CA3016867A1 - Human cytomegalovirus gb polypeptide - Google Patents

Human cytomegalovirus gb polypeptide Download PDF

Info

Publication number
CA3016867A1
CA3016867A1 CA3016867A CA3016867A CA3016867A1 CA 3016867 A1 CA3016867 A1 CA 3016867A1 CA 3016867 A CA3016867 A CA 3016867A CA 3016867 A CA3016867 A CA 3016867A CA 3016867 A1 CA3016867 A1 CA 3016867A1
Authority
CA
Canada
Prior art keywords
polypeptide
seq
amino acid
acid sequence
cmv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA3016867A
Other languages
French (fr)
Inventor
Xiaoyuan Sherry CHI
Xinzhen Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc filed Critical Pfizer Inc
Publication of CA3016867A1 publication Critical patent/CA3016867A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16171Demonstrated in vivo effect

Abstract

The present invention relates to a polypeptide that includes at least one mutation in the fusion loop 1 region and/or in the fusion loop 2 region and/or in the furin-like cleavage site of a human cytomegalovirus gB polypeptide. In one embodiment, the polypeptide undergoes a structural conformation change in response to pH change.

Description

HUMAN CYTOMEGALOVIRUS GB POLYPEPTIDE
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to United States Provisional Patent Application .. Serial No. 62/307,423 filed on March 11, 2016, the contents of which is incorporated in its entirety by reference herein.
FIELD OF THE INVENTION
The present invention relates to human cytomegalovirus (HCMV) compositions and methods thereof.
BACKGROUND
Human cytomegalovirus (HCMV) is a double stranded DNA virus of the 13-herpesvirus family. HCMV is the leading cause of congenital and neonatal hearing loss resulting from vertical virus transmission following infection or reactivation of latent virus in pregnant women. In addition, HCMV is a common opportunistic pathogen affecting immunosuppressed transplant patients, such as solid organ /
stem cell transplant patients, AIDS patients, etc. Though development of a vaccine against HCMV has been listed as a top priority by the Institute of Medicine, none has been licensed to date.
The HCMV genome encodes several envelope glycoproteins, one of which is glycoprotein B (gB). Glycoprotein B is an important surface target for neutralizing antibody (nAb) response in natural infection and is required for virus entry into cells by functioning as a fusogen.
HCMV subunit vaccines incorporating gB have been under development.
Studies have suggested that gB subunit vaccines were safe and immunogenic, though further improvements in potency and durability of protection were desirable.
Accordingly, safe and effective immunogenic compositions against cytomegalovirus infections, as well diagnostic reagents capable of detecting immunogenic stimuli resulting from CMV infections, guiding the design of gB-based HCMV vaccines, and/or supporting the development of therapeutic antibodies against this medically relevant human pathogen are needed.

SUMMARY OF THE INVENTION
To meet these and other needs, in one aspect, the present invention relates to a polypeptide that includes at least one mutation in the fusion loop 1 (F
Li) region .. of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes at least one mutation in the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes at least one mutation in the fusion loop 1 (FL1) region and the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes at least one mutation in the furin-like cleavage site of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes at least two mutations in the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes at least two mutations in the fusion loop 1 (FL1) region and the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes at least two mutations in the furin-like cleavage site of an HCMV gB polypeptide.
In one aspect, the invention relates to a polypeptide that includes a mutation at position Y155, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes a mutation at positions Y155 and 1156, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes a mutation at positions Y155, 1156, and H157, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes a mutation at positions 1156 and H157, as compared to SEQ ID NO: 6.
2 In one aspect, the invention relates to a polypeptide that includes a mutation at positions Y155, 1156, H157, and W240, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes a mutation at positions Y155 and W240, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes a mutation at positions Y155, H157, and W240, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes a mutation at positions Y155 and H157, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutation Y155G, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations Y155G and I156H, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations Y155G, I156H, and H157R, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations I156H and H157R, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations Y155G, I156H, H157R, and W240A, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations Y155G and W240A, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations Y155G, H157R, and W240A, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a polypeptide that includes the mutations Y155G and H157R, as compared to SEQ ID NO: 6.
3
4 In one aspect, the invention relates to a polypeptide that includes an amino acid sequence that is at least about 60% identical to SEQ ID NO: 1.
In one aspect, the invention relates to a polypeptide that includes an amino acid sequence that is at least about 60% identical to SEQ ID NO: 2.
In one aspect, the invention relates to a polypeptide that includes an amino acid sequence that is at least about 60% identical to SEQ ID NO: 3.
In one aspect, the invention relates to a polypeptide that includes an amino acid sequence that is at least about 60% identical to SEQ ID NO: 5.
In one aspect, the invention relates to a polypeptide that includes an amino acid sequence that is at least about 60% identical to SEQ ID NO: 7.
In one aspect, the invention relates to a polypeptide that includes an amino acid sequence that is at least about 60% identical to SEQ ID NO: 8.
In one aspect, the invention relates to a polypeptide that includes the amino acid sequence set forth in SEQ ID NO: 1.
In one aspect, the invention relates to a polypeptide that includes the amino acid sequence set forth in SEQ ID NO: 2.
In one aspect, the invention relates to a polypeptide that includes the amino acid sequence set forth in SEQ ID NO: 3.
In one aspect, the invention relates to a polypeptide that includes the amino acid sequence set forth in SEQ ID NO: 5.
In one aspect, the invention relates to a polypeptide that includes the amino acid sequence set forth in SEQ ID NO: 7.
In one aspect, the invention relates to a polypeptide that includes the amino acid sequence set forth in SEQ ID NO: 8.

