mar-10-2015, each of which is incorporated by reference.

The term "fusion protein" or "F protein" as used herein, means any protein that projects from the envelope surface and mediates host cell entry by inducing fusion between the viral envelope and the cell membrane. However, it is not intended that the present invention be limited to functional F proteins. For example, an F protein may be encoded by a mutant F gene such as, but not limited to, F- K1 15Q. F-K.115Q is believed to eliminate the normal cleavage and subsequent activation of the fusion protein. F-K 1 15Q mimics naturally occurring F-protein mutations in avirulent NDV strains, and in cell culture, eliminates any potential side effects of cell-cell fusion on the release of VLPs. Exemplary NDV F protein sequences include those described in U.S. Pat. No. 7,951 ,384 issued to Morrison et al. on May 01, 2011; U.S. Pat. No. 8,974,797, issued to Morrison on Mar-10-2015, each of which is incorporated by reference. "HR2 domain," "hcptad repeat domain 2'' "HR-B domain," and "heptad repeat domain B" interchangeably refer to a sequence that is present in the F protein of a Paramyxovirus, and that folds as an amphipathic alpha helix. The HR2 domain of NDV is exemplified by those described in U.S.

Pat. No. 8,974,797, issued to Morrison on Mar- 10-2015, incorporated by reference.

The term 'haemagglutinin-neuramiiiidase protein 1 '. "HN protein", or G protein as used herein, means any protein that spans the viral envelope and projects from the surface as spikes to facilitate celt attachment and entry (i.e., for example, by binding to sialic acid on a cell surface). These proteins possess both haemagglutination and neuraminidase activity. Exemplary NDV IIN protein sequences include those described in U.S. Pat. No. 7,951 ,384 issued to Morrison ct al. on May 01 , 201 1 : U.S. Pat. No. 8,974,797. issued to Morrison on Mar- 10-2015, each of which is incorporated by reference.

The term "glycoprotein" as used herein, refers to any protein conjugated to a carbohydrate. "Epstcin-Barr Virus," "EBV," "human herpesvirus 4" and "I IHV-4" interchangeably refer to an oncogenic human herpesvirus. EBV is the cause of acute infectious mononucleosis (AIM, also known as glandular fever). It is also associated with particular forms of cancer, such as Hodgkin's lymphoma, Burkitt's lymphoma, nasopharyngeal carcinoma, and conditions associated with human immunodeficiency virus (HIV), such as hairy leukoplakia and central nervous system lymphomas. EBV infects B cells of the immune system and epithelial cells. Once the virus's initial lytic infection is brought under control. EBV latently persists in the individual's B cells for the rest of the individual's life due to a complex life cycle (Figure 48) that includes alternate latent and lytic phases.

"Symptom of EBV infection" includes acute infectious mononucleosis (AIM, also known as glandular fever) and/or the presence of EBV-associatcd cancer. "EBV-associated cancer" refers to cancer that is caused and/or aggravated, at least in part, by infection with EBV, such as Hodgkin's lymphoma, Burkitt's lymphoma, nasopharyngeal carcinoma, cervical cancer, hairy leukoplakia and central nervous system lymphomas.

The terms "antigen," ''immunogen," "antigenic." "immunogenic," "antigenically active," and

"immunologically active" when made in reference to a molecule, refer to any substance that is capable of inducing a specific humoral and/or ccll-mcdiatcd immune response. In a particular embodiment, the antigen comprises at least a portion or an cctodomain.

The term "ectodomain" when in reference to a membrane protein refers to the portion of the protein that is exposed on the extracellular side of a lipid bilaycr of a cell, virus and the like.

"EBV antigen" refers to an antigen from EBV, such as "'gB, gH, gL, and gp350/220" and tumor-associated EBV antigens. The term "gB" refers to glycoprotein gpl 10 precursor of Human herpesvirus 4 and is exemplified in Figure 55 (SEQ ID NO:02), NCBI Reference Sequence: YP_401713.1.

The term "gH" refers to glycoprotein gp85 precursor of human herpesvirus 4 and is

exemplified by in Figure 56 (SEQ ID NO:03), NCBI Reference Sequence: YP_401700.1.

The term "gL" and "BKRF2" are interchangeably used, and exemplified in Figure 57 by

BKRF2 protein of human herpesvirus 4 type 2 (SEQ ID NO:04), NCBI Reference Sequence:

YP_001129472.1.

The term "gp350/220" is the predominant EBV envelope protein. Interactions between

EBVgp350/220 and complement receptor type 2 (CR2)/CD21 and/or (CR1)/CD35 on B-cells is required for cellular attachment and initiation of latent infection.

'Tumor-associated EBV antigens" are EBV antigens that are associated with tumors in subjects who are infected with EBV. Exemplary tumor-associated EBV antigens include EBNA1 , LMP1, LMP2, and BARF1 , those described in Lin et al. "CD4 and CD8 T cell responses to tumour-associated Epstcin-Barr virus antigens in nasopharyngeal carcinoma patients," Cancer Immunol Immunother. 2008 Jul;57(7):963-75; Kohrt et al. "Dynamic CD8 T-cell responses to tumor-associated Epstein-Barr virus antigens in patients with Epstein-Barr virus-negative Hodgkin's disease," Oncol Res. 2009; 18(5- 6):287-92; Parmita et al., "Humoral immune responses to Epstein-Barr virus encoded tumor associated proteins and their putative extracellular domains in nasopharyngeal carcinoma patients and regional controls," J Med Virol. 201 1 Apr,83(4):665-78.

"EBNA 1 " is exemplified in Figure 8 by human herpesvirus 4 SEQ I D NO:08, NCBI Reference

Sequence: YP_401677.1.

"Truncated EBNA" and "tEBNA 1 " interchangeably refer to EBN A 1 that lacks the Gly-Ala rich domain known to impair presentation of cis-linked sequences, and are exemplified by the sequence from amino acid 325-641 of Figure 8's EBNA SEQ ID NO:08. Constructs containing tEBNAl are exemplified in Figures 50 and 52.

"LMP2" is exemplified in Figure 54 by the terminal protein LMP2A of human herpesvirus 4 (SEQ ID NO:01), NCBI Reference Sequence: YP_40l63 I.l.

"L1" is exemplified in Figure 58 by human papillomavirus type 16 SEQ ID NO:05, Gen Bank: AAD33259.1.

"L2" is exemplified in Figure 59 by human papillomavirus type 16 SEQ ID NO:06, GenBank:

AAD33258.1, and in Figure 60 by human papillomavirus type 18 SEQ ID NO:07, GenBank:

AGG40790.1. Physiologically acceptable "carrier" and "diluents" for vaccine preparation include water, saline solution, human serum albumin, oils, polyethylene glycols, aqueous dextrose, glycerin, propylene glycol or other synthetic solvents. Carriers may be liquid carriers (such as water, saline, culture medium, saline, aqueous dextrose, and glycols) or solid carriers (such as carbohydrates exemplified by starch, glucose, lactose, sucrose, and dextrans, antioxidants exemplified by ascorbic acid and glutathione, and hydrolyzed proteins).

The term "expression vector" refers to a nucleotide sequence containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression (i.e., transcription into

RNA and/or translation into a polypeptide) of the operably linked coding sequence in a particular host cell. Expression vectors arc exemplified by, but iioi limited to, plasimd, phagemid, shuttle vector, cosmid, virus, chromosome, mitochondrial DNA, plastid DNA, and nucleic acid fragments thereof. Nucleic acid sequences used for expression in prokaryotes include a promoter, optionally an operator sequence, a ribosome binding site and possibly other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals.

"Mammalian subject" includes human, non-human primate, murine, ovine, bovine, ruminant, lagomorph, porcine, caprine, equine, canine, felines, ave, etc.).

A subject "in need" of reducing one or more symptoms of a disease, and/or "in need for a particular treatment (such as immunization) against a disease includes a subject that exhibits and/or is at risk of exhibiting one or more symptoms of the disease. For Example, subjects may be at risk based on family history, genetic factors, environmental factors, etc. This term includes animal models of the disease. Thus, administering a composition (which reduces a disease and/or which reduces one or more symptoms of a disease) to a subject in need of reducing the disease and/or of reducing one or more symptoms of the disease includes prophylactic administration of the composition (i.e., before the disease and/or one or more symptoms of the disease are detectable) and/or therapeutic achninistration of the composition (i.e., after the disease and/or one or more symptoms of the disease are detectable). The invention's compositions and methods are also useful for a subject "at risk" for disease refers to a subject that is predisposed to contracting and/or expressing one or more symptoms of the disease. This predisposition may be genetic (e.g., a particular genetic tendency to expressing one or more symptoms of the disease, such as heritable disorders, etc.), or due to other factors {e.g., environmental conditions, exposures to detrimental compounds, including carcinogens, present in the environment, etc.). The term subject "at risk" includes subjects "suffering from disease," i.e., a subject that is experiencing one or more symptoms of the disease. It is not intended that the present invention be limited to any particular signs or symptoms. Thus, it is intended thai the present invention encompass subjects that are experiencing any range of disease, from sub-clinical symptoms to lull-blown disease, wherein the subject exhibits at least one of the indicia (e.g., signs and symptoms) associated with the disease.

"Immunogenically effective amount" refers to that amount of a molecule that elicits and/or increases production of an "immune response" (i.e., production of specific antibodies and/or induction of a cytotoxic T lymphocyte (CTL) response) in a host upon vaccination with the molecule.

"Antibody" refers to an immunoglobulin (e.g., IgG. IgM, IgA, IgE, IgD, etc. ) and/or portion ihereol ' lhal contains a "variable domain" (also referred to as the "Fv region") that specifically binding to an antigen.

The term "specifically binds" and "specific binding" when made m reference to the binding of antibody to a molecule (e.g., peptide) or binding of a cell (e.g.. T-cell) to a peptide, refer to an interaction of the antibody or cell with one or more epitopes on the molecule where the interaction is dependent upon the presence of a particular structure on the molecule. For example, if an antibody is specific for epitope "A" on the molecule, then the presence of a protein containing epitope A (or free, unlabeled A) in a reaction containing labeled "A" and the antibody will reduce the amount of labeled A bound to the antibody. In one embodiment, the level of binding of an antibody to a molecule is determined using the "IC50" i.e., "half maximal inhibitory concentration" that refer to the

concentration of a substance (e.g., inhibitor, antagonist, etc.) that produces a 50% inhibition of a given biological process, or a component of a process (e.g., an enzyme, antibody, cell, cell receptor, microorganism, etc.). It is commonly used as u measure of an antagonist substance's potency.

