CA2701727C - Cytomegalovirus vaccines and methods of production - Google Patents

Cytomegalovirus vaccines and methods of production Download PDF

Info

Publication number
CA2701727C
CA2701727C CA2701727A CA2701727A CA2701727C CA 2701727 C CA2701727 C CA 2701727C CA 2701727 A CA2701727 A CA 2701727A CA 2701727 A CA2701727 A CA 2701727A CA 2701727 C CA2701727 C CA 2701727C
Authority
CA
Canada
Prior art keywords
cmv
cells
cell
population
virus
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.)
Active
Application number
CA2701727A
Other languages
French (fr)
Other versions
CA2701727A1 (en
Inventor
Thomas Shenk
Dai Wang
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.)
Princeton University
Original Assignee
Princeton University
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 Princeton University filed Critical Princeton University
Publication of CA2701727A1 publication Critical patent/CA2701727A1/en
Application granted granted Critical
Publication of CA2701727C publication Critical patent/CA2701727C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • 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
    • C12N2710/16151Methods of production or purification of viral material
    • 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
    • C12N2710/16161Methods of inactivation or attenuation
    • C12N2710/16164Methods of inactivation or attenuation by serial passage

Abstract

Methods of increasing diversity in cytomegalovirus vaccines through the selection of cell type in which the virus is propagated, and the use of cytomegalovirus produced by those methods in the development of vaccine compositions, are disclosed. Vaccine compositions comprising CMV isolated from epithelial cells are also disclosed.

Description

CYTONIEGALOVIRUS VACCINES AND METHODS OF PRODUCTION
FIELD OF THE INVENTION
The invention relates generally to the field of vaccine development. More specifically, the invention relates to methods of increasing diversity in cytomegalovirus vaccines through the selection of cell type in which the virus is propagated.
and to the use of cytomegalovirus produced by those methods in the development of vaccine compositions.
BACKGROUND OF THE INVENTION
Various publications, including patents, published applications, technical articles and scholarly articles are cited throughout the specification. Each of these cited publications is incorporated by reference herein, in its entirety. Full citations for publications referenced by numbers in parentheses or otherwise not cited fully within the specification are set forth at the end of the specification.
Cytomegalovirus (CMV) is a herpes virus classified as being a member of the beta subfamily of herpesviridae. .According to the Centers for Disease Control and Prevention.
CMV infection is found fairly ubiquitously in the human population, with an estimated 40-80% of the United States adult population infected. The virus is spread primarily through bodily fluids, and is frequently passed from pregnant mothers to the fetus or newborn. In most individuals. CMV infection is latent. although virus activation can result in high fever, chills, fatigue, headaches. nausea. and splenomegaly.
Although most human CMV infections are asymptomatic. CMV infections in immunologically immature or immunocompromised individuals, such as newborns, HIV-positive patients. allogeneic transplant patients and cancer patients, can be particularly problematic. CMV infection in such individuals can cause severe morbidity.
including pneumonia, hepatitis. encephalitis. colitis, uveitis. retinitis, blindness, and neuropathy, among other deleterious conditions. In addition, CMV is a leading cause of birth defects. At present, there is no cure or preventive vaccine for CMV infection.
The entry of herpesviruses into cells is a complex process initiated by adsorption and receptor binding and followed by fusion of the virus envelope with a cell membrane. Fusion occurs at either the plasma membrane or an endosomal membrane. For instance, Epstein-Barr virus (EBV) enters primary B cells via receptor-mediated endocytosis (1,
2). yet it infects epithelial cells or transformed B cells by fusion of the virion envelope with the plasma membrane (1). Herpes simplex virus fuses with the plasma membrane of some cell types. but enters others by endocytosis (3-6). Human cytomegalovirus (HCMV) infects multiple cell types in vivo. including epithelial cells, endothelial cells and fibroblasts (7). It fuses with the plasma membranes of fibroblasts (8), but enters retinal pigmented epithelial cells and umbilical vein endothelial cells via endocytosis (9, 10).