In one embodiment, the polypeptide does not include a mutation at any one of the following positions: (i) R236, (ii) G237, (iii) T158; (iv) Y242. In one embodiment, the polypeptide does not include any one of the following mutations: (i) R236N, (ii) G237N, (iii) T158N; (iv) Y242T; (v) Y242S; (vi) Y2420. In one embodiment, the amino acid sequence SEQ ID NO: 9 is a part of the polypeptide sequence. In one embodiment, the amino acid sequence SEQ ID NO: 10 is a part of the polypeptide sequence. In one embodiment, the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10 is a part of the polypeptide sequence. In one embodiment, the polypeptide does not include a protease cleavage site. In one embodiment, the polypeptide does not include a wild-type CMV protease cleavage site. In one embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type CMV protease cleavage site. In one embodiment, the polypeptide does not include an N- glycosylation site that includes N-X-S/T/C motif, wherein X is any amino acid residue. In one embodiment, the polypeptide does not include a modified amino acid sequence that introduces an linked glycosylation site. In one embodiment, the polypeptide does not include a deletion or substitution of any one of the amino acid residues selected from the group consisting of 154, 158, 159, 160, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 241, and 242, according to the numbering of SEQ ID NO: 6. In one embodiment, the polypeptide does not include a mutation of any one of the amino acid residues: Y160, R236, S238, T239, and Y242, according to the numbering of SEQ ID NO: 6. In one embodiment, the polypeptide does not include the cytoplasmic tail of HCMV gB.
5 In one embodiment, the polypeptide does not contain an insect cell pattern of glycosylation. In one embodiment, the polypeptide is contacted with ethylenediaminetetraacetic acid (EDTA).
In one aspect, the invention relates to a composition that includes the .. polypeptide described herein, and a diluent.
In one aspect, the invention relates to a composition that includes the polypeptide described herein, and an adjuvant. In one embodiment, the composition is immunogenic. In one embodiment, the composition is for use in inducing an immune response against cytomegalovirus.
In one aspect, the invention relates to a recombinant nucleic acid molecule encoding the polypeptide described herein, wherein the polypeptide undergoes a structural conformation change in response to a pH change. In one embodiment, said recombinant nucleic acid (a) is not a self-replicating RNA molecule; (b) is not an alphavirus replicon; (c) does not encode any alphavirus nonstructural proteins, such as NSP1, NSP2, NSP3 and NSP4; (d) does not contain: an Internal Ribosomal Entry Site (IRES), such as EMCV or EV71; and/or (e) does not contain a viral 2A
site, such as FMDV.
In one aspect, the invention relates to a method for raising antibodies using the polypeptide described herein. In one embodiment, said antibody is for use in a diagnostic assay. In one embodiment, said antibody is labelled directly or indirectly.
In one embodiment, said antibody is for use in therapy.
In one aspect, the invention relates to a method of eliciting an immune response in a mammal, the method that includes administering to the mammal an effective amount of the polypeptide described herein.
6 BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1- illustration of HCMV gB705(SEQ ID NO: 1). SS represents a signal sequence; D1 represents the Domain I; FL represents a fusion loop; RxxR
mutation represents knock-out mutations of the furin-like cleavage site; 6h represents an optional 6xHis tag.
FIG. 2 - sequence of HCMV gB705 (SEQ ID NO: 2).
FIG. 3 - The gB705 protein was transiently expressed in Expi293 and 293T cells after transfection respectively. The target protein was secreted into culture medium, precipitated by an affinity tag, and resolved on SDS-PAGE under reducing condition.
The presence of the gB705 protein was visualized by a Western blot analysis using poly-clonal anti-gB serum. As shown, gB705 exhibited a single band with an apparent molecular weight about 130kDa, consistent with the design of the furin-cleavage site being knocked out by mutations.
FIG. 4A-B - FIG. 4A Binding of human anti-HCMV gB monoclonal antibodies to gB705 protein; FIG. 4B Binding of human anti-HCMV gB monoclonal antibodies to Sino gB protein (gB from Sino Biologicals, Inc.) (ELN# 00710043-0177).
FIG. 5 - HCMV gB705 protein (SEQ ID NO: 2) was analyzed on SDS-PAGE and exhibited a single band under a fully reduced condition and showed one additional high molecular weight band under partially reduced conditions, representing a trimer.
FIG. 6- Sedimentation Velocity analysis of gB705 (ELN# 00708337-0110).
FIG. 7 - Velocity sedimentation analysis of 10 mM EDTA-treated gB705 (ELN#
00708337-0110).
FIG. 8A-C- Sedimentation velocity analysis of EDTA-treated gB705 at pH 5.2 (FIG.
8A) and pH 8.7 (FIG. 8B); Cryo electron microscopy analysis of EDTA-treated gB705 proteins pH 5.2 (left) or pH 8.7 (right) (FIG. 8C) FIG. 9A-B - FIG. 9A Far UV CD analysis; FIG. 9B intrinsic fluorescence spectroscopy (ELN# 00708337-0115, 0117) of gB705 at pH 5.2 and pH 8.7.
7 FIG. 10- Near UV CD spectroscopy analysis of gB705 at pH 5.2 and pH 8.7.
Buffer 1, 50 mM citrate, 100 mM phosphate; Buffer 2, 50 mM Na0Ac; Buffer 3, 100 mM Na carbonate/bicarbonate; Buffer 4, 50 mM Na borate (ELN# 00708337-0115, 0117).
FIG. 11A-B - DSC analysis of gB705 at (FIG. 11A) pH 5.2 and (FIG. 11B) pH 8.7.
FIG. 12A-C - (A) ANS binding of gB705 at pH 5.2, pH 7.4 and pH 8.7 (ELN#
00708337-0115, 0117); (B) ANS binding of gB705 at pH 8.7 and 5.2 and shifted from pH 8.7 to pH 5.2; (C) ANS binding of gB705 at pH 8.7 and 5.2 and shifted from pH
5.2 to pH 8.7 (ELN# 00708329-0153) FIG. 13- illustration of RhCMV gB674 (SEQ ID NO: 7). SS represents a signal sequence; FL represents a fusion loop; 6h represents a 6xHis tag FIG. 14- Graph showing anti-gB binding titers from six vaccines on week 9 after three doses of vaccination FIG. 15A-C - Six RhCMV(-) monkeys were immunized with placebo (A), recombinant RhCMV pentamer (B), or pentamer + RhCMV gB674 (C) intramuscularly in QS-21 adjuvant at week 0, 4 & 8. The animals were then challenged with live RhCMV
(UCDE52 strain) orally for five times. Virus shedding in saliva samples were measured using qPCR (low limit of quantitation (LLOQ) is 125 DNA copy per ml) after virus challenge. Week 9 was one week after the third vaccination and one week prior to the first oral challenge. Week 15 was first sample at one week post the fifth virus challenge.
FIG. 16- TEM image of negative staining of gB705 (62000X magnification) (ELN#
00702423-0124) FIG. 17A-B - (A) SDS-PAGE analysis of rhesus gB674, demonstrating the lack of protease cleavage as designed and >95% purity, NR = non-reducing; R =
reducing;
(B) Size Exclusion chromatography of rhesus gB674 protein showing elution as a major peak (MW -313 kDa), consistent with a homogenous individual trimer
8 SEQUENCE IDENTIFIERS
SEQ ID NO: 1 sets forth the amino acid sequence for HCMV VR1814 gB705.
SEQ ID NO: 2 sets forth the amino acid sequence for HCMV VR1814 gB705, without an initial methionine. SEQ ID NO: 2 is identical to SEQ ID NO: 1 but for the absence of the initial methionine.
SEQ ID NO: 3 sets forth the amino acid sequence for a fragment of HCMV VR1814 gB705 (residues 25-705 OF SEQ ID NO: 1).
SEQ ID NO: 4 sets forth the amino acid sequence for the signal sequence of an HCMV VR1814 gB.
SEQ ID NO: 5 sets forth the amino acid sequence for HCMV VR1814 gB705 with linker SEQ ID NO: 6 sets forth the amino acid sequence for GenBank Accession #
A0Z79977.1 for envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5].
SEQ ID NO: 7 sets forth the amino acid sequence for an analogous construct of HCMV gB705 construct, named as RhCMV gB674, which was designed and tested in RhCMV(-) monkeys SEQ ID NO: 8 sets forth the amino acid sequence for an analogous construct of HCMV gB705 construct, named as RhCMV gB674, which was designed and tested in RhCMV(-) monkeys, without an initial methionine. SEQ ID NO: 8 is identical to SEQ ID NO: 7 but for the absence of the initial methionine.
SEQ ID NO: 9 sets forth the amino acid sequence for VVDPLPP705, a part of the membrane proximal region (MPR) of HCMV gB.
SEQ ID NO: 10 sets forth the amino acid sequence for RA457TKA4595.
SEQ ID NO: 11 sets forth the amino acid sequence for GenBank Accession #
GU552457.1 for macacine herpesvirus 3 isolate 21252 glycoprotein B (RhUL55).
9 SEQ ID NO: 12 sets forth the amino acid sequence for the predicted fusion loop region of the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], from GenBank Accession # ACZ79977.1.
SEQ ID NO: 13 sets forth the amino acid sequence for the predicted fusion loop region of the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession # ACZ79977.1.
SEQ ID NO: 14 sets forth the amino acid sequence for the furin-like cleavage site of the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession # ACZ79977.1.
SEQ ID NO: 15: sets forth the amino acid sequence for the protease cleavage site of the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession # ACZ79977.1.
10 DETAILED DESCRIPTION
The inventors surprisingly discovered mutations that can be introduced into a cytomegalovirus (CMV) gB polypeptide, which can, among other things, greatly facilitate the production and subsequent purification of a stable gB antigen;
significantly improve the efficiency of production of a gB polypeptide;
maintain and/or increase antigenicity of a gB polypeptide, as compared to the wild-type gB
polypeptide; facilitate a focused immune response to gB; and reduce and/or eliminate steric occlusion of neutralizing epitopes of gB. The modified gB
polypeptide also surprisingly demonstrated strong structural integrity and a potential to undergo structural conformational changes in tertiary structure in response to significant pH changes. The structural changes were strictly pH-dependent.
Without being bound by theory, the potential to undergo structural conformational changes in response to significant pH changes may be a newly recognized property of the HCMV gB ectodomain.
gB is an envelope glycoprotein B having numerous roles, one of which is the involvement in the fusion of the cytomegalovirus with host cells. It is encoded by the UL55 gene of HCMV genome.
In one aspect, the invention relates to a modified HCMV gB that is based on the VR1814 gB sequence (GenBank# ACZ79977.1; SEQ ID NO: 6). However, the .. present invention is applicable to gB proteins originating from any CMV
strain.
Unless otherwise stated, references to the numbering of amino acid residue positions of a CMV gB polypeptide as used herein are in relation to the amino acid sequence of the gB protein of SEQ ID NO: 6, from the clinical isolate VR1814 strain.
Comparable amino acid positions in a gB protein of any other CMV strains can be .. determined by those of ordinary skill in the art. Accordingly, the term "CMV gB
protein" or "HCMV gB protein" as used herein is to be understood as a HCMV gB
protein from any human HCMV strain (not limited to the VR1814 strain). The actual residue position number may need to be adjusted for gB proteins from other human CMV strains depending on the actual sequence alignment.
In one embodiment, the modified HCMV gB polypeptide includes amino acids 1-705 of SEQ ID NO: 6. In another embodiment, the modified HCMV gB polypeptide includes at least one mutation in the fusion loop 1 region of an HCMV gB
polypeptide. The precise boundaries of the fusion loop 1 region of a wild-type VR1814 gB polypeptide are currently not fully defined, however, the predicted
11 sequence of the fusion loop 1 region of a wild-type VR1814 gB polypeptide is Y153AYIHT158 (SEQ ID NO: 12).
In another embodiment, the modified HCMV gB polypeptide includes at least one mutation in the fusion loop 2 region of an HCMV gB polypeptide. The precise .. boundaries of the fusion loop 2 region of a wild-type VR1814 gB polypeptide are currently not fully defined, however, the predicted sequence of the fusion loop 2 region of a wild-type VR1814 gB polypeptide is G237STWLYRE244 (SEQ ID NO: 13).
In a preferred embodiment, the modified HCMV gB polypeptide includes at least one mutation in the fusion loop 1 region and in the fusion loop 2 region of an .. HCMV gB polypeptide. In another preferred embodiment, the modified HCMV gB
polypeptide includes a total of at most four mutations in the fusion loop 1 region and in the fusion loop 2 region of an HCMV gB polypeptide.
In another embodiment, the modified HCMV gB polypeptide includes at least one mutation in the furin-like cleavage site of an HCMV gB polypeptide. In a preferred embodiment, the modified HCMV gB polypeptide includes at most two mutations in the furin-like cleavage site of an HCMV gB polypeptide.
In another preferred embodiment, the modified HCMV gB polypeptide includes at least one mutation in the fusion loop 1 region, at least one mutation in the fusion loop 2 region, and at least one mutation in the furin-like cleavage site of an HCMV gB polypeptide. The sequence of the furin-like cleavage site of a wild-type VR1814 gB polypeptide is R458TKR459 (SEQ ID NO: 14).
In another preferred embodiment, the modified HCMV gB polypeptide includes at least two mutations in the fusion loop 1 region, at least one mutation in the fusion loop 2 region, and at least one mutation in the furin-like cleavage site of an HCMV gB polypeptide.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at position Y155, as compared to SEQ ID NO:
6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions Y155 and 1156, as compared to SEQ
ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions Y155, 1156, and H157, as compared to SEQ ID NO: 6.
12 In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions 1156 and H157, as compared to SEQ
ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions Y155,1156, H157, and W240, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions Y155 and W240, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions Y155, H157, and W240, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having a mutation at positions Y155 and H157, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutation Y155G, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations Y155G and I156H, as compared to SEQ ID
NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations Y155G, I156H, and H157R, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations I156H and H157R, as compared to SEQ ID
NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations Y155G, I156H, H157R, and W240A, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations Y155G and W240A, as compared to SEQ ID
NO: 6.
13 In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations Y155G, H157R, and W240A, as compared to SEQ ID NO: 6.
In one aspect, the invention relates to a CMV gB polypeptide or immunogenic fragment thereof having the mutations Y155G and H157R, as compared to SEQ ID
NO: 6.
In one embodiment, the gB polypeptide does not include a mutation at any one of the following positions, individually or in combination: (i) R236, (ii) G237, (iii) T158; (iv) Y242.
For example, in one embodiment, the gB polypeptide does not include any one of the following mutations, individually or in combination: (i) R236N, (ii) G237N, (iii) T158N; (iv) Y242T; (v) Y2425; (vi) Y242C.
14 Exemplary Modified Human CMV gB polypeptides In one aspect, the invention relates to a polypeptide having the amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%.
85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 1.
In one embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO:
1, includes the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) as a part of the polypeptide sequence.
Without being bound by theory, the sequence set forth in SEQ ID NO: 9 may be highly hydrophobic and structurally stiff, which facilitates immunogenicity of the polypeptide. Accordingly, in one embodiment, the sequence set forth in SEQ ID
NO:
9 is hydrophobic.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 1, includes the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.
Without being bound by theory, the sequence set forth in SEQ ID NO: 10 may prevent cleavage of the gB polypeptide by cellular proteases. Preferably, the polypeptide of the invention does not include a protease cleavage site. For example, in one embodiment, the polypeptide does not include a wild-type, natural CMV
protease cleavage site, e.g., RTKR459 (SEQ ID NO: 15) for the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession #
A0Z79977.1. In another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site. In yet another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type, natural CMV protease cleavage site. In one embodiment, the polypeptide does not include a protease cleavage site at or encompassing residue R459 at position 459 of a human CMV gB, according to the numbering of SEQ ID
NO:
6, wherein the protease cleavage site includes at most 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 0r3 amino acid residues in length. For example, the polypeptide does not include a protease cleavage site encompassing residue R459 at position 459 of a human CMV gB, according to the numbering of SEQ ID NO: 6, wherein the protease cleavage site includes at most 6, more preferably at most 5, and most preferably at most 4 amino acid residues in length.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 1, includes both the amino acid sequence VVDPLPID705 (SEQ ID NO: 9) and the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.

In one aspect, the invention relates to a polypeptide having the amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%.
85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 2.
In one embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO:
2, includes the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) as a part of the polypeptide sequence.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 2, includes the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.
Preferably, the polypeptide does not include a protease cleavage site. For example, in one embodiment, the polypeptide does not include a wild-type, natural CMV protease cleavage site, e.g., RTKR459 (SEQ ID NO: 15) for the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession # A0Z79977.1. In another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site. In yet another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type, natural CMV protease cleavage site.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 2, includes both the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) and the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.

In one aspect, the invention relates to a polypeptide having the amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%.
85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 3.
In one embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO:
3, includes the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) as a part of the polypeptide sequence.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 3, includes the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.
Preferably, the polypeptide does not include a protease cleavage site. For example, in one embodiment, the polypeptide does not include a wild-type, natural CMV protease cleavage site, e.g., RTKR459 (SEQ ID NO: 15) for the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession # A0Z79977.1. In another embodiment, the polypeptide does not include .. a non-naturally occurring protease cleavage site. In yet another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type, natural CMV protease cleavage site.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 3, includes both the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) and the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.

In one aspect, the invention relates to a polypeptide having the amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%.
85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 5.
In one embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO:
5, includes the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) as a part of the polypeptide sequence.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 5, includes the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.
Preferably, the polypeptide does not include a protease cleavage site. For example, in one embodiment, the polypeptide does not include a wild-type, natural CMV protease cleavage site, e.g., RTKR459 (SEQ ID NO: 15) for the envelope glycoprotein B of strain VR1814 [Human beta-herpesvirus 5], based on GenBank Accession # A0Z79977.1. In another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site. In yet another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type, natural CMV protease cleavage site.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 5, includes both the amino acid sequence VVDPLPP705 (SEQ ID NO: 9) and the amino acid sequence RA457TKA4595 (SEQ ID NO: 10) as a part of the polypeptide sequence.

Exemplary Modified Rhesus CMV gB polypeptides In one aspect, the invention relates to a polypeptide having the amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%.
85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 7.
In one embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO:
7, includes the amino acid sequence VVDPLPP (SEQ ID NO: 9) as a part of the polypeptide sequence.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 7, includes the amino acid sequence RATKAS (SEQ ID NO: 10) as a part of the polypeptide sequence. See, for example, residues 428-433 of SEQ ID NO: 7.
Preferably, the polypeptide does not include a protease cleavage site. For example, in one embodiment, the polypeptide does not include a wild-type, natural CMV protease cleavage site. In another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site. In yet another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type, natural CMV protease cleavage site.

In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 7, includes both the amino acid sequence VVDPLPP (SEQ ID NO: 9) and the amino acid sequence RATKAS (SEQ ID NO: 10) as a part of the polypeptide sequence.