SUMMARY OF THE INVENTION

The invention provides a recombinant virus-like particle (VLP) comprising, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, b) NDV nucleocapsid (NP) protein, and c) one or more tumor-associated EBV antigen, wherein said one or more tumor-associated EBV antigen is inside said VLP. In one embodiment, said tumor-associated EBV antigen is selected from the group consisting of CBN A I , tEBN A I and LMP2. In a further embodiment, said tumor- associated EBV antigen comprises ttiBNA l and LMP2. In another embodiment, the VLP further comprises, in operable combination, one or more Epstein- Barr Virus (EBV) antigens, wherein at least one of said one or more antigens is selected from the group consisting of gB. gl I, and gL. In yet another embodiment, said VLP further comprises, in operable combination, EBV gp350/220. In one embodiment, said VLP further comprises, in operable combination, one or more NDV proteins. In a further embodiment, said one or more NDV proteins comprise NDV heptad repeat domain 2 (HR2) protein. In one embodiment, said one or more NDV proteins comprise NDV fusion (F) protein. In another embodiment, said one or more NDV proteins comprise NDV heamagglutinin-neuraminidase

(HN) protein. In yet another embodiment, the VLP further comprises, in operable combination, one or more human papillomavirus antigens. In yet another embodiment, said one or more human

papillomavirus antigens comprises one or more of LI and L2.

The invention also provides a vaccine comprising any one or more of the VLPs described herein, and a physiologically acceptable carrier.

Also provided by the invention is an expression vector encoding any one or more of the recombinant VLPs described herein.

The invention additionally provides a method for immunizing a mammalian subject against cancer, comprising administering an immunologically effective amount of one or more vaccine of Claim 12 to a mammalian subject in need thereof to produce a treated subject, wherein said administering is under conditions to produce an immune response to one or more tumor-associated EBV antigen. In one embodiment, said cancer comprises an Epstein-Barr Virus (EBV) -associated cancer. In another embodiment, said EBV- associated cancer comprises cervical cancer. In one embodiment, said immune response comprises T lymphocytes that specifically bind to said one or more tumor-associated EBV antigen. In a further embodiment, said immune response lacks antibody that specifically binds to said one or more tumor-associated EBV antigen. In yet another embodiment, said T lymphocytes are selected from CD4 + lymphocytes and CD8 + lymphocytes. In a further embodiment, said method further comprises administering a recombinant VLP that contains, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, and b) EBV gp350/220. In another embodiment, said method further comprises one or more of a) detecting said immune response to said one or more tumor-associated EBV antigen, and b) detecting a reduction in one or more symptoms of said cancer in said treated subject. In one particular embodiment, said administering is before manifestation of one or more symptoms of said cancer. In another embodiment, said administering is after manifestation of one or more symptoms of said cancer.

DESCRIPTION OF THE INVENTION

The present invention provides prophylactic and/or therapeutic vaccines that contain Newcastle disease virus (NDV) virus-like particles (VLPs) comprising one or more Epstein-Barr Virus (EBV) antigens. The invention's prophylactic and/or therapeutic vaccines are useful for preventing and/or treating infection with EBV and/or disease associated Epstein-Barr Virus, such as cancer.

In one embodiment, the invention relates to the use of virus-like particles (VLPs) as a multivalent vaccine for EBV, as well as the method of designing and producing these VLPs for this purpose. The VLPs stimulate both the production of nAbs (neutralizing antibodies) and EBV specific

T-cells.

In one embodiment, the present invention involves the development of a safe and feasible method for generating VLPs to be used as a vaccine that targets both nAbs and to stimulate a T-cell response to viral antigens. Specifically, this invention uses the NDV platform to produce a multivalent vaccine to EBV, a worldwide virus associated with AIM and several cancers. In one embodiment, this process utilizes FDA-approved CHO cells to generate VLPs, mimic EBV on the surface, but do not contain viral DNA, avoiding safety concerns about the oncogenetic properties of the virus. Whereas past attempts have focused on inducing nAbs to gp350/220, which is required but not necessary for viral entry into the cell, in one embodiment, the invention's multivalent vaccine includes gH/gL and gB, both of which are necessary for the fusion and subsequent entry of EBV into both epithelial and B- cells (19, 22). In addition to these surface proteins, the VLPs can be used as a vehicle for latent EBV antigens, EBNA1 and LMP2, which can be used to generate CD4+ T-cells that are specific for these antigens. A) Epstein-Barr virus fEBV) and current approaches

Epstein-Barr virus (EBV) is an oncogenic herpesvirus infecting over 95% of the adult population globally. It is implicated in the development of various types of lymphoproliferative diseases (LPDs) and carcinomas (1). Every year, EBV infection is estimated to be responsible for ~200,000 cancers globally (2). In low-income settings, primary EBV infection typically occurs during early childhood and is thought to be largely asymptomatic. However, in malaria endemic regions, childhood acquisition poses an increased risk of EBV positive Burkitt lymphoma (BL) (1, 3, 4). In high-income settings, primary EBV infection is often delayed until adolescence (4); and causes acute infectious mononucleosis (AIM) in 50-70% of adolescents (3). Although the disease is self-limiting, prolonged forms of AIM or chronic active EBV infection, may lead to fatal outcomes (5) or significantly increase the risk of developing EBV positive Hodgkin lymphoma (1). EBV is also highly associated with nasopharyngeal and gastric carcinomas, reflecting the epithelial tropism of the virus (3). Among infected individuals, EBV is controlled by T-cells and normally remains quiescent in memory B-cells (6). However, under conditions of immune suppression, the virus can reactivate, leading to an expansion of EBV-infected cells and increasing the likelihood of de novo infection, and transformation of infected B-cells as seen in BL, EBV positive post-transplant lymphoproliferative disorders (PTLDs), and AIDS-associated B-cell lymphomas (7-9). Management of EBV-associated diseases is problematic due to difficulties with diagnosis, surveillance, and treatment. In a meeting convened at the National Institutes of Health (NIH) in 201 1 , participants agreed that the need for a safe and effective vaccine to prevent and/or treat EBV-associated diseases is urgent (2). Several strategies to generate an EBV vaccine based on viral glycoprotein 350/220 (gp350/220), latent membrane proteins (LMP1 -2), and EBV nuclear antigen 1 (EBNA-1 ) are currently in experimental stages and/or clinical trials (10, 11). However, most of these strategics have low safety profiles, and are designed to elicit the production of neutralizing antibodies (nAbs) to EBV envelope proteins (prophylactic), or a T- cell response to latent EBV antigens (therapeutic). None of the current proposed vaccines address both arms of immunity in a single candidate vaccine. Our vaccine utilizes a VLP platform which incorporate select multiple viral surface glycoproteins in addition to intracellular T-cell antigens to generate a polyvalent vaccine. These VLPs have high safety profiles and are efficiently produced in the FDA-approved Chinese hamster ovary (CHO) cell line.

Antibodies (Abs) provide the first line of defense against virus infection. Neutralizing Abs directed to EBV envelope glycoproteins are present in humans, maternal nAbs prevent neonatal infection, and it has been shown that they are induced in response to immunization both in humans and in other animals (12-14). However, persistent EBV infection and the limited evidence of immune selection of viral antigenic variants indicate that in vivo neutralization of EBV infection is suboptimal. This was observed in four independent phase I/II clinical trials, in which vaccination with either vector constructs expressing gp350/220, or with purified recombinant non-splicing variant gp350 soluble protein, did not prevent infection although incidence of AIM was reduced in young adults by more than 70% (14-17). Importantly, primary B-cells can be infected with recombinant EBV lacking gp350/220; suggesting that additional viral ligands may be mediating EBV infection in the absence of gp350/220 (18-22). These observations indicate that using gp350/220 as the only immunogen (monovalent vaccine) to target viral neutralization is too simplistic and may account for the variable success in using this protein in EBV vaccine development.

In EBV infection, EBVgp350/220, the attachment protein, binds to B-cell receptors CD21 and

CD35, initiating the first contact of the virus and the host cells (23-26), and subsequently triggering endocytosis of the virions (27). This interaction enhances infection, but is not essential (18). Fusion between the viral envelope and the cellular membrane is a required step in the entry of all human herpesviruses (28). For EBV, the viral glycoproteins necessary for fusion of the viral envelope with the host cell receptors are glycoprotein B (gB) (22), the complex of gH and gL (gH/gL), and gp42 (19).

These complexes mediate infection and confer host cell specificity. EBV entry into B-cells is mediated by gB, gH/gL, and gp42; whereas entry into epithelial cells is facilitated by interaction between gB and gH/gL (22, 29, 30). It is important to note that co-expression of EBVgH and gL is required for transport of gH to the cell surface which results in the formation of a stable complex of gH/gL (29).

Recently, integrins have been identified as the epithelial receptors for EBVgH/gL and this interaction initiates fusion in a two-step cascade (20). Recombinant EBV lacking gH does not infect either epithelial or primary B-cells (31).

Although Abs to EBVgH/gL are not robustly produced in vivo during natural infection (perhaps due to masking by the immunodominant gp350/220), immunization of mice with recombinant gH can boost irnmunogenicity and generate Abs capable of blocking EBV infection (20, 21, 32). The ability of gH/gL Abs to neutralize infection is also well-conserved in herpes simplex virus- 1 (33, 34), cytomegalovirus (35, 36) and Kaposi sarcoma herpesvirus (KSHV) (37). Monoclonal Abs to the gH protein or the gH/gL complex block EBV infection, indicating a critical role for gH/gL in EBV infection (38, 39). No specific nAbs to EBVgL or -gB have been reported so far (10). NAbs directed to EBVgp42, have been identified (40).

T-cell-mediated responses are effective in controlling persistent EBV infection, as evidenced by some form of immunosuppression usually preceding EBV-associated lymphomas and PTLDs (1, 41). Furthermore, adoptive transfer of EBV-specific T-cells can induce remission in transplant patients (42, 43).

The current hypothesis is that protection against EBV relies on inducing CD4 + and CD8 + T-cell immune responses, and the development of EBV therapeutic vaccine candidates have focused on enhancing such responses (44). EBNAl-specific CD4 + and CD8 + T-cells are frequently detected in EBV-infected individuals (45, 46), and both T-cell subsets can be effective in controlling growth of EBV-immortalized B-cells (47). Notably, EBNA1, LMP2, and EBVgp350/220 antigens have been developed and independently tested in various clinical trials as vaccine candidates against EBV infection and EBV* cells with promising results (17, 48, 49). Recent phase I clinical trials of recombinant modified vaccinia Ankara vector encoding deletion of Gly-Ala regions from the EBNA1 sequence (known to impair presentation of cu-linked sequences) fused to LMP2 as a vaccine candidate elicited a robust EBV-specific CD4 + and CD8 + T-cell response in humans (44, 48, 50). However, the strategy used to deliver these two important EBV antigens (a DNA vaccine), known for their oncogenic potential, may pose major health risks, such as inducing an antibody response to DNA or integration of DNA in an undesired location in the host genome causing unchecked cell growth, particularly in immunosuppressed individuals. Furthermore, these vaccine candidates cannot elicit nAbs to eliminate reactivated or new EBV infections. There is also a risk of vaccine tolerance since the quantity of proteins produced and secreted in vivo is unregulated. EBV DNA packaging mutants (51 ), and disabled virions that lack the major oncoprotiens have also been proposed as an alternative strategy (52).