The mechanism by which herpesviruses 'choose' their route of entry remains unclear.
It is generally assumed that entry pathways are mainly determined by the host cell, but there is precedent for tropic roles of virion glycoproteins (11). EBV virions contain two 01 complexes, gH/gL and gH/gL/gp42 (12, 13), which have mutually exclusive functions (11).
Fusion with the plasma membrane of B cells is mediated by gH/gL/gp42 (14-16).
but entry into epithelial cells is triggered by gH/gL (11, 12, 17). The cell type in which EBV is produced can alter its tropism. B-cell-derived EBV virions contain less gH-gL-gp42 than epithelial-cell-derived virions. As a result. B-cell-generated virus is more infectious for an epithelial cell and epithelial cell-derived virus is B cell tropic (18).
HCMV also encodes two gH/gL complexes: gEl/gL/g0 and gli/gL1pUL128/pUL130/
pUL131 (19, 20). The gO-containing complex is sufficient for fibroblast infection, whereas the pUL128/pUL130/pUL131-containing complex is required to infect endothelial and epithelial cells (19-21). The AD169 laboratory strain contains only the gli/gL/g0 complex in its virions (19). The absence of the second gH/gL complex is responsible for the loss of epithelial and endothelial cell tropism in HCMV laboratory strains (19-22).
There is a need for variety and diversity of CMV vaccines, and for effective means to control the spread and activation of the virus, particularly in immunocompromised individuals and pregnant women. The present invention addresses that need.
_ -SUM-MARY OF THE INVENTION
One aspect of the present invention features a method of making a cytomegalovirus (CMV) vaccine. The method comprises propagating strains or isolates of CMV in cultured cells of a selected cell type, thereby producing a cell type-conditioned CMV, and producing a CMV vaccine from the cell type-conditioned CMV. In certain embodiments, the CMV strain or isolate is a human CMV (HCMV) strain or isolate. A wide variety of cell types are suitable for the method, including but not limited to epithelial cells, endothelial cells.
fibroblasts, neuronal cells, smooth muscle cells, macrophages, dendritic cells and stromal cells. In a specific embodiment, the selected cell type is an epithelial cell.
The aforementioned method can further comprise producing the cell type-conditioned CMV in two or more different selected cell types and combining those CMV to produce the CMV vaccine. Alternatively or additionally, the method comprises providing two or more CMV strains or isolates. growing each of the strains or isolates in the cultured cells comprising the selected cell type or two or more different selected cell types, and combining all the CMV produced therefrom to make the CMV vaccine.
In certain embodiments, the method comprises producing a live attenuated CMV
vaccine. In other embodiments. it comprises producing an inactivated or killed CMV
vaccine. In still other embodiments, it comprises producing combination vaccines comprising one or more live attenuated viruses, inactivated viruses and other immunogenic components, e.g., immunogenic CMV proteins and peptides. and the like.
CMV vaccines produced by the aforementioned methods are also within the scope of the present invention.
Another aspect of the invention features kit for practicing the methods of the invention. Such kits typically include a package in which is contained one or CMV strains or clinical isolates, cultured cells of one or more selected cell types, and instructions for using the cultured cells and the CMV strains or isolates to produce cell type-conditioned CNIV for use in a CMV vaccine.
Another aspect of the invention features a vaccine composition comprising a cytomegalovirus (CMV) population or virion components thereof. admixed with a suitable pharmaceutical carrier or adjuvant, wherein the CMV population is isolated from an cultured cells of a selected cell type. In one embodiment, the selected cell type is an epithelial cell type. In one embodiment, the vaccine composition comprises HCMV.