In one aspect, the invention relates to a polypeptide having the amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%.
85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 8.
In one embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO:
8, includes the amino acid sequence VVDPLPP (SEQ ID NO: 9) as a part of the polypeptide sequence.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 8, includes the amino acid sequence RATKAS (SEQ ID NO: 10) as a part of the polypeptide sequence.
Preferably, the polypeptide does not include a protease cleavage site. For example, in one embodiment, the polypeptide does not include a wild-type, natural CMV protease cleavage site. In another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site. In yet another embodiment, the polypeptide does not include a non-naturally occurring protease cleavage site that replaces the wild-type, natural CMV protease cleavage site.
In another embodiment, the polypeptide having at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID
NO: 8, includes both the amino acid sequence VVDPLPP (SEQ ID NO: 9) and the amino acid sequence RATKAS (SEQ ID NO: 10) as a part of the polypeptide sequence.

GLYCOSYLATION
Although CMV gB may be referred to as a glycoprotein, this nomenclature should not be taken to mean that the polypeptides described herein must be glycosylated when used with the invention. In some embodiments of the invention, the modified gB polypeptide is not glycosylated.
In one preferred embodiment, the gB polypeptide does not include a glycosylation site in the fusion loop regions of the gB polypeptide. That is, the gB
polypeptide does not include a glycan moiety attached to the gB polypeptide within the fusion loop 1 region or within the fusion loop 2 region. The fusion loop 1 region of the VR1814 (SEQ ID NO: 6) is located at residues 153-158. The fusion loop 2 region of the VR1814 (SEQ ID NO: 6) is located at residues 237-244.
In one preferred embodiment, the gB polypeptide does not include a modified amino acid sequence that introduces an N-linked glycosylation site. For example, the modified gB polypeptide does not include an N- glycosylation site comprising N-X-S/T/C motif, wherein X is any amino acid residue.
In another preferred embodiment, the modified gB polypeptide does not include a modified amino acid sequence that introduces an 0-linked glycosylation site. For example, the modified gB polypeptide does not include a carbohydrate moiety linked to the hydroxyl oxygen of serine and threonine. As another example, the modified gB polypeptide does not include an 0- linked glycosylation at tyrosine, 5-hydroxylysine, or 4-hydroxyproline.

UNMODIFIED HYDROPHOBIC SURFACE RESIDUES
In one preferred embodiment, the modified gB polypeptide does not include a deletion or substitution of any one of the amino acid residues selected from the group consisting of 154, 158, 159, 160, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 241, and 242, according to the numbering of SEQ ID NO: 6, or any combination thereof. For example, the gB polypeptide does not include a mutation of any one of the amino acid residues selected from the group consisting of Y160, R236, S238, T239, and Y242, according to the numbering of SEQ ID NO: 6, or any combination thereof.
DELETION OF C-TERMINAL CYTOPLASMIC DOMAIN
The inventors further discovered that eliminating the cytoplasmic tail of HCMV
gB may help to focus the mammal's immune response to the ectodomains of gB.
Additionally, deletion of the cytoplasmic tail may reduce steric occlusion and facilitate access of antibodies to important neutralizing epitopes on gB.
Accordingly, in one embodiment, the native C-terminal cytoplasmic domain of the gB polypeptide (e.g., 100% of the amino acids of the cytoplasmic domain) is deleted. For example, the native C-terminal cytoplasmic domain is not deleted to a varying extent. That is, no less than 100% of the native C-terminal cytoplasmic domain is deleted. As used herein, the cytoplasmic tail of HCMV gB refers the amino acid sequence located at positions 771-905 of SEQ ID NO: 6.

EXPRESSION OF GB
In one aspect, the invention relates to nucleic acids that encode a gB
polypeptide of the invention. Preferably, the recombinant nucleic acid molecule: (a) is not a self- replicating RNA molecule; (b) is not an alphavirus replicon;
(c) does not .. encode any alphavirus nonstructural proteins, such as NSP 1 , NSP2, NSP3 and NSP4; (d) does not contain: an Internal Ribosomal Entry Site (IRES), such as EIV1CV or EV71 ; and/or (e) does not contain a viral 2A site, such as FM DV.
In one embodiment, the invention relates to a nucleic acid sequence that encodes an amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 1.
In one embodiment, the invention relates to a nucleic acid sequence that encodes an amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, .. 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 2.
In one embodiment, the invention relates to a nucleic acid sequence that encodes an amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 3.
In one embodiment, the invention relates to a nucleic acid sequence that encodes an amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 5.
In one embodiment, the invention relates to a nucleic acid sequence that encodes an amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 7.
In one embodiment, the invention relates to a nucleic acid sequence that encodes an amino acid sequence that is at least about 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%. 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 8.
The invention also provides a host cell comprising the nucleic acids described herein. When the host cell is cultured under a suitable condition, the nucleic acids can express a gB polypeptide. Preferably, said gB polypeptide forms a monodispersed trimer. For example, the gB polypeptide of the invention does not form a dimeric association of two trimers. In another exemplary embodiment, the gB
polypeptide of the invention does not form a higher order association of trimers.
Preferably, the monodispersed trimer can be secreted from the host cell.
Most preferably, the host cells are mammalian cells (e.g., human, non-human primate, horse, cow, sheep, dog, cat, and rodent (e.g., hamster). Suitable mammalian cells include, for example, Chinese hamster ovary (CHO) cells, human embryonic kidney cells (HEK-293 cells, typically transformed by sheared adenovirus type 5 DNA), NIH-3T3 cells, 293-T cells, Vera cells, HeLa cells, PERC.6 cells (ECACC deposit number 96022940), Hep G2 cells, MRC-5 (ATCC CCL-171), WI-38 (ATCC CCL-75), fetal rhesus lung cells (ATCC CL-160), Madin-Darby bovine kidney ("MDBK") cells, Madin-Darby canine kidney ("MDCK") cells (e.g., MDCK (NBL2), ATCC CCL34; or MDCK 33016, DSM ACC 2219), baby hamster kidney (BHK) cells, such as BHK21 -F, HKCC cells, and the like.
In a most preferred embodiment, the host cell is EXPI293FTM (ThermoFisher Scientific) human cells, derived from the 293 cell line.
In certain embodiments, the host cell is a CHO cell. In certain embodiments, the polynucleotide encoding the gB polypeptide described herein is stably integrated into the genome of the CHO cell.
Various CHO cell lines are also available from European Collection of Cell Cultures (ECACC), or American Type Culture Collection (ATCC), such as CHO cell lines hCBE1 1 (ATCC PTA-3357Tm), E77.4 (ATCC PTA-3765Tm), hLT-B: R-hG1 CHO #14 (ATCC CRL-1 1965Tm), MOR-CHO- MORAb-003-RCB (ATCC PTA-7552Tm), AQ.C2 clone 11 B (ATCC PTA-3274Tm), AQ.C2 clone 11 B (ATCC
PTA-3274Tm), hsAQC2 in CHO-DG44 (ATCC PTA-3356Tm), xr55 (ATCC CRL-2348Tm), CHO-K1 (ATCC CCL-61TM), Led [originally named Pro-5WgaRI3C]
(ATCCO CRL-1735Tm), Pro-5 (ATCCO CRL-1781Tm), ACY1 -E (ATCC 65421TM), ACY1 -E (ATCC 65420Tm), pgsE-606 (ATCC CRL-2246Tm), CHO-CD36 (ATCC
CRL-2092Tm), pgsC- 605 (ATCC CRL-2245Tm), MC2/3 (ATCC CRL-2143Tm), CHO-ICAM-1 (ATCC CRL-2093Tm), and pgsB-618 (ATCC CRL-2241Tm). Any one of these CHO cell lines may be used.
Other commercially available CHO cell lines include, e.g., FREESTYLETm CHO-S Cells and Flp-InTm-CHO Cell Line from Life Technologies.

Alternative suitable host cells may also include, for example, insect cells (e.g., Aedes aegypti, Auto grapha califomica, Bombyx mori, Drosophila melanogaster, Spodoptera frugiperda, and Trichoplusia ni), mammalian cells (e.g., human, non-human primate, horse, cow, sheep, dog, cat, and rodent (e.g., hamster)), avian cells (e.g., chicken, duck, and geese), bacteria (e.g., E. coli, Bacillus subtilis, and Streptococcus spp.), yeast cells (e.g., Saccharomyces cerevisiae, Candida albicans, Candida maltosa, Hansenual polymorpha, Kluyveromyces fragilis, Kluyveromyces lactis, Pichia guillerimondii, Pichia pastoris, Schizosaccharomyces pombe and Yarrowia lipolytica), Tetrahymena cells (e.g., Tetrahymena thermophila) or combinations thereof.
Suitable insect cell expression systems, such as baculovirus systems, are known to those of skill in the art. Materials and methods for baculo virus/insect cell expression systems are commercially available. Suitable insect cells include, for example, Sf9 cells, Sf21 cells, Tn5 cells, Schneider S2 cells, and High Five cells (Invitrogen)).
In a most preferred embodiment, the host cell is a mammalian cell, not an insect cell. Accordingly in a preferred embodiment, the polypeptide does not have an insect cell pattern of glycosylation.
In certain embodiments, the recombinant nucleic acids are codon optimized for expression in a selected prokaryotic or eukaryotic host cell.
To facilitate replication and expression, the nucleic acids can be incorporated into a vector, such as a prokaryotic or a eukaryotic expression vector.
Exemplary vectors include plasmids that are able to replicate autonomously or to be replicated in a host cell. Typical expression vectors contain suitable promoters, enhancers, and terminators that are useful for regulation of the expression of the coding sequence(s) in the expression construct. The vectors may also comprise selection markers to provide a phenotypic trait for selection of transformed host cells (such as conferring resistance to antibiotics such as ampicillin or neomycin).
Also provided herein is a process of producing cytomegalovirus (CMV) gB
polypeptide comprising: (i) culturing the host cell described herein under a suitable condition, thereby expressing said gB polypeptide, or immunogenic fragment thereof;

and (ii) harvesting said gB polypeptide, or immunogenic fragment thereof, from the culture.
In one embodiment, the gB polypeptide described herein is purified. The gB
polypeptide can be purified using any suitable methods, such as HPLC, various types of chromatography (such as hydrophobic interaction, ion exchange, affinity, chelating, and size exclusion), electrophoresis, density gradient centrifugation, solvent extraction, or the like. As appropriate, the gB polypeptide may be further purified, as required, so as to remove substantially any polypeptides which are also secreted in the medium or result from lysis of host cells, so as to provide a product which is at least substantially free of host debris, e.g., polypeptides, lipids and polysaccharides.
As used herein, a "purified" protein or polypeptide is a protein or polypeptide which is recombinantly or synthetically produced, or produced by its natural host, and has been isolated from other components of the recombinant or synthetic production system or natural host such that the amount of the protein relative to other macromolecular components present in a composition is substantially higher than that present in a crude preparation. In general, a purified protein will be at least about 50% homogeneous and more preferably at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95% or substantially homogeneous.
In another embodiment, the process of purifying the modified gB polypeptide of the invention allows for production of the polypeptide at a purity of >85%, >86%, >87%, >88%, >89%, >90%, >91%, >92%, >93%, >94% or >95% of total protein by mass, as determined by gel electrophoresis. These high levels of purity make the modified gB polypeptide suitable for use as an immunogen in diagnostic applications or as an antigen in immunogenic compositions.