However, incomplete knowledge of all the virion proteins functions, concerns about inadvertent association of oncogenic DNA/RNA fragments with the assembled VLPs, limited production and release of the virus, as well as current requirements for their propagation in tranformed human cell lines suggest that such strategies may not meet the stringent FDA safety guidelines. Our strategy will address all these limitations by combining select multiple EBV antigens in a VLP platform which is efficiently produced in CHO cells, immunogenic, and safe to use in all populations, irrespective of immune status.

B) Compositions and methods for treating cancer

The invention provides prophylactic and/or therapeutic vaccines that contain Newcastle disease virus (NDV) virus-like particles (VLPs) comprising one or more tumor-associated Epstein-Barr Virus (EBV) antigens.

In one embodiment, the invention's VLPs induce both nAbs and protective human T-cell responses will not only be an invaluable candidate vaccine in preventing EBV infection, but also of utmost importance in preventing BL in African children, acute infectious mononucleosis in 50-70% of US adolescents, nasopharyngeal carcinoma (endemic in Southeast Asia), post-transplant

lymphoproliferative disorders, and non-Hodgkin lymphoma (in iatrogenic and AlDS-immunodeficient patients). The Newcastle virus disease vaccine platform is also invaluable for the development of candidate prophylactic and therapeutic vaccines against other human viruses, as it provides a safe and effective method of targeting both arms of the immune system.

In one embodiment the invention provides a recombinant virus-like particle (VLP) comprising, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, b) NDV nucleocapsid (NP) protein, and c) one or more tumor-associated EBV antigen, wherein the one or more tumor- associated EBV antigen is inside the VLP. In one embodiment, the tumor-associated EBV antigen is selected from the group consisting of EBNA1, tEBNAl and LMP2. In one embodiment, the tumor- associated EBV antigen comprises tEBNAl and LMF2. In one embodiment, the VLP further comprises, in operable combination, one or more Epstein-Barr Virus (EBV) antigens, wherein at least one of the one or more antigens is selected from the group consisting of gB, gH, and gL. In one embodiment, the VLP further comprises, in operable combination, EBV gp350/220. In one

embodiment, the VLP further comprises, in operable combination, one or more NDV proteins, exemplified by, NDV HR2 protein, NDV F protein, and NDV HN protein. In one embodiment, the VLP, further comprises, in operable combination, one or more human papillomavirus antigens. In one embodiment, the one or more human papillomavirus antigens comprises one or more of LI and L2.

The invention also provides a vaccine comprising one or more of any of the VLPs described herein, and a physiologically acceptable carrier.

The invention also provides an expression vector encoding any one or more of the recombinant VLPs described herein.

The invention also provides a method for immunizing a mammalian subject against cancer, comprising administering an immunologically effective amount of any one or more of the vaccines described herein to a mammalian subject in need thereof to produce a treated subject, wherein the adrmnistering is under conditions to produce an immune response to one or more tumor-associated EBV antigen. In one embodiment, the cancer comprises an Epstein-Barr Virus (EBV) -associated cancer. In one embodiment, the EBV-associated cancer comprises cervical cancer. In one

embodiment, the immune response comprises T lymphocytes that specifically bind to the one or more tumor-associated EBV antigen. In one embodiment, the immune response lacks antibody that specifically binds to the one or more tumor-associated EBV antigen. In one embodiment, the T lymphocytes are selected from CD4 + lymphocytes and CD8 + lymphocytes. In one embodiment, the method further comprises administering a recombinant VLP that contains, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, and b) EBV gp350/220. In one embodiment, the method further comprises one or more of a) detecting the immune response to the one or more tumor- associated EBV antigen, and b) detecting a reduction in one or more symptoms of the cancer in the treated subject. In one embodiment, administering the vaccine is prophylactic before manifestation of one or more symptoms of the cancer. In one embodiment, administering the vaccine is therapeutic after manifestation of one or more symptoms of the cancer. In one embodiment, the vaccine prevents cancer and/or the spread of cancer. C) Compositions and methods for immunizing against Epstein-Barr Virus (EBV)

The invention provides prophylactic and/or therapeutic vaccines that contain Newcastle disease virus (NDV) virus-like particles (VLPs) comprising one or more Epstein-Barr Virus (EBV) antigens. The invention provides evidence that viral antigens capable of eliciting T cells response can be incorporated into the virus-like particle by fusing NDV-nucleoprotein.

Approaches to EBV vaccine development are limited due in part to the oncogenic potential of the EBV genome and a lack of animal models for testing vaccine candidates. VLPs are safe, immunogenic and have been successfully used to prevent infections against other oncogenic viruses such as human papillomavirus and hepatitis B virus. The EBV envelope glycoprotein gp350/220 (EBVgp35G/220) has been proposed as a potential vaccine antigen. However, in four independent phase I/II clinical trials, vaccination with EBVgp350/220 did not prevent EBV infection. Importantly, recombinant EBV lacking gp350/220 can infect both epithelial and primary B cells in vitro. These preliminary studies demonstrate that using pooled sera from mice immunized four times with EBVgp350/220 VLPs without adjuvants neutralized EBV infection in vitro, resulting in 46% inhibition. Whereas neutralization with sera from mice immunized with UV-inactivated EBV resulted in -88% inhibition. This suggests that other envelope glycoproteins may have a role in virus entry, independent of gp350/220. The EBVgH/gL envelope glycoprotein complex is definitively required for EBV fusion and entry, and thus is a potential target for developing a prophylactic vaccine. Antibodies to the EBVgH/gL complex can neutralize virus infection, and recombinant EBV lacking gH cannot infect both epithelial and primary B cells. To our knowledge, these proteins have not yet been tested as part of candidate vaccines in any animal model or clinical trial.

Data herein provides evidence that generation of VLPs containing both EBVgH/gL and EBV EBNA1 and/or LMP2 consistently expressed in most EBV positive tumors is feasible. Recent phase I clinical trials of recombinant modified vaccinia Ankara vector encoding deletion of Gly-Ala regions from the EBNA1 sequence fused to LMP2 as a vaccine candidate elicited a robust EBV-specific CD4 + and CD8 + T-cell response. Because EBV is a human pathogen, the process described takes advantage of both human peripheral blood mononuclear cells (PBMCs) and immunodeficient mice reconstituted with human immune system components (humanized BLT model, bone marrow/liver/thymus) as models to test EBN A 1 -specific CD4 + and CD8 + T-cell responses.

Current approaches to EBV vaccine development are limited due in part to the oncogenic potential of the EBV genome (Figure 49). To generate a potent EBV vaccine candidate we included multiple vaccine antigens including tumor-associated antigens in our VLPs (exemplified in Figures 1 -

3)·

Despite strong evidence that antibodies (Abs) to gH/gL can neutralize EBV infection, these proteins have not been used to generate an EBV vaccine. In one embodiment, the invention's VLP vaccines contain two tumor-associated antigens (EBNA1 and LMP2); EBNA1 is consistently expressed in B cells of all BL patients and recognized by CD4+T cells and LMP2 is primarily targeted by CD8+ T cells.

Data herein demonstrate that VLPs containing a functionally inactive truncated EBNA1 (tEBN A 1, lacking the Gly-Ala rich domain known to impair presentation of cis-linked sequences), LMF2, and gH/gL, stimulate robust humoral and cellular responses against EBV. Thus, in one embodiment, the invention provides NDV VLPs containing tEBNA I, LMP2, gB and gH/gL for use as a vaccine to induce neutralizing antibodies (nAbs) and/or EBV specific CD4 + and CD8 + T cells responses. In a particular, embodiment, the invention provides EBVgH/gL:tEBNA I -LMP2 VLPs that can be used alone or together with EBVgp350/220 VLPs to prevent EBV infection and its associated diseases. In another embodiment, the invention's vaccines are therapeutic and/or prophylactic vaccines against infectious diseases and cancers, such as cervical cancer.

In one embodiment, the invention provides a recombinant virus-like particle (VLP) comprising, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, and b) two or more Epstein-Barr Virus (EBV) antigens, wherein at least one of the two or more antigens is selected from the group consisting of gB, gH, and gL. In one embodiment, the two or more EBV antigens comprise a tumor-associated EBV antigen, wherein the tumor-associated EBV antigen is exemplified by, but not limited to, EBNA1 and LMP2. In a particular embodiment, EBNA1 is truncated (tEBNAI, i.e., EBNA1 that lacks the Gly-Ala rich domain). In a further embodiment the VLP comprises EBV gp350/220. In a particular embodiment, the VLP further comprises, in operable combination, one or more NDV proteins, exemplified by, but not limited to NDV nucleocapsid (NP) protein, NDV heptad repeat domain 2 (HR2) protein, NDV fusion (F) protein, and NDV heamagglutinin-neuraminidase (HN) protein.

The invention further provides a VLP that contains, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, and b) EBV gp350/220 (Ogembo et al., "5A chimeric EBV gp350/220-based VLP replicates the virion B-cell attachment mechanism and elicits long-lasting neutralizing antibodies in mice," Journal of Translational Medicine (2015) 13:50). In a particular embodiment, the VLP further comprises, in operable combination, one or more NDV proteins, exemplified, by, but not limited to NDV nuclcocapsid (NP) protein. NDV heptad repeal domain 2

(IIR2) protein, NDV fusion (Γ) protein, and NDV heamagglutinin-neuraminidase (UN) protein.

The invention further provides a vaccine comprising any one or more of the VLPs described herein, and a physiologically acceptable carrier.

Also provided is an expression vector encoding any one or more of the recombinant Vl.Ps described herein.