- 3 -In various embodiments of the vaccine composition, the CMV population isolated from epithelial cell cultures is characterized by one or more features in subsequently infected host cells including but not limited to: (a) entry into the host cells by fusion with host cell plasma membranes; (b) greater virion-mediated cell-cell fusion of the host cells as compared with an equivalent CMV population isolated from cultured fibroblasts; (c) accelerated virus growth in the host cells as compared with an equivalent CMV population isolated from culture fibroblasts: (d) elicitation of a cellular response involving changes in expression greater than or equal to 2.5 fold of about two thirds fewer genes than a response elicited by an equivalent CMV population isolated from culture fibroblasts at 10 hours post-infection; or (e) elicitation of a cellular response involving a change in expression of one or more genes as shown in Table 2 and Table 4 herein, the latter being represented by GenBank Accession Nos: AK094860, NM 145023. NM 133492, NM_001039580, NM 001004301, NM 001034. A1369525, AK123066, NM 005345. NM 020731. BC071797. NM 003414, NM 000800. NM 138467, AK090803, AL133118. NM 001165. BG001037. NM 024861.
NM 001043, NM 016239. NM 001018084, NM 001037442, NM 017600, NM 022097, NM 175868, NM 032266. NM 003841. NM 005039. NM_14505 1 NM 004294.
AW856073. NM 024050, AF085968, NM 080927, NM 022115, AK056703. NM 000808, _ NM 012377 NM 006793 NM 031466, NM 005185, NM 139173, BX360933, NM 016125, NM 00104 NM 032188. NM 004185, NM 004843 or NM 173550.
_ In certain embodiments, the vaccine composition comprises a CMV population or virion components thereof isolated from a cell culture of two or more different selected cell types. For instance, the CMV population may be isolated from as an epithelial cells and cells of another cell type. such as a fibroblast cell type. In other embodiments, the CMV
population comprises two or more CMV strains or clinical isolates grown in the selected cell type. Certain embodiments can comprise a plurality of CMV strains or clinical isolates grown in cell cultures of a plurality of different cell types.
In one embodiment. the vaccine composition comprises a live attenuated CMV
vaccine. In another embodiment. it comprises an inactivated CMV vaccine. In still other embodiments, the vaccine composition can be a combination vaccine comprising one or more strains of live attenuated virus or components thereof, inactivated virus or components thereof, ancUor other immunogenic CMV peptides or proteins.
Another aspect of the invention features a method of immunizing an individual against CMV, comprising administering to the individual a CMV vaccine composition
- 4 -produced by the aforementioned methods and/or comprising the aforementioned features. In one embodiment, the individual to be immunized is a human.
Other features and advantages of the invention will be understood by reference to the drawings, detailed description and examples that follow.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Kinetics of HCMV 1E1 expression in ARPE-19 cells. (A) Infected cells (0.1 pfu/cell) were fixed at indicated times, and stained forIE1 (green in color photo. light gray in black and white photo), Sp100 (red in color photo, very dark gray in black and white photo) and DNA (blue in color photo, dark gray in black and white photo). (B) At various times after infection (0.1 pfu/cell), the percentage oflEl-expressing cells was quantified;
results are shown on the graph.
Figure 2. Electron microscopic analysis of HCMV entry into ARPE-19 cells.
ep1BADrUL131 orfibroBADrUL131 particles (50 pfu/cell) were bound to cells at 4 C and then allowed to internalize at 37 C for 15 min. Representative images are displayed.
Figure 3. Effects of inhibitors of endosome acidification and virion source on HCMV entry into ARPE-19 cells. Experiments were performed in triplicate, and the number of positive cells in drug-treated relative to untreated cultures is reported.
(A) Cells were pretreated with NH4C1 or BFA for 1 h, inoculated with epiBADrUL131 orfibroBADrULI31 (1 pfu/cell) and stained for 1E1 16 h later. (B) Cells were pretreated with 50 mM NH4C1 or 40 nivl BFA for 1 h. and then inoculated with BADrUL131 (0.1 pfu/cell) or FIX.wt (0.01 pfu/cell) produced in the indicated cell types and stained for 1E1 16 h later.