PREPARATION OF ANTIBODIES AGAINST CMV EPITOPES
The immunogenic polypeptides prepared as described above may be used to produce antibodies, both polyclonal and monoclonal. If polyclonal antibodies are desired, a selected mammal (e.g., mouse, rabbit, goat, guinea pig, horse, etc.) is .. immunized with an immunogenic polypeptide bearing a CMV epitope(s). Serum from the immunized animal is collected and treated according to known procedures.
If serum containing polyclonal antibodies to a CMV epitope contains antibodies to other antigens, the polyclonal antibodies can be purified by immunoaffinity chromatography. Techniques for producing and processing polyclonal antisera are known in the art.
Monoclonal antibodies directed against CMV epitopes can also be readily produced by one skilled in the art. The general methodology for making monoclonal antibodies by hybridomas is known. Immortal antibody-producing cell lines can be created by cell fusion, and also by other techniques such as direct transformation of B lymphocytes with oncogenic DNA, or transfection with Epstein-Barr virus.
Panels of monoclonal antibodies produced against CMV epitopes can be screened for various properties; i.e., for isotype, epitope affinity, etc.
Antibodies, both monoclonal and polyclonal, which are directed against CMV
epitopes are particularly useful in diagnosis, and those which are neutralizing are useful in passive immunotherapy. Monoclonal antibodies, in particular, may be used to raise anti-idiotype antibodies.
IMMUNOASSAY AND DIAGNOSTIC KITS
Both the recombinant polypeptides which react immunologically with serum containing CMV antibodies, and the antibodies raised against these recombinant polypeptides, are useful in immunoassays to detect the presence of CMV
antibodies, or the presence of the virus, in biological samples, including for example, blood or serum samples. Design of the immunoassays is subject to a great deal of variation, and a variety of these are known in the art. For example, the immunoassay may utilize the polypeptide having the sequence set forth in SEQ ID NO: 2.
Alternatively, the immunoassay may use a combination of viral antigens derived from the gB
polypeptides described herein. It may use, for example, a monoclonal antibody directed towards one modified gB polypeptides described herein, a combination of monoclonal antibodies directed towards the modified gB polypeptides described herein, monoclonal antibodies directed towards different viral antigens, polyclonal antibodies directed towards the modified gB polypeptides described herein, or polyclonal antibodies directed towards different viral antigens. Protocols may be based, for example, upon competition, or direct reaction, or may be sandwich type assays. Protocols may also, for example, use solid supports, or may be by immunoprecipitation. Most assays involve the use of labeled antibody or polypeptide;
the labels may be, for example, fluorescent, chemiluminescent, radioactive, or dye molecules. Assays which amplify the signals from the probe are also known;
examples of which are assays which utilize biotin and avidin, and enzyme-labeled and mediated immunoassays, such as ELISA assays.
Kits suitable for immunodiagnosis and containing the appropriate labeled reagents are constructed by packaging the appropriate materials, including the recombinant polypeptides of the invention containing CMV epitopes or antibodies directed against epitopes in suitable containers, along with the remaining reagents and materials required for the conduct of the assay, as well as a suitable set of assay instructions.
The polynucleotide probes can also be packaged into diagnostic kits.
Diagnostic kits include the probe DNA, which may be labeled; alternatively, the probe DNA may be unlabeled and the ingredients for labeling may be included in the kit. The kit may also contain other suitably packaged reagents and materials needed for the particular hybridization protocol, for example, standards, as well as instructions for conducting the test.

COMPOSITIONS AND METHODS OF TREATMENT
The invention relates to compositions and methods of treatment using the cytomegalovirus gB polypeptide described herein, or a nucleic acid encoding such gB polypeptide described herein. For example, the polypeptide of the invention can be delivered directly as a component of an immunogenic composition.
Alternatively, nucleic acids that encode the gB polypeptide of the invention can be administered to produce the CMV protein or immunogenic fragment in vivo. Certain preferred embodiments, such as protein formulations, recombinant nucleic acids (e.g., DNA, RNA, self-replicating RNA, or any variation thereof) and viral vectors (e.g., live, single-round, non-replicative assembled virions, or otherwise virus-like particles, or alphavirus VRP) that contain sequences encoding gB polypeptides are further described herein and may be included in the composition.
In one aspect, the invention provides an immunogenic composition comprising the recombinant CMV gB polypeptide described herein. The immunogenic composition can include additional CMV proteins, such as gO, gH, gL, pUL128, pUL130, pUL131 , an immunogenic fragment thereof, or a combination thereof. For example, the gB polypeptide can be combined with CMV pentameric complex comprising: gH or a pentamer-forming fragment thereof, gL or a pentamer-forming fragment thereof, pUL128 or a pentamer-forming fragment thereof, pUL130 or a pentamer-forming fragment thereof, and pUL131 or a pentamer-forming fragment thereof. The gB polypeptide of the invention can also be combined with CMV trimeric complex comprising: gH or a trimer-forming fragment thereof, gL
or a trimer-forming fragment thereof, and g0 or a trimer- forming fragment thereof.
The immunogenic composition may include an adjuvant. Exemplary adjuvants to enhance effectiveness of the composition include: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc.; (2) oil-in-water emulsion formulations (with or without other specific adjuvants such as muramyl peptides (see below) or bacterial cell wall components), such as for example (a) MF59 (PCT Publ. No. WO 90/14837), containing 5% Squalene, 0.5% TWEEN 80, and 0.5% Span 85 formulated into submicron particles using a microfluidizer, (b) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer L121 , and thr-MDP either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RIBI TM adjuvant system (RAS), (Ribi lmmunochem, Hamilton, Mont.) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL+CWS (DetoxTm); (3) saponin adjuvants, such as QS-21, STIMU LON TM (Cambridge Bioscience, Worcester, Mass.), which may be used or particles generated therefrom such as ISCOMs (immunostimulating complexes);
(4) Complete Freunds Adjuvant (CFA) and Incomplete Freunds Adjuvant (I FA); (5) cytokines, such as interleukins (IL-1 , IL-2, etc.), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TN F), etc.; and (6) other substances that act as adjuvants to enhance the effectiveness of the composition. In a preferred embodiment, the adjuvant is a saponin adjuvant, namely QS-21.
Each of the immunogenic compositions discussed herein may be used alone or in combination with one or more other antigens, the latter either from the same viral pathogen or from another pathogenic source or sources. These compositions may be used for prophylactic (to prevent infection) or therapeutic (to treat disease after infection) purposes.
In one embodiment, the composition may include a "pharmaceutically acceptable carrier," which includes any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition.
Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles. Such carriers are well known to those of ordinary skill in the art.
Additionally, these carriers may function as adjuvants. Furthermore, the antigen may be conjugated to a bacterial toxoid, such as a toxoid from diphtheria, tetanus, .. cholera, H. pylori, and etc. pathogens.
In one embodiment, the composition includes a diluent, such as water, saline, glycerol, ethanol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
The compositions described herein may include an immunologically effective amount of the polypeptide, as well as any other of the above-mentioned components, as needed. By "immunologically effective amount," it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for eliciting an immune response. The immune response elicited may be sufficient, for example, for treatment and/or prevention and/or reduction in incidence of illness, infection or disease. This amount varies depending upon the health and physical condition of the individual to be treated, the taxonomic group of individual to be treated (e.g., nonhuman primate, primate, etc.), the capacity of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
The composition may be administered parenterally, e.g., by injection, either subcutaneously or intramuscularly. Additional formulations suitable for other modes of administration include oral and pulmonary formulations, suppositories, and transdermal applications. Oral formulations may be preferred for certain viral proteins. Dosage treatment may be a single dose schedule or a multiple dose schedule. The immunogenic composition may be administered in conjunction with .. other immunoregulatory agents.
In another aspect, the invention provides a method of eliciting an immune response against cytomegalovirus (CMV), comprising administering to a subject in need thereof an immunologically effective amount of a modified CMV gB
polypeptide and/or an immunogenic composition described herein, which comprises the proteins, DNA molecules, RNA molecules (e.g., self-replicating RNA molecules), or VRPs as described above. In certain embodiments, the immune response comprises the production of neutralizing antibodies against CMV.
The immune response can comprise a humoral immune response, a cell-mediated immune response, or both. In some embodiments an immune response is .. induced against each delivered CMV protein. A cell-mediated immune response can comprise a Helper T-cell (Th) response, a CD8+ cytotoxic T-cell (CTL) response, or both. In some embodiments the immune response comprises a humoral immune response, and the antibodies are neutralizing antibodies.
Neutralizing antibodies block viral infection of cells. CMV infects epithelial cells and also fibroblast cells. In some embodiments the immune response reduces or prevents infection of both cell types. Neutralizing antibody responses can be complement-dependent or complement- independent. In some embodiments the neutralizing antibody response is complement- independent. In some embodiments the neutralizing antibody response is cross-neutralizing; i.e., an antibody generated against an administered composition neutralizes a CMV virus of a strain other than the strain used in the composition.
The modified CMV gB polypeptide and/or immunogenic composition described herein may also elicit an effective immune response to reduce the likelihood of a CMV infection of a non-infected mammal, or to reduce symptoms in an infected mammal, e.g., reduce the number of outbreaks, CMV shedding, and risk of spreading the virus to other mammals.
For example in one aspect, the modified CMV gB polypeptide and/or immunogenic composition described herein reduces viral shedding in a mammal.
The term "viral shedding" is used herein according to its plain ordinary meaning in medicine and virology and refers to the production and release of virus from an infected cell. In some embodiments, the virus is released from a cell of a mammal.
In some embodiments, virus is released into the environment from an infected mammal. In some embodiments the virus is released from a cell within a mammal.
In one aspect, the invention relates to a method for reducing CMV viral shedding in a mammal. The method includes administering the modified CMV gB
polypeptide and/or immunogenic composition described herein to the mammal that is infected with or is at risk of a CMV infection. In one embodiment, the reduction in CMV viral shedding in a mammal is as compared to the viral shedding in mammals that were not administered the modified CMV gB. In another embodiment, the reduction in CMV viral shedding in a mammal is as compared to the viral shedding following an administration of a CMV pentamer alone or following an administration of a CMV pentamer in the absence of the modified CMV gB.
In one embodiment, the challenge cytomegalovirus strain is a human CMV
strain. In one embodiment, the challenge cytomegalovirus strain is homologous to the CMV strain from which the modified CMV gB polypeptide is derived. In another embodiment, the challenge cytomegalovirus strain is homologous to the VR1814 CMV strain.
In one embodiment, the challenge cytomegalovirus strain is a human CMV
strain that is heterologous to the CMV strain from which the modified CMV gB
polypeptide is derived. In another embodiment, the challenge cytomegalovirus strain is a human CMV strain that is heterologous to the VR1814 CMV strain. In another embodiment, the challenge cytomegalovirus strain is the VR1814 CMV strain.

In another embodiment, the challenge cytomegalovirus strain is a rhesus CMV strain homologous to the macacine herpesvirus 3 isolate 21252 CMV strain.
In another embodiment, the challenge cytomegalovirus strain is the macacine herpesvirus 3 isolate 21252 CMV strain.
A useful measure of antibody potency in the art is "50% neutralization titer."
Another useful measure of antibody potency is any one of the following: a "60%

neutralization titer"; a "70% neutralization titer"; a "80% neutralization titer"; and a "90% neutralization titer." To determine, for example, a 50% neutralizing titer, serum from immunized animals is diluted to assess how dilute serum can be yet retain the ability to block entry of 50% of infectious viruses into cells. For example, a titer of 700 means that serum retained the ability to neutralize 50% of infectious virus after being diluted 700-fold. Thus, higher titers indicate more potent neutralizing antibody responses. In some embodiments, this titer is in a range having a lower limit of about 200, about 400, about 600, about 800, about 1000, about 1500, about 2000, about 2500, about 3000, about 3500, about 4000, about 4500, about 5000, about 5500, about 6000, about 6500, or about 7000. The 50%, 60%, 70%, 80%, or 90% neutralization titer range can have an upper limit of about 400, about 600, about 800, about 1000, about 1500, about 2000, about 2500, about 3000, about 3500, about 4000, about 4500, about 5000, about 5500, about 6000, about 6500, about 7000, about 8000, about 9000, about 10000, about 11000, about 12000, about 13000, about 14000, about 15000, about 16000, about 17000, about 18000, about 19000, about 20000, about 21000, about 22000, about 23000, about 24000, about 25000, about 26000, about 27000, about 28000, about 29000, or about 30000. For example, the 50% neutralization titer can be about 3000 to about 6500. "About"
means plus or minus 10% of the recited value. Neutralization titer can be measured as described in the specific examples, below.
An immune response can be stimulated by administering proteins, DNA
molecules, RNA molecules (e.g., self-replicating RNA molecules), or VRPs to an individual, typically a mammal, including a human. In some embodiments the immune response induced is a protective immune response, i.e., the response reduces the risk or severity of or clinical consequences of a CMV infection.
Stimulating a protective immune response is particularly desirable in some populations particularly at risk from CMV infection and disease. For example, at-risk populations include solid organ transplant (SOT) patients, bone marrow transplant patients, and hematopoietic stem cell transplant (HSCT) patients. VRPs can be administered to a transplant donor pre-transplant, or a transplant recipient pre-and/or post-transplant. Because vertical transmission from mother to child is a common source of infecting infants, administering VRPs to a woman who is pregnant or can become pregnant is particularly useful.
Any suitable route of administration can be used. For example, a composition can be administered intramuscularly, intraperitoneally, subcutaneously, or transdermally. Some embodiments will be administered through an intra-mucosal route such as intra-orally, intra- nasally, intra-vaginally, and intra-rectally.
Compositions can be administered according to any suitable schedule.
Also provided herein is a method of inhibiting cytomegalovirus (CMV) entry into a cell, comprising contacting the cell with the immunogenic composition described herein.
The invention will be further described by reference to the following, non-limiting, examples.