The invention also provides a method lor immunizing a mammalian subject against Epslein- Barr Virus (EBV), comprising administering an immunologically effective amount of one or more of the VLP vaccines described herein to a mammalian subject in need thereof to produce a treated subject, wherein the administering step is under conditions to produce an immune response to one or more EBV antigen. In one embodiment, the method comprises administering (A) a first recombinant VLP comprising, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, and b) two or more Epstein- Barr Virus (HBV) antigens, wherein at least one of the two or more antigens is selected from the group consisting of gB, gH, and gL, and (B) a second recombinant VLP that contains, in operable combination, a) Newcastle disease virus (NDV) matrix (M) protein, and b) EBV gp350/220. In one preferred embodiment, administering the vaccine docs not produce EBV-associaled cancer. In another embodiment, the method further comprises one or more of a) detecting the immune response to the one or more EBV antigen, b) delecting a reduction in one or more symptoms of EBV infection in the treated subject, and c) determining the presence and/or absence of EBV-associatcd cancer in the treated subject. In one embodiment, the invention's vaccination method is used prophylactically by administering the vaccine before manifestation of one or more symptoms of infection of the subject with the EBV. In a further embodiment, the invention's vaccination method is used therapeutically by administering the vaccine alter manifestation of one or more symptoms of infection of the subject with the EBV. In a particular embodiment, the immune response by the treated subject comprises antibody that specifically binds to the one or more EBV antigen. In a particular embodiment, immune response comprises T lymphocytes that specifically bind to the one or more EBV antigen. In a further embodiment, the T lymphocytes arc selected from CD4 lymphocytes and CDS ' lymphocytes. EXPERIMENTAL

The following examples serve to illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof. EXAMPLE 1

Materials And Methods. The following is a brief description of the exemplary materials and methods used in the subsequent Examples.

Virus and Cell lines

B95-8 strain harboring FBfaV-GFP was obtained from Dr. Richard Longnecker (Northwestern University, Chicago). EGFP-EBV was prepared as described (53). Purified Kaposi's sarcoma herpesvirus (KSHV) and EBV-EGFP from the AGS cell line were generous gifts from Dr. Christine King (SUNY Upstate* Medical School) and Dr. Liisa Selin (University of Massachusetts Medical School), respectively.

Human embryonic kidney I IEK-293A cell line, a cloned derivative of 293 was purchased from Life Science Technologies. HEK-293T, a 293 derivative expressing SV-40 T antigen, CHO, Vcro (African green monkey kidney). ELL-0 (chicken embryo), KS62 (human crythrolcukcmia), Raji (EBV+ Burkitt's lymphoma), HB168 (72A1 murine hybridoma) were all purchased from the American Type Culture Collection. Cell lines 293A. CHO, and ELL-0 were cultured in Dubclco's Modified Eagle's medium (DMEM). B95-8, K562, HB168 and Raji cell line were grown in RPMI media. All media contained 1% L-glutamine, 10% heat-inactivated fetal bovine serum (FBS) and 2% penicillin- streptomycin unless otherwise specified.

Antibodies

Primary monoclonal antibodies (mAb)-72A 1 and -2LI 0 anti-gp350/220 were purchased from EMD Millipore. Polyclonal rabbit anti-Newcastle disease virus (NDV) and anti-IIR2 have been previously described (54). MAb anti-CD35 (clone El l ) and anli-CD21 (clone LT21 ) were purchased from BioLegend. mAb EBNA1 (clone IH4-1) was a gift of Dr. F. Grasser, Institut fur Virologie. Germany. Both goat polyclonal anti-EBNA l Gly-Arg regions and rat monoclonal anti-EBV LMP2A (clone 14137) were purchased from Abeam. Monoclonal anti-EGFP (clone GSN 149) was purchased from Sigma. All the antibodies to EBVgH, EBVgH/gL and EBVgB were gifts of Dr. Lindscy Hutt- Flctchcr (Louisiana State University Health Sciences Center, Shrcvcport, LA). mAb anti EBVgH/gL (clone CL59) recognizes gl l alone and binds to an epitope between residues 501 -628 which is part of the C-terminal flap structure in domain IV: E1D1 recognizes only gH/gL complex and partially blocks binding to an integrin so probably binds somewhere on the domain I/domain II interlace. CL40 recognizes gHL/gL complex. mAb anti-gB (clone CLS5) recognizes the ectodomain of the glycoprotein, while rabbit polyclonal anti-gB (clone BA23) recognizes C-terminal domain.

Secondary: Horseradish peroxidase (HRP)-conjugatcd goat anti-mouse IgG (total and isotypc specific) antibodies, goat anti-rabbit or goat anti-rat antibodies for immunoblot or ELISA were purchased from Sigma. Goat F(ab ' )2 anti-mouse IgG (H+l .) AF4X8 or AF594 was used for cytometric and confocal analyses (Invitrogcn). Goat anti-mouse IgG (I H L) immunogold was used for electron microscopy (EM) (Aurion).

Plasmid vectors

pCAGGS-EBVgp350/220-F or pCAGGS-EBVgp350/22u-iiR2 F

Full length EBVgp35(V220 (BLLF1) from the BamHI I . fragment of EBV genome strain B95-8 (55) was amplified by PCR and cloned into pCAGGS as described (56). A chimeric fragment consisting of amino acids 1 -864 encoding the gp350/220 cctodomain (ED) fused to the amino acids 466-553 of the NDV-F heptad repeat 2 (HR2). transmembrane (TM) and cytoplasmic (CT) domains was constructed by three-way ligation to obtain EBVgp350/220-F as described (56). Full length chimeric EBVgp350/220-F lacking the heptad region of the NDV TM region was synthesized by Genewiz, cloned into pCAGGS vector (pCAGGS-EBVgp350/220-HR2 F) and sequence fidelity verified. A lull-length gp350/220 wild type (WT) were also synthesized and cloned into pCAGGS vector to be used as a control in various experiments. pCAGGS-F, pCAGGS-M and pCAGGS-NP derived from NDV (Avulavirus) have been described (57). pCAGGS EBVgH/gL (gp85/gp25)

To construct chimeric EBVgll-F and EBVgL-IIN proteins, sequences encoding the TM domain lacking the heptad region and the cytoplasmic tail of the NDV F protein (amino acids 499-553) or hemagglutinin -neiiraminidase (HN) protein (amino acids 1-46) were fused to the sequence encoding the ectodomain of gH, amino acids 1-679 and to amino acids 25-137 of gL, respectively. These chimera proteins were synthesized and cloned individually into the pCAGGS vector. cDNAs of full length gH WT (amino acids 1 -707) and gL WT (amino acids 1 -137) were also synthesized and individually cloned into pCAGGS vector as controls. pCAGGS-EBVgB-F (gpl 10) A chimeric fragment consisting of amino acids 1-736 encoding the EBVgB cctodomain fused to the NDV-F lacking IIR2 region of the TM and CT domains (amino acids 466-553) was synthesized. The synthesized sequence was cloned into pCAGGS vector and sequence fidelity verified. A full- length EBVgB WT (amino acids I -790) was also synthesized and cloned into pCAGGS vector as a control.

Generation of fusion protein between NP and EGKP, truncated ΕΒΝA1 or LIVIP2.

Fusion proteins between NP (amino acids 1-489) and EGFP (amino acids 1-241), truncated EBNA 1 encoding deletion of Gly-Ala regions from the EBNA1 sequence known to impair presentation of c ls-linked sequences (ammo acids 326-641), or full length LMP2 (ammo acids 1-497) were synthesized by Genewiz and fidelity of the sequences verified. The specific fusion sequences were individually cloned into pCAGGS vector. Full length EGFP, EBNA1 and LMP2 were also synthesized and cloned into pCAGGS vector as controls. Transfection, Generation and Purification of EBVgp350/220-F, EBVgp350/220-EGFP, gp350/220-EBNA1, EBVgH/gL, E BVgH/gL-EBN A 1 and EBVgB-LMP2 VLPs.

To determine surface expression of the EBV glycoproteins, 1.0μg/well of pCAGGS, pCAGGS- gp350/220 WT, pCAGGS-EBVgp350/220-F, pC'AGGS-EBVgp350/220-HR2 F, pC AGGS-EBVgH WT. pCAGGS-EBVgH-K, pCAGGS- EBVgL WT, pCAGGS-EBVgL-HN or pCAGGS-EGFP were individually transfected into 80% subconfluent CHO cells seeded in six-well tissue culture plates using Lipofcctaminc and Reagent Plus (Life Sciences Technologies) according to the manufacturer's direction. Both gH-F/gL-HN or gH WT/gL WT plasmids were also transfected into CHO cells to assess the formation of the gll/gL complex. Cells were harvested at 48h posi-transieciion to assess surface expression of individual protein by cylmelry after staining with specific primary antibody followed by secondary antibody. In certain cases, surface expression of individual proteins were also assessed transfected 293A, Vero or ELL-0 cell lines. Cytometric analysis was performed on a LSRII benchtop FC (Bccton-Dickinson, B-D) and data analyzed using CcllQucst Pro Version 4.0.1 (B-D) and'or FlowJo Cytometry Analysis software (Tree Star Inc) as described (56).

For VLP preparation, equal amounts 8μg/plasmid) of pCAGGS-NDV M, NP and pCAGGS- EBVgp350/220-F plasmids were co-transfcctcd into cells seeded in T- 175cm" flasks. DNA- Lipolcctaminc complexes were incubated at 37"C for 5h with 293T and ELL-0 or overnight with CHO cells. Complexes were removed by washing and 20ml of complete media with or without 4mM of sodium butyraic (Millipore ED) and 20ng'ml of TP A ( 12-O-tetradecanonylphorbol 13-acciaic) (Sigma) were added. VLPs were isolated by sucrose gradient purification as described (58). Similar strategy was used to generate EBVgp350/220-HR2 F. EBVgp350/220-HR2 F-EGFP, EBVgp350/220-HR2 F-

EBNA I , EBVgH/gL-EBN A 1 and EBVgB-LMP2 VLPs in CHO cells.

Silver stain, Immunoblot Analysis, Electron Microscopy and Immunogold Staining

Purified EBV, VLPs or cells were lysed in RIPA buffer (Boston Bioproducls) containing complete protease inhibitor cocktail (Roche Applied Science). Cell lysates were incubated on ice lbr 15 min. and then centrifuged for 5 min at 14,000 x rpm in microcentrifuge. The protein content of lysates was determined by Bradford assay using Coomassie Brilliant Blue (Sigma). Lysates were boiled for 5 min in I .Laemmli SDS-sample buffer (Boston Bioproducls) under non-reducing or reducing conditions. A known quantity of protein lysatc was loaded onto a 4-12% polyacrylamidc gel for protein separation. Protein was detected by Pierce's silver stain kit following manufacturer's recommendation. For immunoblot analyses, separated proteins were transferred to a PVDF membrane (Life Sciences Tech.) using iblot (Life Sciences Tech.). Membranes were pre-incubated with 5% non- iat dry milk (LabScicntiilc) lbr 30 min and delected with specific antibodies as previously outlined (26 f 59).