Figure 4. Fusion from without of ARPE-19 cells induced by epithelial cell-derived virus. (A) Cells were inoculated with epiBADrUL131 orfibroBADrUL131 (20 pfu/cell) and then maintained in medium containing 200 [ig/m1 of PFA. Phase contrast images were taken at 16 h post infection. (B) A mixture of reporter and effector cells were infected by epiBADrUL131 orfibroBADrUL131 (20 pfu/cell) for at 4 C for 1 h. The culture was then shifted to 37 C for 6 h. after which relative luciferase activity was measured.
Figure 5. Effect of pUL130-specific neutralizing antibody on 1-1CMV infection and entry. (A) Epithelial cell- or fibroblast-derived viruses were incubated with various concentrations of anti-pUL130. and residual infectivity was determined. (B) Epithelial cell-or fibroblast-derived virus particles were pretreated with anti-pUL130 at a final concentration of 20 vg/ml or with PBS, and then adsorbed to ARPE-I9 cells at 4 C for 1 h.
The cells were
- 5 -washed twice with cold PBS. and viral DNA associated with the cells was extracted to determine the relative numbers of particles attached to the cells.
Alternatively, the cells were shifted to 37 C for 2 h to allow the virus entry. Virions that did not penetrate the cells were removed by EDTA-trypsin treatment. Internalized viral DNA was subsequently quantified by real-time PCR.
Figure 6. Modulation of the ARPE-19 transcriptome by HCMV produced in epithelial cells versus fibroblasts. (A) Venn diagrams depict the distribution of differentially regulated genes at 6 h or 10 hpi with epiBADrUL131 orfibroBADrUL131(3 pfu/cell) relative to mock infection. (B) Changes in relative RNA levels assayed by real-time RT PCR.
The genes tested are hydroxymethylbilane synthase (HMBS. NNI_000190). GLI
pathogenesis-related 1 (glioma) (GliPR, NM_006851), pentraxin-related gene.
rapidly induced by IL-1 beta (PTX3, NM_002852). 2'-5'-oligoadenylate synthetase 3 (OAS3, NM_006187), interferon-induced protein 44 (IF144, NM 006417). v-rdl reticuloendotheliosis viral oncogene homolog B. nuclear factor of kappa light polypeptide gene enhancer in B-cells 3 (relB, NM 006509), and ATP-binding cassette, sub-family C (CFTR/MRP), member (MRP3, NM_003786).
DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
Various terms relating to the methods and other aspects of the present invention are used throughout the specification and claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although any methods and materials similar or equivalent to those described herein can be used in the practice for testing of the present invention, the preferred materials and methods are described herein.
In describing and claiming the present invention, the following terminology will be used. It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only. and is not intended to be limiting.
Definitions:
As used in this specification and the appended claims, the singular forms "a-, "an"
and "the" include plural referents unless the content clearly dictates otherwise. Thus, for example. reference to "a cell- includes a combination of two or more cells, and the like.
"About- as used herein when referring to a measurable value such as an amount.
a temporal duration, and the like, is meant to encompass variations of 20% or 10%, more
- 6 -preferably 5%, even more preferably 1%, and still more preferably 0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
The terms -amplifying,- "propagating," and "growing." or "amplification,"
-propagation.- and -growth" are used interchangeably herein to refer to the general process of introducing virus into cultured cells or infecting cells with virus under conditions permitting the virus to replicate and multiply within the cells, in accordance with methods well known to virologists and medicinal biologists. In particular, these terms are used herein to refer to the step of the inventive method in which the CMV is "conditioned"
by propagation on a selected cell type, as the step prior to using the conditioned CMV for the production of a vaccine.
"Biomolecules" include proteins, poplypepticles, nucleic acids, lipids.
polysaccharides, monosaccharides, and all fragments, analogs. homologs, conjugates, and derivatives thereof.
-Cell culture- refers generally to cells taken from a living organism and grown under controlled conditions (-in culture" or -cultured"). A -primary cell culture"
is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture. A "cell line- is a population of cells formed by one or more subcultivations of a primary cell culture.