EXAMPLES
The following Examples illustrate embodiments of the invention.
EXAMPLE 1: Description of the gB705 expression construct Pfizer's expression construct of HCMV gB705 (SEQ ID NO: 1) is based on the VR1814 gB sequence (GenBank# A0Z79977.1) that contains amino acids 1-705. A
6xHis affinity tag is optionally included at the C-terminus in a pLH115 vector (Pfizer).
Pfizer's gB705 contains four mutations ((Y155G/I156H/H157R/VV240A) as well as mutations (R456A/R459A) in the furin-like cleavage site as illustrated in FIG.
1, wherein SS represents a signal sequence; D1 represents the Domain I; FL
represents a fusion loop; 6h represents a 6xHis tag. FIG. 2 shows the sequence of HCMV gB705 (SEQ ID NO: 2) EXAMPLE 2: Expression of gB705 The initial assessment of gB705 expression was carried out by transfecting the construct DNA into HEK293T or Expi293F cells using Effectene Transfection Reagent (Qiagen) or Expifetamin 293 Transfection kit (Gibco), respectively.
The culture supernatants were harvested -48 hours after transfection. The gB705 protein was pulled down from the culture supernatant of transfected cells using a MAGNEHISTM Protein Purification System (Promega) and was analyzed on 4-12%
Bis-Tris protein gel (NUPAGEO NOVEXO) under reducing condition. Presence of the target proteins was detected by Western blot using anti-HCMV polyclonal antibody (CytoGam). The result showed that recombinant gB705 existed as a single protein band with molecular weight about 120 kDa (FIG. 3). This apparent molecular weight is higher than that predicted from the peptide sequence of residues 25-705 [of SEQ ID NO: 1] from the mature gB705 polypeptide, indicating that the gB705 protein is heavily glycosylated. The gB705 protein is expressed efficiently as a homogeneous protein in transfection of HEK293 or Expi293F cells.

EXAMPLE 3: Recombinant gB705 contains intact neutralizing epitopes recognized by human anti-HCMV gB monoclonal antibodies (mAbs) To evaluate the structural integrity of recombinant gB, a panel of five neutralizing human anti-HCMV gB mAbs were tested for the binding of gB705 (SEQ
ID NO: 2). Briefly, the culture supernatants from gB705 transfected Expi293F
cells were added on a pre-blocked HISGRABTM Nickel Coated Plate (Pierce) and incubated for 1 hour at room temperature. After wash, serial diluted antibody solutions were added to the plate and incubated for 1 hour at room temperature, followed by addition of HRP-conjugated anti-human IgG secondary antibody.
After 1 hour incubation, the plate was washed and the peroxidase substrate TMB was added to be read by a plate reader when the color was developed. The results showed all five human anti-gB mAbs bound efficiently to gB705 (FIG. 4A).
Recombinant gB705 binds to three anti-AD5 mAbs including 2B11, 5F1 and 4H9 (each described in Table 1 of US patent publication no. 20160280770 and Table 1 of WIPO publication no. W02010007533), whereas recombinant gB proteins from a commercial source (Sino gB, from Sino Biologicals, Inc.) did not (FIG. 4B).
Similarly, these three mAbs did not bind to a recombinant gB protein expressed in insect cells from RedBiotech, Inc. (data not shown). The recombinant gB
protein from the commercial source and from RedBiotec do not include any one of the following mutations: Y155G, I156H, H157R, and W240A, according to the numbering of SEQ ID NO: 6. Without being bound by theory, the inventive combination of mutations Y155G, I156H, H157R, and W240A include deletions of the cytoplasmic tail and mutations in hydrophobic fusion loops of gB. These mutations appear to relieve steric hindrance that potentially would have otherwise blocked antibody access to these sites in an unmodified gB protein or in a protein that does not include the inventive combination of mutations.
The 2B11 mAb has a sequence set forth in SEQ ID NO: 367 for the heavy chain and SEQ ID NO: 368 for the light chain; 5F1 mAb has a sequence set forth in SEQ ID NO: 290 for the heavy chain and SEQ ID NO: 291 for the light chain; and 4H9 mAb has a sequence set forth in SEQ ID NO: 308 for the heavy chain and SEQ
ID NO: 309 for the light chain, as disclosed in US patent publication no.
20160280770 and WIPO publication no. W02010007533, each sequence of which is incorporated by reference herein).

FIG. 4A Binding of human anti-HCMV gB monoclonal antibodies to gB705 protein. Culture supernatants from gB705 transfected Expi293F cells were captured on a pre-blocked HISGRABTM Nickel Coated Plate (Pierce) and incubated serially diluted monoclonal antibodies followed by detection with HRP conjugated anti-s human Ig secondary antibody (ELN# 00710043-0177). FIG. 4B Binding of human anti-HCMV gB monoclonal antibodies to Sino gB protein (gB from Sino Biologicals, Inc.) (ELN# 00710043-0177).

EXAMPLE 4: Scale-up production and purification of gB705 The gB705 construct was transiently transfected into 1.8 liters of Expi293F
cells and about 95 mg of protein was purified from the conditioned media collected on day 4 through a series of processes of diafiltration, Ni-sepharose and size exclusion chromatography. The protein was analyzed on SDS-PAGE and exhibited a single band under reducing condition, as detected by Coomassie blue staining (FIG.
5).
Under non-reducing conditions, there were higher molecular weight bands that may be corresponding to a trimeric form of the protein. FIG. 5 shows an SDS-PAGE
of gB705 under reducing and non-reducing conditions (ELN#00709755-0086).

EXAMPLE 5: Structural characterization of recombinant gB705 expressed in Expi293F cells: Oligomer formation and stability Postfusion gB protein is in a trimer form. To characterize whether the gB705 forms higher order of oligomers, a series of experiments were carried out as summarized .. below.
Singular and oligomeric trimers of gB705 with characteristics of post-fusion gB observed by transmission electron microscopy Recombinant gB705 protein was subjected to electron microscopy analysis after negative staining. The image (FIG. 16) shows that the protein forms a singular trimer as well as larger oligomeric complexes (that is, associations between trimers). The singular trimer of gB705 exhibited strong similarity in general morphology with the postfusion gB structure as published in Burke et al., PLoS Pathog. 2015 Oct;
11(10).
In contrast to the singular trimer formed by the inventive gB705, a modified gB
protein, termed "gB706," which does not include the inventive combination of fusion .. loop mutations, forms a dimer and trimer of trimers, as discussed in in Sharma et al., Virology. 2013 Jan 20;435(2):239-49, namely on page 243 and in Figure 20. More specifically, the gB706 protein of Sharma et al. is described as encoding the mature ectodomain of HCMV (strain AD169) gB, residues 25-706, with a signal sequence.

Sharma et al. further describe gB706 as lacking the hydrophobic membrane-proximal region, the transmembrane region, and the cytoplasmic domain of gB.
Multiple species of gB705 observed in velocity analytical ultracentrifugation analysis The purified gB705 protein (SEQ ID NO: 2) was also subjected to velocity analytical ultracentrifugation analysis and the result showed that the protein formed multiple peaks corresponding to the sizes of singular trimer or larger / high-order complexes (FIG. 6). FIG. 6 velocity analytical ultracentrifugation analysis of gB705 (ELN#
00708337-0110).

Transforming recombinant gB705 to homogeneous singular trimer by treatment of ethylenediaminetetraacetic acid (EDTA) To assess the stability of the gB705 higher order oligomers, the gB705 protein was subjected to various treatment including surfactants, chaotropes, heat, reductant and EDTA (data not shown). Among these conditions, it was observed that EDTA
can break down the high molecular weight oligomers to homogenous monodispersed trimers. This was confirmed by velocity sedimentation analysis as shown in FIG
7.
This homogenous material establishes a strong base entity for studying its structural characteristics and its potential for change under biologically relevant conditions.
FIG. 7 shows Velocity sedimentation analysis of 10 mM EDTA-treated gB705 ( ELN#
00708337-0110).
In the structure of a postfusion HCMV gB protein, a calcium ion (Ca++) was observed in the core of the central coiled coil, coordinated by residue D508.
To evaluate whether Ca++ ion has an effect on the overall shape of the gB705 after EDTA treatment, sedimentation velocity analysis was performed on the EDTA-treated gB705 at pH 5.2 with and without adding back 10 mM Ca++ ion. The result showed that adding back Ca++ ion had no impact on hydrodynamic properties of gB705 at pH 5.2 (data not shown). Furthermore, adding back Ca++ ion did not cause the protein to form high molecular weight oligomers seen in the non-EDTA-treated gB705 protein. In contrast, adding back Ni+ or Cu++ ions led to formation of high-order oligomeric complexes of gB705 trimer (data not shown). Taken together, oligomer formation of gB705 trimer may be a result of the existence of the 6xHis affinity tag, rather than due to some unknown intrinsic properties of gB705 trimer.

EXAMPLE 6: Structural characterization of gB705 trimer at different pH
conditions:
To evaluate the effect of pH on the HCMV gB705 protein, EDTA-treated gB705 protein was dialyzed into buffers at pH 5.2 and pH 8.7 and subjected to a series of .. biophysical characterization experiments to determine the changes, if any, in the secondary, tertiary and quaternary structure or hydrodynamic properties.
Sedimentation velocity analysis Sedimentation velocity analysis was performed on EDTA-treated gB705 at pH 5.2 and pH 8.7 (Fig. 8). No significant difference in migration or apparent molecular weight of the protein at different pHs was observed. FIG. 8A-B Sedimentation velocity analysis of EDTA-treated gB705 at pH 5.2 (A) and pH 8.7 (B).
Cryo electron microscopy analysis Cryo electron microscopy analysis was performed on EDTA-treated gB705 proteins. The two images were collected on a Tecnia F20 electron microscope at the magnification of x50,000 .. using low dose beam condition on a cryo frozen grid. The images showed that the gB705 was in monodispersed trimer form at either pH5.2 or pH 8.7 (FIG. 8C).
Far UV CD and intrinsic fluorescence spectroscopy Both the Far UV CD (FIG. 9A) and intrinsic fluorescence spectroscopy (FIG. 9B) analyses suggest little change in the secondary structure of gB705 at pH 5.2 and pH
.. 8.7. Fig. 9A Far UV CD analysis; FIG. 9B intrinsic fluorescence spectroscopy (ELN#
00708337-0115, 0117) Change in tertiary structure of gB705 trimer at different pHs detected by near-UV CD spectroscopy Near-UV CD analysis was also performed to determine whether there is any difference in the tertiary structure of gB705 trimer at pH 5.2 and pH 8.7. The gB705 trimer exhibited differences in the spectroscopy profile under the two pH
conditions (Fig. 10). This difference was strictly pH-dependent, because it was consistently observed when two different buffers were used at both pH 5.2 and pH 8.7. The lower fluorescence of gB705 in the normalized near-UV CD spectrum at pH 5.2 may be indicative of looser packing of the aromatic residues in gB705 trimer under acidic conditions. In short, change in environmental pH condition has a noticeable influence on the tertiary structure of gB705 trimer. FIG. 10 Near UV CD spectroscopy analysis of gB705 at pH 5.2 and pH 8.7. Buffer 1, 50 mM citrate, 100 mM phosphate;
Buffer 2, 50 mM Na0Ac; Buffer 3, 100 mM Na carbonate/bicarbonate; Buffer 4, 50 mM Na borate (ELN# 00708337-0115, 0117).