Purified VLPs and viruses were analyzed by EM as described (60). Briefly, particles were dialyzed against 2 L of I X TNE ( lOOmM Tris; 2.0M NaCI; l OmM EDTA; pH 7.4) to remove residual sucrose. Purified KSHV, KBV and VI P gp350/22( ) /HR2 F were incubated with 3% BSA in TNK for 45 min and embedded on the grid. Primary antibody (l Oug/ml) was diluted in 1 % BSA/TNE and adsorbed to the grid for I hour at room temperature. Following three washes with I x TNE, secondary gold- conjugated antibody was added for 1 hour at room temperature (R.T). The grids were washed twice with TNE, and negatively stained with 12% phosphotungstic acid (pH 7) lbr 15 sec; air dried lbr 30 min and examined using a Tecnai transmission electron microscope.

Confocal Microscopy, Cell Binding Assays

Cells were washed three times with 1 x phosphate buffer saline (PBS) and confocal microscopy performed as previously outlined (26). Briefly, I x 10 6 cells were seeded onto Labtck slides and incubated at RT for I hour with standard amounts of VLP predetermined based on silver stain and/or Bradford assay quantification of total protein. Mixture of cells and VLPs were stained with mAb Alexa-fluor (AF) 594 anti-CD21 or mAb AF488 anli-CD35 for 30 min on ice. Nuclei were stained with DAPI 33342 (Sigma) for 5 min at RT. Stained cells were washed three times, mounted (Mounting

Medium, DakoCytomaiion) and imaged using an UPlanApo 60x1.42 NA objective on an Olympus

BX62 microscope fitted with a cooled Hamamatsu Orca AG CCD camera. The microscope, filters, and camera were controlled as outlined (61 ). The dcconvolution process is described (26).

Immunization, Enzyme-linked Immunosorbent Assay (ELISA) for Antibodies and I FN- γ

Sera from terminal bleed of mice immunized with F.BVgB-LMP2 VLP, RBVgH/gL VLP, EBVgp350/220-IIR2 F VLP, EBVgp350/220-IIR2 F VLF, or UV-inaclivated EBV were used to determine IgG titers measured by ELISA (62). Soluble gp35()/22() ED or lysatcs from ELL-0 cells transfected with gB or gH/gL were used as target antigens. Briefly, 96-wcll microtiter plates (Nunc- Imimino Plate Maxisorp) were coated with 5()ng/well of recombinant gp350/220 HI). gB or gH/gl. in a carbonate buffer (pi I 6.2) at 4 o C overnight and blocked with 1% BSA. Serially diluted sera in PBS was added for 2 h at RT and washed. Antibody binding was detected with HRP-labclcd goat anti- mouse IgG, secondary antibodies at RT for 1 h. Plates were washed 5x and the substrate letramethylbenzidine (Life Science Technologies) was added. Reactions were stopped with 2M sulfuric acid. To determine antibody liter, optical density was read at 450nm with an ELISA reader (Spectramax® Plus 384, Molecular Devices). The highest antibody dilution yielding an OD 450 2x higher than that of TNE-treated mice was designated the endpoint liter. Anti-gp350/220 mAbs served as positive control for gp350/220.

Peptides corresponding to the immunization components, as well as control peptides were used to stimulate 5 x 10 5 splcnocytcs in. Synthetic peptides derived from

LMP2 I CLGGUXMV) or Promix EBV peptide pool consisting of 26 peptides, each corresponding to a defined HLA class I-restricted T cell epitope from EBV were used at concentration of μg/ml in the assay. After overnight culturing, the supernatants were tested for IFN -γ release by ELISA. SI1NFEKL ovalbumin was used as a negative control and concavalin A and IL1B were used as model antigens.

Serum EBV-neutralization Studies and Statistical Analysis

Terminal bleed sera from mice serially immunized with UV-EBV, soluble gp350/220 ED, TNE, EBVgp35()/22()-F and EBVgp350/220-HR2 VLPs were heat-inactivated at 56°C to remove complement. Sera were prc-incubatcd 1 : 1 with purified EGFP-EBV at different concentrations (typically a 1:5 virus dilution from a frozen stock) for 1 h at RT before infecting 10 5 Raji or 293 cell lines seeded in 24 -well tissue culture plates as previously outlined (53, 56, 63). Anti-gp350/220 mAb- 72A1 (neutralizing). mAb-2L10 (non-neutralizing) and sera from TNE-trealed mice (vehicle) served as controls. Experiments were repeated at least 3 times. Plates were incubated at 37°C for 3 days and visualized daily to enumerate GFP cells. Cytometry was performed on day 3 tor end-point analysis.

Neutralization data were analyzed using Graph Pad Prism 6 Software (GraphPad Software, Inc., San Diego) as described (56).

EXAMPLE 2

Construction and purification of Epstein-Barr virus-like particle (EBV-VLP) gp350/220 using Newcastle Disease virus platform as a prophylactic vaccine candidate against EBV infection. Sec Figures 4-11.

EXAMPLE 3

Development of a novel gH/gL VLPs. Sec Figures 12- 16. EXAMPLE 4

Development of a novel gH/gL-EBNAl-LMJP2 VLPs. Sec Figures 17-29.

EXAMPLE S

NDV [NP as a Carrier for EBNA1-LMP2 in EBVgp350/220 or gH/gL VLPs. Sec Figures 30-34.

EXAMPLE 6

Generating N P-tE ΒΝΛ I -L.M P2 Chimera Protein. Sec Figures 35-37.

EXAMPLE 7

NP-EGFP Blots. See Figures 38-47.

EXAMPLE 8

Generating VLPS containing NP-tEBNAl Chimera Protein and containing N P-tE ΒΝΛ 1 -LM P2 Chimera Protein

Sec Figures 50-53 and 62-71.

EXAMPLE 9 Detection of chimeric EBV-gp350/220-F, EBVgH-F/gL-HN and EBVgB-F on the surface of mammalian and avian cell lines

In this study, wc generated novel VLPs known for their safety, adjuvanicity and efficacy; to present select EBV glycoproteins (gp350/220, gH/gL or gB) and/or latent genes expressed in EBV positive tumors (ΗΒΝΛ Ι and/or I.MP2) to elicit neutralizing antibodies and T-cell responses, respectively. These VLPs arc devoid of EBV genomic DNA and arc efficiently produced in CI 10 cell line; an FDA approved vehicle. Newcastle disease VLP-based platform has been shown to efficiently assemble from three or four virion proteins, the envelope glycoproteins, F (fusion, a type 1 glycoprotein) and/or HN (hcmaglutinin-ncuraminidasc, a type 2 glycoprotein), together with core proteins M (matrix) and N'P (nucleocapsid protein) that drive assembly and release as described (58, 64); patent issued to Morrison et al., U.S. Pat. No. 7,951 ,3X4 issued to Morrison et al. on May 01 , 201 1 ; U.S. Pat. No. 8.974,797, issued to Morrison on Mar- 10-2015, each of which is incorporated by reference and sec also Figure 72. Further novel methods for producing NDV-bascd VLPs arc disclosed herein.

To generate EBVgp350/220 bearing VLPs; we first constructed an EBVgp350'220 F,

EBVgp350/220-HR2 F, chimeric plasmids ( Figure 73). To assess whether the expressed chimera proteins appropriately localized lo the plasma membrane, pCAGGS-EBVgp350/220-F, pCAGGS- LBVgp350/220-HR2 F and pC"AGGS-EBVgp350/220 WT plasmids were individually transfected into CHO, ELL-0 and 293A cell lines from three different species. Membrane expression of the gp350/220 was analyzed by cytometry. MAb-72A 1 directed to the N-terminal attachment protein binding epitope of gp350/220 (65) detected expression of both gp350/ ' 220 WT and chimeric EBVgp350/220 F proteins at the surface of all relevant transfectants ( Figure 74).

EXAMPLE 10

Assembly and characterization of chimeric EBVgp350/220 F VLPs

Following confirmation of plasma membrane expression, as required for particle assembly, pCAGGS-EBVgp350/220-F was co-transfcctcd with NDV core proteins M, NP into CHO. 293T or ELL-0 cell lines to generate chimeric VLPs as diagram in Figure 75A. Particles from distinct preparations released into cell supernatants were purified, characterized by immunoblot (Figure 75A, 75C; electron microscopy (Figure 75B), and silver stain (Figure 75D) as outlined (56). These analyses confirmed thai proteins of correct sizes (350 and 220 kDa) were made and these VLPs arc similar in size, shape and structure to the native virus, by negative staining and immunogold- conjugated antibody analysis. EXAMPLE 11

Visualization of EBVgp350/220-F VLP attachment to CD21 and CD35 bearing cells

EBVgp350/220 binds CD21 and/or CD35 on human cells (23, 26). To determine whether chimeric VLPs expressed from CHO cells retained the receptor-binding specificity of the virion envelope protein, we incubated EBVgp350/220-F VLPs with Raji, a latently EBV-infected B-cell line that naturally expresses high amounts of CD21 (23) and can be superinfected with EBV. In addition, a panel of receptor negative cell lines, Nalm6 and K.562 (not shown), together with their CD21 or CD3S transfected sublines was investigated (26). Receptors (red) and VLPs (green) were visualized by indirect immunofluorescence using a confocal microscope for detection as shown in Figure 76. EBVgp350/220 F VLPs abundantly bound Raji, no attachment to Nalm6 was detected. Nalm6CD21, Nalm6CD35 K562CD21 and K562CD35 all bound EBVgp350/220-F VLPs. EXAMPLE 12

Development of specific IgG responses to EBVgp350/220 in BALB/c mice immunized with EBVgp350Y220-F VLPs

To determine whether chimeric VLPs elicit EBVgp350/220 specific antibody responses, a group of five mice were immunized intraperitoneally with 10^g of EBVgp350/220-F VLP derived from CHO cells. UV-EBV and soluble gp350/220 ED served as positive controls and TNE as vehicle/negative control. Equivalence of gp350/220 protein content among the different immunogens was determined by both silver stain and Bradford assay (not shown). All animals received booster immunizations on days 43, 172, 183 and 218. Sera were collected two weeks post-boost. None of the animals displayed signs of local or systemic inflammation or changes in feeding or body weight that would indicate toxicity. Soluble recombinant gp350/220 ED served as the target antigen in an IgG ELISA. Anti-gp350/220 specific total IgG antibody titers significantly increased among mice immunized with the chimeric VLP, UV-EBV and soluble recombinant gp350/220 ED compared with pre-vaccination and control titers (Figure 77). Historical controls using NDV-F VLPs as immunogen were non-reactive in gp350/220-based ELISAs (not shown). The increase in EBV-gp350/220 specific antibody appeared to plateau on day 84 after the initial boost, but then further increased after the second and third boosts. There was a significant difference in antibody titers of mice immunized with UV-EBV, compared with soluble gp350/220 ED and EBVgp350/220-F VLP, although the slopes of the response curves were similar. Gp350/220 specific antibody was absent from TNE-immunized mice. All gp350/220-based immunogens produced long-term gp350/220-specific responses, though mice immunized with native EBV maintained significantly higher titers of gp350/220 antibody compared to mice immunized with VLP or soluble recombinant protein.