A -coding region- of a gene consists of the nucleotide residues of the coding strand of the gene and the nucleotides of the non-coding strand of the gene which are homologous with or complementary to. respectively, the coding region of an mRNA molecule which is produced by transcription of the gene.
A "coding region- of an mRNA molecule also consists of the nucleotide residues of the mRNA molecule which are matched with an anti-codon region of a transfer RNA
molecule during translation of the mRNA molecule or which encode a stop codon.
The coding region may thus include nucleotide residues corresponding to amino acid residues which are not present in the mature protein encoded by the mRNA molecule (e.g., amino acid residues in a protein export signal sequence).
The terms -conditioned virus." "cell type-conditioned virus," -conditioned CMV-or -cell type-conditioned CMV" refer to CMV that has been propagated in a selected cell type prior to its use in vaccine production, in accordance with the methods described herein.
These terms are intended to be analogous to the term "conditioned medium.-which describes culture medium in which a particular cell type or cell line has been grown and then removed.
and which contains components or factors produced by the cells. thereby altering the
- 7 -
8 functionality of the medium. For purposes of the present application, the term -conditioned virus- similarly refers to virus that has been grown in a selected cell type and then removed from those cells, wherein the virus thereafter exhibits one or more altered functional features resulting from its growth in that cell type.
"Encoding- refers to the inherent property of specific sequences of nucleotides in a polynucleotide. such as a gene. a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e.. rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand.
used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA. Unless otherwise specified, a "nucleotide sequence encoding an amino acid sequence" includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns.
"Effective amount" or "therapeutically effective amount- are used interchangeably herein, and refer to an amount of a compound. formulation, material, or composition, as described herein effective to achieve a particular biological result. Such results may include.
but are not limited to. the inhibition of virus infection as determined by any means suitable in the art.
As used herein "endogenous" refers to any material from or produced inside an organism, cell, tissue or system. -Exogenous- refers to any material introduced from or produced outside an organism, cell, tissue or system.
The term "expression- as used herein is defined as the transcription and/or translation of a particular nucleotide sequence driven by its promoter.
As used herein, "immunization" or -vaccination" are use interchangeably herein and are intended for prophylactic or therapeutic immunization or vaccination. -Therapeutic vaccination- is meant for vaccination of a patient with CMV infection.
"Isolated" means altered or removed from the natural state. For example, a nucleic acid or a peptide naturally present in a living animal is not "isolated," but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is "isolated." An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell.
Unless it is particularly specified otherwise herein, the proteins. virion complexes.
antibodies and other biological molecules forming the subject matter of the present invention are isolated, or can be isolated.
The terms "patient.- "subject." -individual,- and the like are used interchangeably herein, and refer to any animal. or cells thereof whether in vitro or in situ, that can be infected with CMV. In certain non-limiting embodiments, the patient, subject or individual is a human.
"Parenteral- administration of an immunogenic or vaccine composition includes.
e.g..
subcutaneous (s.c.). intravenous (i.v.), intramuscular (i.m.), or intrasternal injection, or infusion techniques.
The term -polynucleotide- as used herein is defined as a chain of nucleotides.

Furthermore, nucleic acids are polymers of nucleotides. Thus, nucleic acids and polynucleotides as used herein are interchangeable. One skilled in the art has the general knowledge that nucleic acids are polynucleotides, which can be hydrolyzed into the monomeric "nucleotides." The monomeric nucleotides can be hydrolyzed into nucleosides.
As used herein polynucleotides include, but are not limited to, all nucleic acid sequences which are obtained by any means available in the art, including, without limitation.
recombinant means. i.e.. the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning and amplification technology, and the like, and by synthetic means.
As used herein, the terms -peptide," ''polypeptide.- and -protein" are used interchangeably, and refer to a compound comprised of amino acid residues covalently linked by peptide bonds. A protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein's or peptide's sequence. Polypeptides include any peptide or protein comprising two or more amino acids joined to each other by peptide bonds. As used herein, the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types. "Polypeptides- include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion
- 9 -proteins, among others. The polypeptides include natural peptides, recombinant peptides, synthetic peptides, or a combination thereof.