Difference in thermodynamic melting at high and low pH conditions DSC analysis was performed to study the thermodynamic properties of gB705 at pH
5.2 and pH 8.7 (Fig. 11). The gB705 protein underwent a two-phase transition at pH
5.2, with a transitional melting temperature of 81.0 C. In stark contrast, the same protein prep demonstrated a three-phase transition at pH 8.7, with two transitional melting points at 72 C and 78 C respectively. The observed difference here indicates that there is significant structural difference, in terms of biophysical stability, in the gB705 trimer at pH 5.2 versus pH 8.7. FIG. 11 A-B: DSC analysis of gB705 at (FIG.
11A) pH 5.2 and (FIG. 11B) pH 8.7. At pH 8.7, the observed melting curve (in block line) was fit to the 4-state unfolding model and resolved to a three-phase transition pattern (in red lines) using the software provided by the DSC manufacturer.
(ELN#
00708337-0115, 0117).
Increase in exposure of hydrophobic elements in gB705 trimer at acidic condition.
Difference in ANS staining of gB705 at different pH
ANS is a fluorescent dye that binds to hydrophobic surfaces of protein, resulting in increased fluorescence intensity and shift in the emission maximum towards lower wavelengths. It was observed that ANS fluorescence intensity of gB705 was increased and its maximal emission was shifted to shorter wavelength of UV at pH
5.2, relative to those at pH 8.7 or pH 7.4 (FIG. 12A). These data are consistent with that hydrophobic surfaces of gB705 trimer are more exposed under acidic pH
than under neutral or basic pH conditions.
The exposure of hydrophobic surfaces of gB705 trimer at different pH
conditions is reversible To assess whether the difference in ANS binding ability of gB705 at different pHs is reversible or not, the gB705 protein at pH 5.2 was dialyzed into pH 8,7 buffer and vice versa, and then subjected to ANS analysis. The results showed that the protein gained ANS binding when shifted from high pH to low pH (FIG. 12B) and lost ANS

binding when shifted from low pH to high pH (FIG. 12C). Therefore, the observed difference in ANS binding to gB705 are reversible.
FIG. 12A-C: (A) ANS binding of gB705 at pH 5.2, pH 7.4 and pH 8.7 (ELN#
00708337-0115, 0117); (B) ANS binding of gB705 at pH 8.7 and 5.2 and shifted from pH 8.7 to pH 5.2; (C) ANS binding of gB705 at pH 8.7 and 5.2 and shifted from pH
5.2 to pH 8.7 (ELN# 00708329-0153) Accordingly, a HCMV gB705 protein was produced. The protein contains the ectodomain of the VR1814 gB and mutations in the fusion loops and furin cleavage site. This protein may be in the postfusion conformation under neutral pH. The protein forms various oligomers after purification but can be transformed into homogenous singular trimer upon EDTA treatment.
No significant difference was observed in the secondary structure of EDTA-treated gB705 at pH 5.2 and pH 8.7, based on assays of far-UV CD spectroscopy, intrinsic fluorescence as well as sedimentation velocity analysis. However, significant differences in conformation of gB705 trimer are observed with various assays.
Data from near-UV spectroscopy revealed significant difference in tertiary structure in gB705 trimer between pH 5.2 and pH 8.7. Data on extrinsic fluorescence after ANS
staining suggest that the protein exhibits more exposed hydrophobic surfaces at pH
5.2, compared with at pH 8.7 and pH 7.2. The thermodynamic melting pattern of gB705 was significantly different between pH 5.2 and pH 8.7. Finally, the ANS
staining analysis demonstrated that the conformational states of gB705 trimer exist in a reversible equilibrium, a property similar to VSV-G as a prototypic type Ill viral fusogen.
The gB705 protein exhibits a pH-dependent transition of conformation, which is a unique property and is biologically important. A non-postfusion conformation of gB
(for example, gB705 at pH 8.7) may be a presented in a specific chemical/physical condition or by stabilizing its conformation with treatment of crosslinking agents.
Such an immunogen may assume conformation state(s) that have a potential to elicit more robust nAb response than for example, recombinant gB protein in the postfusion conformation. Moreover, the pH-dependent transition of tertiary conformation of the gB705 trimer may be useful for improving solubility of the gB
immunogen in an immunogenic composition, for improving stability of the immunogen, for improving binding of the immunogen to aluminum or other adjuvants, and/or for improving immunization of a vaccine comprising the immunogen, as compared, for example, to a recombinant gB protein that does not exhibit a pH-dependent transition of tertiary conformation, and/or as compared to a recombinant gB protein that does not include the following mutations Y155G, I156H, H157R, and W240A.

>HCMV VR1814 gB705 M ESR IWC LVVCVN LC I VCLGAVVSSSSTSHATSSA H NGS HTSRTTSAQTRSVSSQ H
VTSSEAVSH RAN ETIYNTTLKYG DVVGVNTTKYPYRVCSMAQGTDLI RFERNIVCTP
MKPI NEDLDEGIMVVYKRNIVAHTFKVRVYQKVLTFRRSYAGH RTTYLLGSNTEYVA
PPMWEIHHINRHSQCYSSYSRVIAGTVFVAYHRDSYEN KTMQLM LDDYSNTHSTR
YVTVKDQWHSRGSTALYRETCN LNCMVTITTARSKYPYHFFATSTGDVVDISPFYN
GTNRNTSYFGENADKFFI FPNYTIVSDFGRANSAPETHRLVAFLERADSVISWDIQD
EKNVTCQLTFWEASERTI RSEAEDSYHFSSAKMTATFLSKKQEVNMSDPVLDCVR
DQALNKLQQI FNASYNQTYEKYG NVSVFETTGGLVVFWQG I KQKSLLELERLANSS
GVNSTRATKASTGNTTTLSLESESVRNVLYAQLQFTYDTLRSYI NRALAQIAEAWCV
DQRRTLEVFKELSKI NPSAI LSAIYNKPIAARFMGDVLGLASCVTI NQTSVKVLRDMN
VKESPGRCYSRPVVI FN FVNSSYVQYGQ LGEDN El LLGNHRTEECQFPSLKI FIAGN
SAYEYVDYLFKRMI DLSSISTVDSMIALDI DPLENTDFRVLELYSQKELRSSNVFDLE
El MR EFNSYKQRVKYVEDKVVDPLPP (SEQ ID NO: 1) >HCMV VR1814 gB705 (w/o Met) ESR IWC LVVCVN LC I VC LGAVVSSSSTSHATSSA H NGSH TSRTTSAQT RSVSSQ HV
TSSEAVSH RAN ETIYNTTLKYG DVVGVNTTKYPYRVCSMAQGTD LI RFERNIVCTPM
KPI NEDLDEGIMVVYKRN IVAHTFKVRVYQKVLTFRRSYAGHRTTYLLGSNTEYVAP
PMWEI HHINRHSQCYSSYSRVIAGTVFVAYHRDSYENKTMQLMLDDYSNTHSTRY
VTVKDQWHSRGSTALYRETCNLNCMVTITTARSKYPYHFFATSTGDVVDISPFYNG
TN RNTSYFGENADKF Fl FPNYTIVSDFGRANSAPETHRLVAFLERADSVISWDIQDE
KNVTCQLTFWEASERTI RSEAEDSYHFSSAKMTATFLSKKQEVNMSDPVLDCVRD
QALNKLQQI FNASYN QTYEKYGNVSVFETTGG LVVFWQG I KQKSLLELERLANSSG
VNSTRATKASTGNTTTLSLESESVRNVLYAQLQFTYDTLRSYI NRALAQIAEAWCVD
QRRTLEVFKELSKINPSAILSAIYNKPIAARFMGDVLGLASCVTI NQTSVKVLRDM NV
KESPGRCYSRPVVI FNFVNSSYVQYGQLGEDN El LLGNHRTEECQFPSLKI FIAGNS
AYEYVDYLFKRMI DLSSISTVDSMIALDI DPLENTDFRVLELYSQKELRSSNVFDLEEI
MREFNSYKQRVKYVEDKVVDPLPP (SEQ ID NO: 2) >HCMV VR1814 gB705 (residues 25-705 OF SEQ ID NO: 1) SSSTSHATSSAHNGSHTSRTTSAQTRSVSSQHVTSSEAVSH RAN ETIYNTTLKYGD
VVGVNTTKYPYRVCSMAQGTD LI RFE RN IVCTPM KPI N EDLDEG I MVVYKRN IVAHT
FKVRVYQKVLTFRRSYAGHRTTYLLGSNTEYVAPPMWEIHHIN RHSQCYSSYSRVI
AGTVFVAYHRDSYENKTMQLMLDDYSNTHSTRYVTVKDQWHSRGSTALYRETCN
LNCMVTITTARSKYPYHFFATSTGDVVDISPFYNGTNRNTSYFGENADKFFI FPNYTI
VSDFGRANSAPETH RLVAFLERADSVISWDIQDEKN VTCQLTFWEASERTI RSEAE
DSYHFSSAKMTATFLSKKQEVNMSDPVLDCVRDQALNKLQQI FNASYNQTYEKYG
NVSVFETTGGLVVFWQGI KQKSLLELERLANSSGVNSTRATKASTGNTTTLSLESE
SVRNVLYAQLQFTYDTLRSYIN RALAQIAEAWCVDQRRTLEVFKELSKI NPSAI LSAI
YNKPIAARFMGDVLGLASCVTINQTSVKVLRDMNVKESPGRCYSRPVVI FNFVNSS
YVQYGQLGEDN El LLGNH RTEECQFPSLKI FIAGNSAYEYVDYLFKRM I DLSSISTVD
SMIALDI DPLENTDFRVLELYSQKELRSSNVFDLEEI M REFNSYKQRVKYVEDKVVD
PLPP (SEQ ID NO: 3) > signal sequence MESRIWCLVVCVNLCIVCLGAV (SEQ ID NO: 4) >HCMV VR1814 gB705 with linker VSSSSTSHATSSAH NGSHTSRTTSAQTRSVSSQHVTSSEAVSH RAN ETIYNTTLKY
GDVVGVNTTKYPYRVCSMAQGTDLI RFER NI VCTPMKPI N EDLDEGI MVVYKRN IVA
HTFKVRVYQKVLTFRRSYAGHRTTYLLGSNTEYVAPPMWEI HHINRHSQCYSSYSR
VIAGTVFVAYH R DSYEN KTMQLM LDDYSNTHSTRYVTVKDQWHSRGSTALYRETC
NLNCMVTITTARSKYPYHFFATSTGDVVDISPFYNGTNRNTSYFGENADKFFI FPNY
TIVSDFGRANSAPETH RLVAFLERADSVISWDIQDEKNVTCQLTFWEASERTI RSEA
EDSYHFSSAKMTATFLSKKQEVNMSDPVLDCVRDQALNKLQQI FNASYNQTYEKY
G NVSVFETTGG LVVFWQGI KQKSLLELERLANSSGVNSTRATKASTG NTTTLSLES
ESVRNVLYAQLQFTYDTLRSYI NRALAQIAEAWCVDQRRTLEVFKELSKI NPSAI LSA
IYNKPIAARFMGDVLGLASCVTI NQTSVKVLRDMNVKESPGRCYSRPVVI FNFVNSS
YVQYGQLGEDN El LLGNH RTEECQFPSLKI FIAGNSAYEYVDYLFKRM I DLSSISTVD
SMIALDI DPLENTDFRVLELYSQKELRSSNVFDLEEI M REFNSYKQRVKYVEDKVVD
PLPPGSG (SEQ ID NO: 5) >ACZ79977.1 envelope glycoprotein B [Human betaherpesvirus 5]
MESRIWCLVVCVNLCIVCLGAVVSSSSTSHATSSAHNGSHTSRTTSAQTRSVSSQH
VTSSEAVSHRANETIYNTTLKYGDVVGVNTTKYPYRVCSMAQGTDLIRFERNIVCTP
MKPINEDLDEGIMVVYKRNIVAHTFKVRVYQKVLTFRRSYAYIHTTYLLGSNTEYVAP
PMWEIHHINRHSQCYSSYSRVIAGTVFVAYHRDSYENKTMQLMLDDYSNTHSTRY
VTVKDQWHSRGSTWLYRETCNLNCMVTITTARSKYPYHFFATSTGDVVDISPFYNG
TNRNTSYFGENADKFFIFPNYTIVSDFGRANSAPETHRLVAFLERADSVISWDIQDE
KNVTCQLTFWEASERTIRSEAEDSYHFSSAKMTATFLSKKQEVNMSDPVLDCVRD
QALNKLQQIFNASYNQTYEKYGNVSVFETTGGLVVFWQGIKQKSLLELERLANSSG
VNSTRRTKRSTGNTTTLSLESESVRNVLYAQLQFTYDTLRSYINRALAQIAEAWCVD
QRRTLEVFKELSKINPSAILSAIYNKPIAARFMGDVLGLASCVTINQTSVKVLRDMNV
KESPGRCYSRPVVIFNFVNSSYVQYGQLGEDNEILLGNHRTEECQFPSLKIFIAGNS
AYEYVDYLFKRMIDLSSISTVDSMIALDIDPLENTDFRVLELYSQKELRSSNVFDLEEI
MREFNSYKQRVKYVEDKVVDPLPPYLKGLDDLMSGLGAAGKAVGVAIGAVGGAVA
SVVEGVATFLKNPFGAFTIILVAIAVVIIIYLIYTRQRRLCMQPLQNLFPYLVSADGTTV
TSGNTKDTSLQAPPSYEESVYNSGRKGPGPPSSDASTAAPPYTNEQAYQMLLALA
RLDAEQRAQQNGTDSLDGQTGTQDKGQKPNLLDRLRHRKNGYRHLKDSDEEENV
(SEQ ID NO: 6) EXAMPLE 7:
CMV is a species-specific virus. Just has humans are infected by HCMV, rhesus macaques are infected by rhCMV, which has a biology similar to HCMV.
This provides an animal model system for testing CMV vaccine inventions and technologies in ways that may be impractical or unethical in humans, such as certain infectious challenge experiments. Therefore, rhCMV analogues of HCMV antigens may be made to allow experimental investigation. RhCMV has a gB with a structure and function very similar to those of HCMV gB. A satisfactory correlation exists between effects in rhesus macaques relating to CMV and effects ultimately observed in humans. Accordingly, data from testing in rhesus macaques is reasonably predictive of the response in humans. Following this line reasoning, an analogous construct of HCMV gB705 construct, named as RhCMV gB674, was designed and tested in RhCMV(-) monkeys.
RhCMV gB674 Design Following the same design of HCMV gB705, Pfizer's expression construct of RhCMV
gB674 (SEQ ID NO: 7) was based on gB sequence of RhCMV strain UCD52, which contains amino acids 1-674 of the ectodomains and a 6xHis affinity tag at the C-terminus. In addition, the gB674 protein contains three mutations (F128G, I129H, W213A) in the predicted fusion loops, because a fourth change in HCMV gB705 is pre-existing in the natural UCD52 sequence. Similarly, mutations (R429A/R430T/R432A) were introduced to totally inactivate the furin-like cleavage site, as illustrated.