EXAMPLE 12

Anti EBV-gp350/220 antibodies generated following VLP immunization neutralize EBV infection in vitro.

It is well known that titer is not the sole gauge of a protective antibody response, as certain Immunogens can induce antibodies that promote, rather than block infection and high affinity blocking antibodies can be highly effective at low titers (66). To evaluate the protective efficacy of antibodies generated in response to chimeric EBV -VLP, UV-EBV and soluble recombinant gp350/220 ED, we assessed the in vitro neutralizing antibody titers of sera boosted four times (collected day 228) with the immunogens described above. Because EBV docs not plaque and large virus quantities arc difficult to obtain, EBV was titered by the Raji cell infection assay (Figure 78A). As predicted, pre-incubation of EGFP-EBV 1:1 with serum from TNE treated mice (negative control) produced ~ 27% fluorescence of Raji (Figure 78B) as did pre-treatment with the non-neutralizing anti-gp350/220 mAb-2L10. In contrast when terminal sera from mice immunized with EBVgp350/220-F VLPs or UV-EBV was pre- incubated with EGFP-EBV, infection was reduced in comparison with TNE-immunized sera: 15% (p = 0.0217) and 5% fluorescent cells (p = 0.0002), respectively. As expected, purified mAb-72Al (positive control) containing only IgGl antibody directed to the gp350/220 attachment epitope was most effective at neutralization (1% fluorescent cells, p = <0.0001 compared with TNE). Antibodies generated after immunization with soluble recombinant gp350/220 ED were least effective (22% fluorescent cells, p = 0.0298 versus TNE). Though the numbers are small, the comparative ability of antibodies generated in response to immunisation with chimeric VLP versus UV-EBV to neutralize EBV infection of Raji cells was not significant (p = 0.2188).

Because of the inability of sera from mice immunized with gp350/220 VLPs to effectively neutralize EBV infection in vitro (sterile condition), we reasoned that incorporation of EBVgH/gL or EBVgB with tumor-associated EBV antigens EBNA-1 and/or LMP2 as components of VLPs will enhance and sustain both humoral and T-cell responses. Furthermore, adoptive transfer of antigen- specific cytotoxic T lymphocytes (CTLs) offers safe and effective therapy for eradication of EBV- associated cancers, however, they do not expand or persist long term (42, 67). Thus, vaccination strategies that induce a robust antibody response and enhance EBV specific T-cell immunity are crucial for an effective EBV vaccine.

Immunization with inactivated virus particles or a subunit vaccine such as soluble recombinant proteins, in the absence of intracellular replication rarely induces robust CTL responses. An important reason for the poor immunogenicity is due to the difficulty of an exogenous antigen to activate the major histocompatibility complex (MHC) class I pathway (68, 69). Typically, antigens that cannot gain access to the cytosol of the host cell activate the MHC class II pathway while antigens that are endocytosed are processed through the MHC class I pathway. VLPs are an interesting exception, since they can be efficiently processed by the MHC class I pathway through receptor mediated binding and entry (52, 70-72). This attribute makes VLPs promising candidates for the development of suburui vaccines, particularly for oncogenic viruses such as EBV. In nature, EBV only infects humans, however, humanized mouse models harboring functional human immune system components are easily infected with EBV (73). Many aspects of human EBV biology, including EBV latent infection, EBV -associated diseases, and T-cell-mcdiatcd immune responses arc reproducible in humanized mice (74). Antibody responses are also elicited in humanized mice, however, the generation of antigen- specific IgG has been challenging (73). As a proof of concept, we first generated a fusion protein between NP and EGFP as illustrated (Figure 79A), NDV-F and EBVgp350/220 (Figure 79B). The NP-EGFP fusion protein was transfected into CHO cells together with gp350/220/HR2 F chimera, and NDV-M as illustrated in Figure 79C. The transfected CHO cells expressed NP-EGFP protein (Figure 80A), and efficiently assembled and released gp350/220-NP-EGFP VLPs into the supernatant when co-transfected with both pCAGGS-M and gp350/220. These gp350/220-NP-EGFP VLPs incorporated proteins of correct molecular sizes as confirmed by immunoblot (Figure 80B) and bound CD21 expressed on the surface of Raji cells (Figure 81).

Using a similar strategy, we incorporated a truncated form of EBNA1 (tEBNAl) in which the Gly-Ala region known to impair presentation of cur-linked sequences is deleted into EBVgp350/220 (Figure 82) and EBVgH/gL-EBNAl VLPs (Figure. 83). This confirmed that it is possible to generate VLPs containing antigens that are not expressed on the surface of the particles, in this case the latent EBV antigen EBNAI . The sequence of the antigen was present in detectable amounts in the supernatant of the transfected cells, and able to bind the antibody despite the deleted Gly-Ala region.

Construction and schematic illustration of pACGGS-EBVgB WT, chimeric pCAGSS-EBVgB-

NDV F, chimeric pCAGGS -NP-LMP2 plasmids is outlined in Figure 84 A-B. These constructs were transfected into CHO cells to determine expression of EBVgB on the cell surface was determined by cytometry (Figure 85). We further confirmed that proteins (gB, LMP2, NP) were made and were of the right sizes (Figure 86).

Several studies on host immune responses against EBV have suggested that both B and T -eel Is immunity play a critical role in the protection against EBV infection and control of EBV-associated diseases. We hypothesized that incorporation of tumor-associated EBV antigens tEBNA-1 and LMP2 as components of VLPs will enhance and sustain both humoral and T-cell responses in BALB/c mice.

To test our hypothesis, groups of 5 mice were irnmunized thrice at day 0, 29 and 54 intraperitoneally with 10μg of EBVgH/gL-tEBNAl VLPs, EBVgB-LMP2 VLPs, EBVgp350/220 VLPs, or UV- inactivatcd EBV resuspended in 500 μΙ of TNE. The animals were boosted twice at day 29 and 54 without adjuvants as illustrated (Figure 87). TNE served as control for immunogens. At day 161 mice were sacrificed and 5 x 10 5 splenocytes were stimulated in vitro with the corresponding as well as control peptides. 1μg/mL of synthetic peptides derived from EBNA1_(HPVGF.ADYFEY), or Promix EBV peptide pool consisting of 26 peptides, each corresponding to a

defined HLA class I -restricted T cell epitope from EBV were used in the assay. After overnight culturing, the supernatants were tested for IFN-γ release by ELISA. SIINFEKL ovalbumin was used as a negative control and concavalin A and IL1B were used as model antigens. Splenocytes from mice immunized with EB VgH/gL-EBNA 1 VLPs and EBVgB-LMP2 VLPs generated significantly higher IFN-γ than mice immunized with UV-inactivated EBV and EBVgp350/220 VLPs (Figure 88). Experiments are ongoing to determine the ability of the mice, sera from specific time points (including terminal bleed) to neutralize EBV expressing EGFP in an in vitro system as outlined (56).

References

1. Rickinson AB, Kieff E. Epstein-Barr Virus. In: Knipe D, Howley P, editors. Fields Virology. Fifth ed. Philadelphia: Lippincott Wilkins and Williams; 2007. p. 2680-700.

2. Cohen JL Fauci AS, Varmus H, Nabel GJ. Epstein-Barr Virus: An Important Vaccine Target for Cancer Prevention. Science Translational Medicine. 2011;3(107): 107fs7-fs7.

3. Kutok J, Wang F. Spectrum of Epstein-Barr virus-associated diseases. Annu Rev Pathol Mech Dis. 2006;1 :375-404.

4. Hjalgrim H, Friborg J, Mclbye ML The epidemiology of EBV and its association with malignant disease. In: Arvin A, Campadelli-Fiume G, Mocarski E, Moore PS, Roizman B, Whitley R, et al., editors. Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis. Cambridge2007. 5. Luzuriaga K, Sullivan JL. Infectious mononucleosis. New England Journal of Medecine 2010;362:1993-2000.

6. Babcock GJ, Decker LL, Volk M, Thorley-Lawson DA. EBV persistence in memory B cells in vivo. Immunity. 1998;9(3):395-404.

7. Goedert JJ, Cote TR, Virgo P, Scoppa SM, Kingma DW, Gail MH, et al. Spectrum of A IDS- associated malignant disorders. Lancet. 1998;351(91 19): 1833-9. Epub 1998/07/04. PubMed PMID: 9652666.

8. Cote TR, Biggar RJ, Rosenberg PS, Devesa SS, Percy C, Yellin FJ, et al. Non-Hodgkin's lymphoma among people with AIDS: Incidence, presentation and public health burden. International journal οτ cancer. 1998;73(5):645-50.

9. Gottschalk S, Rooney CM, Heslop HE. Post-transplant lymphoproliferative disorders. Annu Rev Med. 2005;56:29-44.

10. Cohen J I. Epstein-barr virus vaccines. Clinical & translational immunology. 2015;4( 1 ):e32. Epub 2015/02/12. doi: I0.1038/cti.2014.27. PubMed PMID: 25671 130; PubMed Central PMCID: PMC4318489.

11. Balfour HH, Jr. Progress, prospects, and problems in Epstein-Barr virus vaccine development. Curr Opin Virol. 2014;6C: 1-5. Epub 2014/03/19. doi: 10.1016/j.coviro.2014.02.005. PubMed PMID: 24632197.

12. Biggar RJ, Henle G, B6cker J, Lennette ET, Fleisher G, Henle W. Primary Epstein-Barr virus infections in African infants. Π. Clinical and serological observations during seroconversion.

International Journal of Cancer. 1978;22(3):244-50.

13. Biggar RJ, Henle W, Fleisher G, Becker J, Lennette ET, Henle G. Primary Epstein-Barr virus infections in african infants. I. Decline of maternal antibodies and time of infection. International Journal of Cancer. 1978;22(3):239-43.

14. Gu SY, Huang TM, Ruan L, Miao YH, Lu H, Chu CM, et al. First EBV vaccine trial in humans using recombinant vaccinia virus expressing the major membrane antigen. Dcv Biol Stand.

1995;84:171-7. Epub 1995/01/01. PubMed PMID: 7796951.

15. Moutschen M, Leonard P, Sokal EM, Smets F, Haumont M, Mazzu P, et al. Phase Ι/Π studies to evaluate safety and immunogenicity of a recombinant gp350 Epstein-Barr virus vaccine in healthy adults. Vaccine. 2007;25(24):4697-705. Epub 2007/05/09. doi: 10.1016/j.vaccine.2007.04.008.