"Pharmaceutically acceptable- refers to those properties and/or substances which are acceptable to the patient from a pharmacological/toxicological point of view and to the manufacturing pharmaceutical chemist from a physical/chemical point of view regarding composition, formulation, stability, patient acceptance and bioavai lability.
"Pharmaceutically acceptable carrier" refers to a medium that does not interfere with the effectiveness of the biological activity of the active ingredient(s) and is not toxic to the host to which it is administered.
The term "single package- means that the components of a kit are physically associated in or with one or more containers and considered a unit for manufacture.
distribution, sale, or use. Containers include, but are not limited to, bags, boxes, bottles.
shrink wrap packages, stapled or otherwise affixed components. or combinations thereof. A
"single package" can also include virtual components. For instance, a kit may contain abbreviated physical instructions contained within the physical package. and instructions for accessing more detailed instructions from a virtual enviroment. such as a website for example.
The term "therapeutic- as used herein means treatment and/or prophylaxis. A
therapeutic effect is obtained by avoidance, delay, suppression, remission, or eradication of a disease state associated with CMV infection.
The term "treatment- as used within the context of the present invention is meant to include therapeutic treatment as well as prophylactic, or suppressive measures for the disease or disorder. Thus. for example. the term treatment includes the administration of an agent prior to or following the onset of a disease or disorder thereby preventing or removing all signs of the disease or disorder. As another example, administration of the agent after clinical manifestation of the disease to combat the symptoms of the disease comprises "treatment" of the disease. This includes for instance, prevention of CMV propagation to uninfected cells of an organism. The phrase "diminishing CMV infection- is sometimes used herein to refer to a treatment method that involves reducing the level of infection in a patient infected with CMV, as determined by means familiar to the clinician.
Description:
Cytomegalovirus (CMV) infects multiple cell types in vivo, including epithelial cells.
endothelial cells and fibroblasts. As summarized above in the background material, various studies have reported that the virus fuses with the plasma membranes of fibroblasts, but enters retinal pigmented epithelial cells and umbilical vein endothelial cells via endocytosis.
Due to the relative ease of propagating CMV in cultured fibroblasts as compared with epithelial or endothelial cell cultures, studies such as the above-summarized studies have been conducted using fibroblast-propagated CMV strains. Likewise, cultured fibroblasts are typically the cell type of choice in propagating CMV for clinical applications, such as the development of attenuated virus strains for vaccines.
It has now been demonstrated in accordance with the present invention that the cell type in which CMV particles are produced has a profound influence on their behavior in subsequent rounds of infection. Thus, for example, while it was heretofore reported that that CMV enters epithelial cells by endocytosis, the present inventors have demonstrated that this is the mode of entry for CMV propagated in fibroblasts. but not for CMV
propagated in cultured epithelial cells. Epithelial cell-propagated CMV enters epithelial cells predominantly via fusion with the plasma membrane. This different mode of entry has a variety of physiological consequences: it influences the kinetics with which the infection proceeds and it markedly influences the cellular response to infection. For instance virus grown in epithelial cells produces a dramatically muted cellular response as compared to cells infected with virus grown in fibroblasts. Many cellular anti-viral genes expressed after infection with fibroblast-grown virus are not expressed after infection with epithelial cell-grown virus. As a consequence, CMV grown in epithelial cells is predicted to perform differently than does a vaccine than CMV grown in fibroblasts. thus offering a new and unexpected source of diversity for the generation of CMV vaccines. Likewise.
propagation of CMV in other cell types. such as endothelial cells or specialized cell types that CMV is able to infect (e.g.. neurons, other cells of the central or peripheral nervous systems. smooth muscle cells, hepatocytes. stromal cells, macrophages or dendritic cells) should produce additional novel sources of diversity for the generation of CMV vaccines.