Amino Acid Sequence:
> RhCMV gB674 1 msknwfpllc asylvvyvai assstgtasa vtpaptentt geiianttlr thevfrvnms 61 kfpyrvcsma qgtdllrfeq nircdsfkpt kedfdegimv vykrdikpyt fkvhiyqkil .. 121 tfrqsysGHr enhlIgfsqe rlavpmwevn yinrInrcyn svvrnvagvt yvnyhkdsyv 181 netmhliedd ysnthsaryv tvkelwhkpg stAlyttscn vncmvtvtta rskypydffv 241 tsggevvdis pfyngsnneh fgenrdkfyi rrnysmvesy grdnaplvah elvafferpd 301 mlmswdivde anntceytfw eqsertirse adetyhftsh smtatfltlk kelnesdsdf 361 dcirdeaner lekifnttyn etyvksgnvs vyetsggliv fwlpvkekai wemqklateh 421 anntnatrAT kAstnsgnst kevlqnvvya qlqftydtlr nyinralrqi aeawckdqkr 481 tlevlkelsk inpsamlsai ydkpiaarfv gdvislarcv evdqnsvqvl rdmhtkekgl 541 cysrpvvlyt fvnsshvqyg qlgedneill grhrteaces pslkifiagn ssyeyvdyly 601 krmipldsis tvdtmisldi dplentdfra lelysrdelr ssnvfdledi mrefntykqr 661 mvhvegkvfd nvpa gsghhhhhh (SEQ ID NO: 7) > RhCMV gB674 sknwfplIcasylvvyvaiassstgtasavtpaptenttgeiianttIrthevfrvnmskfpyrvcsmaqgtdllrfeq nirc dsfkptkedfdegimvvykrdikpytfkvhiyqkiltfrqsysGHrenhlIgfsgerlavpmwevnyinrInrcynsvv r nvagytyvnyhkdsyvnetmhlieddysnthsaryvtvkelwhkpgstAlyttscnvncmvtvttarskypydffvts ggevvdispfyngsnnehfgenrdkfyi rrnysmvesygrdnaplvahelvafferpdm Imswdivdeanntceytf weqsertirseadetyhftshsmtatfltIkkelnesdsdfdcirdeanerlekifnttynetyvksgnvsvyetsggl ivfw I pvkekaiwemqklatehanntnatrATkAstnsgnstkevlqnvvyaql qftydtl rnyi nral rq iaeawckdqkrtl evIkelskinpsamIsaiydkpiaarfvgdvislarcvevdqnsvqvIrdmhtkekgIcysrpvvlytfvnsshvqygq gednei Ilgrhrteacespslkifiagnssyeyvdylykrmi pldsistvdtm isldidplentdfralelysrdelrssnvfdl .. edimrefntykqrmvhvegkvfdnvpagsghhhhhh (SEQ ID NO: 8) > RhCMV gB
MSKNWFPLLCASVLVVYVAIASSSTGTASAVTPAPTENTTGEIIANTTLRTHEVFRVN
MSKF PYRVCSMAQGTDLLR F EQ N I RCDSF KPTKED F DEG I MVVYKR DI KPYTFKVH I
YQKI LTF RQSYSF I R EN H LLGFSQERLAVPMWEVNYI N RLN RCYNSVVRNVAGVTY
.. VNYH KDSYVN ETM H LI EDDYSNTHSARYVTVKELWH KPGSTWLYTTSCNVNCMVT
VTTARSKYPYDFFVTSGGEVVDISPFYNGSN N EH FG EN RDKFYI RRNYSMVESYGR
DNA P LVA H ELVAF F ERP DM LMSWDI VD EAN NTCEYTFWEQSERTI RSEADETYH FT
SHSMTATF LTLKKELN ESDSDF DCI R DEAN ER LEKI FNTTYN ETYVKSGNVSVYETS
GGLIVFWLPVKEKAIWEMQKLATEHANNTNATRRRKRSTNSGNSTKEVLQNVVYA
QLQFTYDTLRNYI N RA LRQ IAEAWCKDQ KRTLEVLKELSKI N PSAM LSA IYDKP IAA R
FVGDVISLARCVEVDQNSVQVLRDM HTKEKGLCYSRPVVLYTFVNSSHVQYGQLG
EDN El LLGRH RTEACESPSLKI FIAGNSSYEYVDYLYKR M I PLDSI STVDTM I SLD I DP L
ENTDFRALELYSRDELRSSNVFDLEDIM REFNTYKQRMVHVEGKVFDNVPAYLRGL
DDM MSGLGSAGKA LGVA I GAVGGAVASFVEGVVG Fl EN PFGSFTVI LFLLAVLGVIY
LI YM RQ KRAYEKP F EH F F PYVVPPTTVKEA PPSYEQSQYEN I KEKAASATKEFSLEE
AYQM LLALQKLDQEKRRKAEADDEDFASNGQSAGFLDRLRN RR RGGYQ KI QN EYE
V (SEQ ID NO: 11) EXAMPLE 8: Analytics of RhCMV gB674 1. SDS-PAGE analysis of gB674 (ELN 00707788-0011) The gB674 protein was transiently expressed in Expi293 cells and purified via the C-terminal 6xHis tag. On SDS-PAGE, the protein exhibited a single band under .. reducing (R) conditions, demonstrating the lack of protease cleavage as designed and >98% purity. Some oligomeric forms were shown under a non-reducing condition. See FIG. 17A.
2. Size Exclusion chromatography (ELN 00707788-0011) The gB674 protein was treated with EDTA after purification and analyzed on Superose-6 column. As observed with HCMV gB705, gB674 protein was eluted as a major peak, consistent with a homogenous individual trimer. See FIG. 17B.
Immunogenicity testing:
RhCMV(-) rhesus monkeys were screened and proven as seronegative for anti-gB by an ELISA test (data not shown). Six animals were immunized intramuscularly with 100 ug of gB674 per dose and 50 pg of RhCMV pentamer in 50 ug of QS-21 adjuvant at week 0, 4 and 8. A group of six animals were injected with a placebo regimen lacking only the gB protein, as controls.
At week 9, anti-gB response was detected in an ELISA using recombinant RhCMV gB lacking a C-terminal 6xHis tag. In so doing, the ELISA avoids detection .. of antibody responses to the 6His tags on the gB and pentamer antigens in the vaccine. Sera from all six animals in the control group (animals C1-C6 in FIG.
14) were negative of anti-gB at a 1:100 dilution on week 9, as expected. The anti-gB
binding titers from the six vaccines (animals V1-V6 in FIG. 14) on week 9 after three doses of vaccination were detected. Thus, recombinant gB674 of RhCMV UCD52 is highly immunogenic. This provides supporting evidence of recombinant gB705 of HCMV as a vaccine antigen.

EXAMPLE 9: Efficacy testing of the Rhesus equivalent of HCMV gB705 Utility of the new design of HCMV post-fusion gB705 was tested by evaluating the protective efficacy of immunization with RhCMV gB674 protein (SEQ ID NO:
7) in an oral RhCMV infectious challenge model in RhCMV(-) rhesus monkeys. The candidate monkeys were screened and proven seronegative for gB by an ELISA
test (data not shown). Six animals in each of three experimental groups were immunized intramuscularly at weeks 0, 4 and 8. All formulations, including the control formulation, had a pH of 7.7 and contained 50 lig of the QS-21 adjuvuant. Six animals in one group of six animals were injected with a placebo formulation, containing no antigen, as negative controls. Six animals in a second group were immunized with 50 lig each of a recombinant RhCMV pentamer complex (containing the gH, gL, pUL128, pUL130, and pUL131 RhCMV antigens). Six animals in a third group were each immunized with 100 lig of gB674 + 50 lig of recombinant RhCMV
pentamer. Two weeks after the third vaccination, all animals were challenged with 8x105 plaque-forming units (PFU) of UCD52 RhCMV viral stock by oral inoculation, and the challenge was repeated weekly for a total of 5 times. This study was designed to demonstrate whether recombinant gB674 would add extra protective efficacy to a vaccine formulation containing recombinant RhCMV pentamer as the sole antigen. Virus infection was monitored by the presence of viremia and by virus shedding in saliva and urine compartments. The presence of virus in these target samples was quantified by a qPCR assay with a limit of quantitation of 125 DNA

copies/ml. As shown in Fig 15 panel A, five oral challenges led to virus shedding in saliva in 5 of 6 animals in the control group. Week 9 and week 15 are one week prior to the first virus challenge and one week after the fifth virus challenge, .. respectively. The same five control animals had viremia and high level of virus shedding in urine (data not shown). Five of six animals in both the pentamer and the pentamer + gB groups of animals also acquired virus infection, but with modestly reduced levels of viremia and urine virus shedding (data not shown).
Vaccination with pentamer alone led to some reduction of virus shedding in saliva, Fig 15 panel .. B. Importantly, inclusion of gB674 in combination with the pentamer resulted in significant delay and reduction in saliva virus shedding, relative to immunization with the pentamer alone, Fig 15 panel C. This result indicates that adding the gB674 to the pentamer reduced viral shedding in the urine. This likely reflects limitation of virus infection of the kidneys of the monkeys that received the combination of pentamer and gB antigens. This result demonstrates the utility of the rhCMV
gB674 antigen as a protective vaccine antigen, and by extension, of the analogous HCMV
gB705 antigen.

The following clauses describe additional embodiments of the invention:
Cl. A polypeptide comprising at least one mutation in the fusion loop 1 (FL1) region of an HCMV gB polypeptide.
02. A polypeptide comprising at least one mutation in the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
03. A polypeptide comprising at least one mutation in the fusion loop 1 (FL1) region and the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
04. A polypeptide comprising at least one mutation in the furin-like cleavage site of an HCMV gB polypeptide.
05. A polypeptide comprising at least two mutations in the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
06. A polypeptide comprising at least two mutations in the fusion loop 1 (FL1) region and the fusion loop 2 (FL2) region of an HCMV gB polypeptide.
07. A polypeptide comprising at least two mutations in the furin-like cleavage site of an HCMV gB polypeptide.
08. A polypeptide comprising a mutation at position Y155, as compared to SEQ
ID NO: 6.
09. A polypeptide comprising a mutation at positions Y155 and 1156, as compared to SEQ ID NO: 6.
010. A polypeptide comprising a mutation at positions Y155, 1156, and H157, as compared to SEQ ID NO: 6.
C11. A polypeptide comprising a mutation at positions 1156 and H157, as compared to SEQ ID NO: 6.
012. A polypeptide comprising a mutation at positions Y155, 1156, H157, and W240, as compared to SEQ ID NO: 6.