PubMed PMID: 17485150. 16. Rees L, Tizard EJ, Morgan AJ, Cubitt WD, Finerty S, Oyewole-Eletu TA, et al. A phase I trial of epstein-barr virus gp350 vaccine for children with chronic kidney disease awaiting transplantation.

Transplantation. 2009;88(8): 1025-9. Epub 2009/10/27. doi: t0.1097/TP.0b013e3181b9d918

00007890-200910270-00013 [pii]. PubMed PMID: 19855249.

17. Sokal EM, Hoppenbrouwers K, Vandermeulen C, Moutschen M, Leonard P, Moreels A, et al.

Recombinant gp350 vaccine for infectious mononucleosis: a phase 2, randomized, double-blind, placebo-controlled trial to evaluate the safety, immunogenicity, and efficacy of an Epstein-Barr virus vaccine in healthy young adults. Journal of Infectious Diseases. 2007;196(12): 1749-53.

18. Janz A, Oezel M, Kurzeder C, Mautncr J, Pich D, Kost M, et al. infectious Epstein-Barr virus lacking major glycoprotein BLLFl (gp350/220) demonstrates the existence of additional viral Iigands.

Journal of virology. 2000;74(21):10142-52. Epub 2000/10/12. PubMed PMID: 1 1024143; PubMed Central PMC1D: PMC102053.

19. Borza CM, Hutt-Fletcher LM. Alternate replication in B cells and epithelial cells switches tropism of Epstein-Barr virus. Nature medicine. 2002;8(6):594-9. Epub 2002/06/04. doi:

10.1038/nm0602-594. PubMed PMID: 12042810.

20. Chesnokova LS, Hutt-Fletcher LM. Fusion of Epstein-Barr virus with epithelial cells can be triggered by alphavbeta5 in addition to alphavbeta6 and alphavbeta8, and integrin binding triggers a conformational change in glycoproteins gHgL. Journal of virology. 2011;85(24): 13214-23. Epub 201 1/10/01. doi: 10.1 128/J VI .05580- I I . PubMed PMID: 21957301; PubMed Central PMCID:

PMC3233123.

21. Miller 1M, Hutt-Fletcher LM. A monoclonal antibody to glycoprotein gp85 inhibits fusion but not attachment of Epstein-Barr virus. Journal of virology. 1988;62(7):2366-72. Epub 1988/07/01. PubMed PMID: 2836619; PubMed Central PMCID: PMC253393.

22. Wang HB, Zhang H, Zhang JP, Li Y, Zhao B, Feng GK, et al. Neuropilin 1 is an entry factor that promotes EBV infection of nasopharyngeal epithelial cells. Nature communications. 2015;6:6240.

Epub 2015/02/12. doi: 10.1038/ncomms7240. PubMed PMID: 25670642.

23. Fingeroth JD, Weis JJ, Tedder TF, Strominger JL, Biro PA, Fearon DT. Epstein-Barr virus receptor of human B lymphocytes is the C3d receptor CR2. Proc Natl Acad Sci U S A.

1984;81(14):4510-4. Epub 1984/07/01. PubMed PMID: 6087328; PubMed Central PMCID:

PMC345620. 24. Nemerow GR, Cooper NR. Early events in the infection of human B lymphocytes by Epstein- Barr virus: the internalization process. Virology. 1984;132(l):186-98. Epub 1984/01/15. PubMed PMID: 6320532.

25. Nemerow GR, Houghten RA, Moore MD, Cooper NR. Identification of an epitope in the major envelope protein of Epstein-Barr virus that mediates viral binding to the B lymphocyte EBV receptor

(CR2). Cell. 1989;56(3):369-77.

26. Ogembo JG, Kannan L, Ghiran I, Nicholson- Weller A, Finberg RW, Tsokos GC, et al. Human complement receptor type 1/CD35 is an Epstein-Barr Virus receptor. Cell Rep. 2013;3(2):371-85. Epub 2013/02/19. doi: 10.1016/j.celrep.2013.01.023. PubMed PMID: 23416052; PubMed Central

27. Tanner J, Weis J, Fearon D, Whang Y, Kieff E. Epstein-Barr virus gp350/220 binding to the B lymphocyte C3d receptor mediates adsorption, capping, and cndocytosis. Cell. 1987;50(2):203-13. Epub 1987/07/17. doi: 0092-8674(87)90216-9 [pii]. PubMed PMID: 3036369.

28. Eiscnbcrg RJ, Atanasiu D, Cairns TM, Gallagher JR, Krummcnacher C, Cohen GH. Herpes virus fusion and entry: a story with many characters. Viruses. 2012;4(5):800-32. Epub 2012/07/04. doi: 10.3390/v4050800. PubMed PMID: 22754650; PubMed Central PMCID: PMC3386629.

29. Li Q, Turk SM, Hutt-Fletcher LM. The Epstein-Barr virus (EBV) BZLF2 gene product associates with the gH and gL homologs of EBV and carries an epitope critical to infection of B cells but not of epithelial cells. Journal of virology. 1995;69(7):3987-94. Epub 1995/07/01. PubMed PMID: 7539502; PubMed Central PMCID: PMC189130.

30. Wang X, Hutt-Fletcher LM. Epstein-Barr virus lacking glycoprotein gp42 can bind to B cells but is not able to infect. Journal of virology. 1998;72(1): 158-63.

31. Molesworth SJ, Lake CM, Borza CM, Turk SM, Hutt-Fletcher LM. Epstein-Barr virus gH is essential for penetration of B cells but also plays a role in attachment of virus to epithelial cells.

Journal of virology. 2000;74(14):6324-32. Epub 2000/06/23. PubMed PMID: 10864642; PubMed Central PMCBD: PMC112138.

32. Wu L, Borza CM, Hutt-Fletcher LM. Mutations of Epstein-Barr virus gH that are differentially able to support fusion with B cells or epithelial cells. Journal of virology. 2005;79(17):10923-30. Epub 2005/08/17. doi: 10.1128/JV1.79.17.10923-10930.2005. PubMed PMID: 16103144; PubMed Central PMCID: PMC1 193614.

33. Fuller AO, Santos RE, Spear PG. Neutralizing antibodies specific for glycoprotein H of herpes simplex virus permit viral attachment to cells but prevent penetration. Journal of virology. 1989;63(8):3435-43. Epub 1989/08/01. PubMcd PMID: 2545914; PubMcd Central PMCID:

PMC250919.

34. Compels UA. Carss AL, Saxby C. Hancock DC. Forrester A, Minson AC. Characterization and sequence analyses of antibody-selected antigenic variants of herpes simplex virus show a

conformationally complex epitope on glycoprotein H. Journal of virology. I991 ;65(5):2393-401. Epub 1991/05/01. PubMcd PMID: 1707982; PubMcd Central PMCID: PMC240591.

35. Nokta M, Tolpin MD, Nadler PI. Pollard RB. Human monoclonal anti-cytomegalovirus (CMV) antibody (MSL 109): enhancement of in vitro lbscarnel- and ganciclovir-induced inhibition of CMV replication. Antiviral research. 1994;24(1 ): 17-26. Epub 1994/05/01. PubMcd PMID: 7944310.

36. Wussow 1 : , Chiuppesi F-, Martinez J, Campo J. Johnson li. 1 flechsig C, et al. Human

cytomegalovirus vaccine based on the envelope gH/gL pentamer complcx20l4.

37. Naranatt PP, Akula SM, Chandran B. Characterization of gamma2 -human hcrpcsvirus-8 glycoproteins gH and gL. Archives of virology. 2002; 147(7): 1349-70. Epub 2002/07/12. doi:

10.1007/s00705-002-0813-7. PubMcd PMID: 121 1 1412.

38. Kirschner AN, Omerovic J, Popov B. Longnecker R, Jardel/ky TS. Soluble Epstein-Barr virus glycoproteins gH. gL. and gp42 form a 1:1:1 stable complex that acts like soluble gp42 in B-cell fusion but not in epithelial cell fusion. Journal of virology. 2006;80(19):9444-54. Epub 2006/09/16. doi: 10.1 128/JV1.00572-06. PubMed PMID: 16973550: PubMed C entral PMC ID: PMC1617263.

39. Rowc CL, Connolly SA, Chen J, Jardctzky TS, Longnecker R. A soluble form of Epstein-Barr virus gH/gL inhibits HBV-induced membrane fusion and does not function in fusion. Virology.

2013;436(l ): l 18-26. Epub 2012/12/04. doi: 10.1016/j.virol .2012.10.039. PubMcd PMID: 23200314; PubMcd Central PMCID: PMC3545092.

40. Li Q, Spriggs MK, Kovats S, Turk SM, Comeau MR, Nepom B, ei al. Epstein-Barr virus uses HLA class II as a cofactor for infection of B lymphocytes. Journal of virology. 1997;71(6):4657-62. 41. Icheva V, Kayser S, Wolff D, Tuve S, Kyzirakos C, Bethge W. et al. Adoptive transfer of Epstein-Barr virus (EBV) nuclear antigen 1 -specific T cells as treatment for EBV reactivation and lymphoprolifcrativc disorders after allogeneic stem-cell transplantation. Journal of Clinical Oncology. 2012:JCO. 201 1.39. 8495.

42. Hcslop HE, Ng CY, Li C. Smith CA, Loftin SK, Krancc RA, et al. Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of genc-modified virus-specific T lymphocytes. Nature medicine. 1996:2(5):551-5. 43. Icheva V, Kayser S, Wolff D, Tuve S, Kyzirakos C, Bethge W, et al. Adoptive transfer of epstein-barr virus (EBV) nuclear antigen 1 -specific t cells as treatment for EBV reactivation and lymphoproliferativc disorders after allogeneic stem-cell transplantation. J Clin Oncol. 2013;31 (1):39-

48. Epub 2012/1 1/22. doi: 10.1200/JCO.201 1.39.8495. PubMed PMID: 23169501.

44. Taylor GS, Jia H, Harrington K, Lee LW, Turner J, Ladell K, et al. A Recombinant Modified

Vaccinia Ankara Vaccine Encoding Epstein-Barr Virus (EBV) Target Antigens: A Phase I Trial in UK

Patients with EBV-Positive Cancer. Clin Cancer Res. 2014;20(19):5009-22. Epub 2014/08/16. doi:

10.1158/1078-0432.CCR-14-1122-T. PubMed PMID: 25124688.

45. Fogg MH, Wirth LJ, Posner M, Wang F. Decreased EBNA-1 -specific CD8+ T cells in patients with Epstein-Barr virus-associated nasopharyngeal carcinoma. Proc Natl Acad Sci U S A.