013. A polypeptide comprising a mutation at positions Y155 and W240, as compared to SEQ ID NO: 6.
014. A polypeptide comprising a mutation at positions Y155, H157, and W240, as compared to SEQ ID NO: 6.
015. A polypeptide comprising a mutation at positions Y155 and H157, as compared to SEQ ID NO: 6.
016. A polypeptide comprising the mutation Y155G, as compared to SEQ ID NO:
6.
017. A polypeptide comprising the mutations Y155G and I156H, as compared to SEQ ID NO: 6.
018. A polypeptide comprising the mutations Y155G, I156H, and H157R, as compared to SEQ ID NO: 6.
019. A polypeptide comprising the mutations I156H and H157R, as compared to SEQ ID NO: 6.
020. A polypeptide comprising the mutations Y155G, I156H, H157R, and W240A, as compared to SEQ ID NO: 6.
021. A polypeptide comprising the mutations Y155G and W240A, as compared to SEQ ID NO: 6.
022. A polypeptide comprising the mutations Y155G, H157R, and W240A, as compared to SEQ ID NO: 6.
023. A polypeptide comprising the mutations Y155G and H157R, as compared to SEQ ID NO: 6.
024. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 1.

025. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 2.
026. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 3.
.. 027. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 5.
028. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 7.
029. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 8.
030. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
1.
031. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
2.
032. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
3.
033. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
5.
034. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
7.
035. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
8.
036. The polypeptide according to any one of clauses 01-035, wherein the polypeptide does not comprise a mutation at any one of the following positions: (i) R236, (ii) G237, (iii) T158; (iv) Y242.
037. The polypeptide according to any one of clauses 01-035, wherein the polypeptide does not comprise any one of the following mutations: (i) R236N, (ii) G237N, (iii) T158N; (iv) Y242T; (v) Y2425; (vi) Y2420.
038. The polypeptide according to any one of clauses 01-037, wherein the amino acid sequence SEQ ID NO: 9 is a part of the polypeptide sequence.

039. The polypeptide according to any one of clauses 01-037, wherein the amino acid sequence SEQ ID NO: 10 is a part of the polypeptide sequence.
040. The polypeptide according to any one of clauses 01-037, wherein the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10 is a part of the polypeptide sequence.
041. The polypeptide according to any one of clauses 01-040, wherein the polypeptide does not comprise a protease cleavage site.
042. The polypeptide according to any one of clauses 01-040, wherein the polypeptide does not comprise a wild-type CMV protease cleavage site.
043. The polypeptide according to any one of clauses 01-040, wherein the polypeptide does not comprise a non-naturally occurring protease cleavage site that replaces the wild-type CMV protease cleavage site.
044. The polypeptide according to any one of clauses 01-040, wherein the polypeptide does not comprise an N- glycosylation site comprising N-X-S/T/C
motif, wherein X is any amino acid residue.
045. The polypeptide according to any one of clauses 01-040, wherein the polypeptide does not comprise a modified amino acid sequence that introduces an 0-linked glycosylation site.
046. The polypeptide according to any one of clauses 01-045, wherein the polypeptide does not include a deletion or substitution of any one of the amino acid residues selected from the group consisting of 154, 158, 159, 160, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 241, and 242, according to the numbering of SEQ ID NO: 6.
047. The polypeptide according to any one of clauses 01-046, wherein the polypeptide does not include a mutation of any one of the amino acid residues:

Y160, R236, S238, T239, and Y242, according to the numbering of SEQ ID NO:
6.
048. The polypeptide according to any one of clauses 01-047, wherein the polypeptide does not include the cytoplasmic tail of HCMV gB.
049. The polypeptide according to any one of clauses 01-048, wherein the polypeptide does not contain an insect cell pattern of glycosylation.
050. The polypeptide according to any one of clauses 01-049, wherein the polypeptide is contacted with ethylenediaminetetraacetic acid (EDTA).
051. The polypeptide according to any one of clauses 01-050, wherein the polypeptide undergoes a structural conformation change in response to a pH
change.
052. The polypeptide according to clause 051, wherein the polypeptide exhibits improved solubility or stability, as compared to a recombinant gB protein that does not include the following mutations Y155G, I156H, H157R, and W240A.
053. A composition comprising the polypeptide according to any one of clauses 01-051, and a diluent.
054. A composition comprising the polypeptide according to any one of clauses 01-051, and an adjuvant.
055. The composition according to any one of clauses 053-054, wherein the composition is immunogenic.
056. The composition of clause 055, for use in inducing an immune response against cytomegalovirus.
057. A recombinant nucleic acid molecule encoding the polypeptide according to any one of clauses 01-051, wherein the polypeptide undergoes a structural conformation change in response to a pH change.

058. The recombinant nucleic acid molecule according to clause 057, wherein said recombinant nucleic acid (a) is not a self-replicating RNA molecule; (b) is not an alphavirus replicon; (c) does not encode any alphavirus nonstructural proteins, such as NSP1, NSP2, NSP3 and NSP4; (d) does not contain: an Internal Ribosomal Entry Site (I RES), such as EMCV or EV71 ; and/or (e) does not contain a viral 2A site, such as FMDV.
059. A method for raising antibodies using the polypeptide according to any one of clauses 01-051.
060. The antibody according to clause 059, wherein said antibody is for use in a diagnostic assay.
061. The antibody according to clause 059, wherein said antibody is labelled directly or indirectly.
062. The antibody according to clause 059, wherein said antibody is for use in therapy.
063. A method of eliciting an immune response in a mammal, the method comprising administering to the mammal an effective amount of the polypeptide according to any one of clauses 01-051.
064. A method for reducing cytomegalovirus viral shedding in a mammal, the method comprising administering to the mammal an effective amount of the polypeptide according to any one of clauses 01-051.
065. The method according to clause 064, wherein the reduction in viral shedding is as compared to the viral shedding following an administration of a CMV
pentamer.
066. The method according to clause 064, wherein the challenge virus is homologous to the VR1814 CMV strain.
067. The method according to clause 064, wherein the challenge virus is homologous to the macacine herpesvirus 3 isolate 21252 CMV strain.

Claims (25)

1. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 1.
2. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 2.
3. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 7.
4. A polypeptide comprising an amino acid sequence that is at least about 60%
identical to SEQ ID NO: 8.
5. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 1.
6. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 2.
7. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 3.
8. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 5.
9. A polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 7.
10.A polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 8.
11.The polypeptide according to any one of claims 1-10, wherein the polypeptide does not comprise a mutation at any one of the following positions: (i) R236, (ii) G237, (iii) T158; (iv) Y242.
12. The polypeptide according to any one of claims 1-10, wherein the polypeptide does not comprise any one of the following mutations: (i) R236N, (ii) G237N, (iii) T158N; (iv) Y242T; (v) Y242S; (vi) Y242C.
13.The polypeptide according to any one of claims 1-12, wherein the amino acid sequence SEQ ID NO: 9 is a part of the polypeptide sequence.
14.The polypeptide according to any one of claims 1-12, wherein the amino acid sequence SEQ ID NO: 10 is a part of the polypeptide sequence.
15.The polypeptide according to any one of claims 1-12, wherein the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10 is a part of the polypeptide sequence.
16.The polypeptide according to any one of claims 1-15, wherein the polypeptide does not comprise a protease cleavage site.
17.The polypeptide according to any one of claims 1-15, wherein the polypeptide does not comprise an N- glycosylation site comprising N-X-S/T/C motif, wherein X is any amino acid residue.
18.The polypeptide according to any one of claims 1-15, wherein the polypeptide does not comprise a modified amino acid sequence that introduces an O-linked glycosylation site.
19.The polypeptide according to any one of claims 1-18, wherein the polypeptide does not include a deletion or substitution of any one of the amino acid residues selected from the group consisting of 154, 158, 159, 160, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 241, and 242, according to the numbering of SEQ
ID NO: 6.
20.The polypeptide according to any one of claims 1-19, wherein the polypeptide does not include a mutation of any one of the amino acid residues: Y160, R236, S238, T239, and Y242, according to the numbering of SEQ ID NO: 6.
21.The polypeptide according to any one of claims 1-20, wherein the polypeptide does not include the cytoplasmic tail of HCMV gB.
22.The polypeptide according to any one of claims 1-21, wherein the polypeptide does not contain an insect cell pattern of glycosylation.
23.The polypeptide according to any one of claims 1-22, wherein the polypeptide is contacted with ethylenediaminetetraacetic acid (EDTA).
24.The polypeptide according to any one of claims 1-23, wherein the polypeptide undergoes a structural conformation change in response to a pH change.
25.A composition comprising the polypeptide according to any one of claims 1-24, and a diluent.
CA3016867A 2016-03-11 2017-03-09 Human cytomegalovirus gb polypeptide Abandoned CA3016867A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662307423P 2016-03-11 2016-03-11
US62/307,423 2016-03-11
PCT/IB2017/051401 WO2017153954A1 (en) 2016-03-11 2017-03-09 Human cytomegalovirus gb polypeptide

Publications (1)

Publication Number Publication Date
CA3016867A1 true CA3016867A1 (en) 2017-09-14

Family

ID=58413151

Family Applications (1)

Application Number Title Priority Date Filing Date
CA3016867A Abandoned CA3016867A1 (en) 2016-03-11 2017-03-09 Human cytomegalovirus gb polypeptide

Country Status (3)

Country Link
EP (1) EP3426676A1 (en)
CA (1) CA3016867A1 (en)
WO (1) WO2017153954A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7179872B2 (en) * 2018-12-10 2022-11-29 Kmバイオロジクス株式会社 Vaccines for preventing or treating congenital infection with cytomegalovirus
KR20230022160A (en) 2020-06-09 2023-02-14 케이엠 바이올로직스 가부시키가이샤 A fusion protein of gB and pentamer of cytomegalovirus and a vaccine comprising the fusion protein

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0832638B2 (en) 1989-05-25 1996-03-29 カイロン コーポレイション Adjuvant formulation comprising submicron oil droplet emulsion
AU2009272284C1 (en) 2008-07-16 2014-05-15 Institute For Research In Biomedicine Human cytomegalovirus neutralizing antibodies and use thereof
AU2011315447A1 (en) * 2010-10-15 2013-05-09 Glaxosmithkline Biologicals S.A. Cytomegalovirus gB antigen
EP3031822A1 (en) * 2014-12-08 2016-06-15 Novartis AG Cytomegalovirus antigens

Also Published As

Publication number Publication date
WO2017153954A1 (en) 2017-09-14
EP3426676A1 (en) 2019-01-16

Similar Documents

Publication Publication Date Title
US20230109393A1 (en) Designer peptides and proteins for the detection, prevention and treatment of coronavirus disease, 2019 (covid-19)
US10364273B2 (en) Cytomegalovirus antigens
US20150086578A1 (en) Human cytomegalovirus vaccine
US11629172B2 (en) Human cytomegalovirus gB polypeptide
CN107531761A (en) Cytomegalovirus antigen and application thereof
TWI445547B (en) Dengue virus peptide vaccine and methods of preparing and using the same
Fowler et al. Marker vaccine potential of a foot-and-mouth disease virus with a partial VP1 GH loop deletion
KR100927221B1 (en) Polypeptide fragment of hepatitis E virus, vaccine composition and diagnostic kit using the same
US11857622B2 (en) Human cytomegalovirus GB polypeptide
KR20130041185A (en) Designer peptide-based pcv2 vaccine
US20180015159A1 (en) Cmv antigens and uses thereof
US20130216566A1 (en) Vectors expressing SARS immunogens, compositions containing such vectors or expression products thereof, methods and assays for making and using
KR20220009959A (en) CSFV subunit vaccine
US10611800B2 (en) Human cytomegalovirus gB polypeptide
CA3016867A1 (en) Human cytomegalovirus gb polypeptide
KR20190096965A (en) Isolation of a Novel Pastivirus That Causes Innate Progression A
KR101966841B1 (en) Recombinant antigen derived from zika virus e protein and use thereof
KR20230054719A (en) Vaccine against SARS-CoV-2 infection
Shigeki et al. Characterization of pseudorabies virus neutralization antigen glycoprotein gIII produced in insect cells by a baculovirus expression vector
Zhang et al. PF-D-Trimer, a broadly protective SARS-CoV-2 subunit vaccine: immunogenicity and application
WO1995004147A1 (en) Recombinant constructs using replacement sequences in hypervariable regions
CN117720628A (en) Preparation method and application of respiratory syncytial virus bivalent antigen
CN116472280A (en) Vaccine against SARS-CoV-2 infection

Legal Events

Date Code Title Description
FZDE Discontinued

Effective date: 20220325

FZDE Discontinued

Effective date: 20220325