2009;106(9):3318-23. Epub 2009/02/13. doi: I0.l073/pnas.0813320106. PubMed PMID: 1921 1798; PubMed Central PMCID: PMC2651339.

46. Long HM, Chagoury OL, Leese AM, Ryan GB, James E, Morton LT, et al. MHC II tetramers visualize human CD4+ T cell responses to Epstein-Barr virus infection and demonstrate atypical kinetics of the nuclear antigen EBNA1 response. The Journal of experimental medicine.

2013;210(5):933-9. Epub 2013/04/10. doi: 10.1084/jem.20121437. PubMed PMID: 23569328;

PubMed Central PMCID: PMC3646497.

47. Lee SP, Brooks JM, Al-Jarrah H, Thomas WA, Haigh TA, Taylor GS, et al. CD8 T cell recognition of endogenously expressed Epstein-Barr virus nuclear antigen 1. The Journal of experimental medicine. 2004; 199(10): 1409-20. Epub 2004/05/19. doi: 10.1084/jem.20040121.

PubMed PMID: 15148339; PubMed Central PMCID: PMC2211813.

48. Taylor GS, Haigh TA. Gudgeon NH, Phelps RJ, Lee SP, Steven NM, et al. Dual stimulation of Epstein-Barr Virus (EBV)-specific CD4+- and CD8+-T-cell responses by a chimeric antigen construct: potential therapeutic vaccine for EBV-positive nasopharyngeal carcinoma. Journal of virology.

2004;78(2):768-78. Epub 2003/12/25. PubMed PMID: 14694109; PubMed Central PMCID:

PMC368843.

49. Apcher S, Daskalogianni C, Manoury B, Fahraeus R. Epstein-Barr virus-encoded EBNA1 interference with MHC class I antigen presentation reveals a close correlation between mRNA translation initiation and antigen presentation. PLoS Pathog. 2010;6(10):c1001151. Epub 2010/10/27. doi: 10.1371/journal.ppat.1001 151. PubMed PMID: 20976201; PubMed Central PMCID:

PMC2954899. 50. Hui EP. Taylor GS, Jia H, Ma BB. Chan SL. Ho R, cl al. Phase I trial of recombinant modified vaccinia ankara encoding Epstein-Barr viral tumor antigens in nasopharyngeal carcinoma patients.

Cancer Res. 2013:73(6): 1676-88. Epub 2013/01 /26. doi: 10.1 158/0O08-5472.CAN- 12-2448. PubMcd

PMID: 23348421.

51. Pavlova S, Feederle R, Gartner K, Fuclis W, Gran/ow H, Deleclluse HJ. An Epstein- Burr virus mutant produces immunogenic defective particles devoid of viral DN A. Journal of virology.

2013;87(4):201 1-22. Rpub 2012/12/14. doi: 10.1 128/JVI.02533-12. PubMed PMID: 23236073;

PubMed Central PMCID: PMC3571473.

52. Ruiss R, Jochum S, Wanner G, Reisbach G, Hammerschmidt W, Zeidler R. A virus-like particle-based Epstem-Barr virus vaccine. Journal of virology. 2011;85(24): 13105-13.

53. Speck P, Longneeker R. Epstein-Barr virus (EBV) infection visualized by EGFP expression demonstrates dependence on known mediators of EBV entry. Archive of Virology. 1999: 144(6): 1 123- 37. Epub 1999/08/14. PubMcd PMID: 10446648.

54. McGinncs LW, Rcittcr JN. Gravel K. Morrison TG. Evidence for mixed membrane topology of the newcasile disease virus fusion protein. Journal of Virolology. 2003 ;77(3): 1951 -63. Epub

2003/01/15. PubMcd PMID: 12525629: PubMcd Central PMCJD: PMC140911.

55. Biggin M, Farrell PJ, Barrel! BG. Transcription and DNA sequence of the BamHI L fragment of B95-8 Epstein-Barr virus. EMBO J. 1984:3(5): 1083-90. Epub 1984/05/01. PubMed PMID:

6203743; PubMcd Central PMCID: PMC557477.

56. Ogeinbo JG, Muraswki MR, McGinncs I.W. Parcharidou A, Sutiwisesak R, Tison T, et al. A chimeric EBV gp350/220-bascd VLP replicates the virion B-ccll attachment mechanism and elicits long-lasting neutralizing antibodies in mice. Journal of Translational Medicine. 20I 5; I 3(I):50.

57. Pantua IID, McGinnes LW, Peeples ME. Morrison TG. Requirements for the assembly and release of Newcastle disease virus-like particles. Journal of Virolology. 2006:80(22): 11062-73. Epub 2006/09/15. doi: 10.1128/ JVI.00726-06. PubMed PMID: 16971425; PubMed Central PMCID:

PMC 1642154.

58. McGinncs LW, Morrison TG. Newcastle Disease Virus-Like Particles: Preparation,

Purification. Quantification, and Incorporation of Foreign Glycoproteins. Current Protocols in

Microbiology. 2013: 18.2. I -.2. 21.

59. Lalibcrtc JP, McGinncs LW, Pccplcs ME, Morrison TG. Integrity of membrane lipid rafts is necessary for the ordered assembly and release of infectious Newcastle disease virus particles. Journal oi ' virology. 2006:80(21): 10652-62. Epub 2006/10/17. doi: 10.1128/ JVI.01183-06. PubMcd PMID:

17041223; PubMed Central PMCID: PMC1641742.

60. Battisti AJ, Mcng G, Winkler DC, McGinncs L\V, Plcvka P, Steven AC. ct al. Structure and assembly of a paramyxovirus matrix protein. Proceedings of the National Academy of Sciences.

2012; 109(35): 13996-4000.

61. Ghiran I, Glodck AM, Weaver G, Klickstcin LB, Nicholson-Wcllcr A. Ligation of erythrocyte CR I induces its clustering in complex with scaffolding protein FAP-1. Blood. 2008: 1 12(8):3465-73. Epub 2008/08/08. doi: blood-2008-04-151845 [pii]

10.1 182/blood-2008-04- l 5 l 845. PubMcd PMID: 18684861 ; PubMcd Central PMCID: PMC2569 I 83. 62. Murawski MR, McGinnes LW. Finberg RW. Kurt-Jones EA, Massarc MJ. Smith G, et al.

Newcastle disease virus-like particles containing respiratory syncytial virus G protein induced protection in BALB/c mice, with no evidence of immunopathology. Journal of virology.

2010;84(2): l 1 10-23. Hpub 2009/1 1/06. doi: 10.1 128/J VI .01709-09. PubMcd PMID: 19889768;

PubMcd Central PMCID: PMC2798376.

63. Sashihara J, Burbelo PD, Savoldo B, Pierson TC, Cohen JI. Human antibody tilers to Epstein -

Barr Virus (EBV) gp350 correlate with neutralization of infectivity better than antibody titers to EBV gp42 using a rapid flow cytometry -based EBV neutralization assay. Virology. 2009:391(2):249-56.

Epub 2009/07/09. doi: S0042 -6822(09)00358-4 [pii]

10.1016/j . vi rol .2009.06.013. PubMcd PMID: 19584018; PubMcd Central PMCID: PMC2728425. 64. Pantua H. McGinncs LW, Leszyk J, Morrison TG. Characterization of an alternate form of Newcastle disease virus fusion protein. Journal of virology. 2005;79( 18): 1 1660-70. Epub 2005/09/06. doi: 10.1 128/JVI.79. I 8. I 1660-1 1670.2005. PubMcd PMID: 16140743; PubMcd Central PMCID: PMC 1212644.

65. Tanner J, Whang Y, Sample J, Sears A, Kieff E. Soluble gp350/220 and deletion mutant glycoproteins block. Epstein-Barr virus adsorption to lymphocytes. Journal of virology.

1988;62( 12):4452-64. Epub 1988/12/01. PubMed PMID: 2460635; PubMed Central PMCID:

PMC253554.

66. Civoli K, Krocnkc MA, Rcynhardt K. Zhuang Y, Kaliyapcrumal A, Gupta S. Development and optimization of neutralizing antibody assays to monitor clinical immunogenicity. Bioanalysis.

2012;4(22):2725-35. Epub 2012/12/06. doi: 10.4155/bio.12.239. PubMcd PMID: 23210655. 67. Gallot G, Vollant S, Saiagh S, Clémenceau B, Vivien R, Cerato E, et al. T-cell Therapy Using a Bank of EBV-specific Cytotoxic T Cells: Lessons From a Phase I/II Feasibility and Safety Study. Journal of Immunotherapy. 2014;37(3): 170-9.

68. Braciale TJ, Morrison LA, Sweetser MT, Sambrook J, Gething MJ, Braciale VL. Antigen presentation pathways to class I and class II MHC-restricted T lymphocytes. Immunological reviews. 1987;98:95-1 14. PubMed PMID: 2443444.

69. Germain RN. Immunology. The ins and outs of antigen processing and presentation. Nature. 1986;322(6081):687-9. doi: 10.1038/322687a0. PubMed PMID: 3489186.

70. Adhikary D, Behrends U, Feederle R, Dclccluse HJ, Mautncr J. Standardized and highly efficient expansion of Epstein-Barr vinis-speciflc CD4+ T cells by using virus-like particles. Journal of virology. 2008;82(8):3903-l 1. Epub 2008/02/15. doi: 10.1 128/JVI.02227-07. PubMed PMID:

18272580; PubMed Central PMCID: PMC2293016.

71. Schirmbeck R, Bohm W, Reimann J. Virus-like particles induce MHC class l-restricted T-cell responses. Intervirology. 1996;39(1-2):1 1 1-9.

72. Paliard X, Liu Y, Wagner R, Wolf H, Baenziger J, Walker CM. Priming of strong, broad, and long-lived HIV type 1 p55gag-specific CD8+ cytotoxic T cells after administration of a virus-like particle vaccine in rhesus macaques. AIDS research and human retroviruses. 2000;16(3):273-82.

73. Yajima M, Imadome K, Nakagawa A, Watanabe S, Terashima K, Nakamura H, et al. A new humanized mouse model of Epstein-Barr virus infection that reproduces persistent infection, lymphoproliferative disorder, and cell-mediated and humoral immune responses. The Journal of infectious diseases. 2008;198(5):673-82. Epub 2008/07/17. doi: 10.1086/590502. PubMed PMID: 18627269.

74. Chatterjee B, Leung CS, Milnz C. Animal models of Epstein-Barr virus infection. Journal of immunological methods. 2014.

Each and every publication and patent mentioned in the above specification is herein incorporated by reference in its entirety for all purposes. Various modifications and variations of the described methods and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific embodiments, the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the .: UMASSW 18176 invention which are obvious to those skilled in the art and in fields related thereto are intended to be within the scope of the following claims.