Thus. one aspect of the invention features methods of making CMV vaccines that exploit the variability associated with choosing a cell type in which to propagate the virus.
Another aspect features a kit for practicing the methods described above.
Another aspect of the invention features vaccine compositions for the prevention or treatment of CMV
infection, and methods of immunizing an individual using such compositions.
Various embodiments of these aspects of the invention are set forth below.

Methods of producing CMV vaccines:
The methods in accordance with an aspect of the invention comprise ( I ) providing a CMV strain or isolate; (2) propagating the strain or isolate in a cell culture of a selected cell type. and (3) harvesting CMV virions produced by growth in that cell type (referred to herein as -cell type-conditioned CMV") for use in producing a CMV vaccine.
The cell type selected for propagating the CMV prior to its use for vaccine development can be any cell line permissive for CMV infection that produces a yield of virus particles. The virus particles might be highly infectious in some assays or the particles might exhibit limited or no infectivity in many assays. Suitable cell types include, but are not limited to, (1) epithelial cell lines such as ARPE-19, which is exemplified herein and other retinal pigmented epithelial cell lines. e.g., epithelial cell line K-1034 (Ando, Y.. et al. 1997, Arch. Viro1.142(8): 1645-1658): HCMC. derived from normal human colonic mucosa (Smith. JD, 1986, J Virol. 60(2): 583-588): Caco-2 intestinal epithelial cells (Esclatine. A.. et al.. 2000, J. of Virol. 74(1): 513-51); SW480, HCT116, HeLa. H1299, and MCF-7 (regarding the latter five. see Wang. D. & T. Shenk, 2005. J. Virol. 79:
10330) (2) endothelial cell lines such as HMEC-1. a human microvascular endothelial line, immortalized with SV-40 virus large T antigen (Guetta. E.. et al., 2001, Cardiovascular Research 50: 538-546);
HUVEC and LMVEC (regarding the latter two, see Wang. D. & T. Shenk, 2005, J.
Virol. 79:
10330); (3) neuronal cells such as SK-N-SH, SK-N-AS and IMR-32 (see Wang, D. &
T.
Shenk, 2005. J. Virol. 79: 10330) as well as primary epithelial, endothelial.
smooth muscle.
macrophage and dendritic cells derived from a variety of tissue/organ sources.
Any CMV or combination of CMVs amenable to development as a vaccine is suitable for use as a source of the CMV for the method, as long as they can be grown in at least one selected cell type. In one embodiment. the CMV is human CMV (HCMV). either an isolate that has been previously isolated and characterized or a new isolate of HCMV
or an HCMV-like virus. In another embodiment, the CMV originates from another primate, including but not limited to chimpanzee (Davison, AJ et al. 2003, J. Gen. Virol. 84: 17-28) and rhesus monkey (Hansen. SG et al. 2003. J. Virol. 77:6620-36: Rivailler, P et al.
2006. J. Virol.
80:4179-82). The CMV can be an unmodified virus from a selected source. or it can be a chimeric virus produced by genetic modification or combination of elements from two or more different CMV strains or isolates.

Methods of making chimeric viruses are known in the art. To this end, at least six strains of human CMV have been cloned as infectious bacterial artificial chromosomes (BAC) and sequenced (Murphy. E et al. 2003. Proc. Natl. Acad. Sci. USA 100:
14976-14981.
The BAC sequences are available at GenBank Accession Nos. AC146999 (laboratory strain AD! 69, from which the BADrUL131 variant described herein was made): AC146851 (laboratory strain Towne): AC146904 (clinical isolate PH): AC146905 (clinical-like isolate Toledo): AC146906 (clinical isolate TR); and AC146907 (clinical isolate FIX).
At least two strains of human CMV have been sequenced without prior BAC cloning, and are available at GenBank Accession Nos. BK000394 (laboratory strain AD169) and AY446894 (clinical isolate Merlin). The entire genome of a chimpanzee CMV strain is available at GenBank Accession No. AF480884. The genome sequence of two rhesus CMV strains is also available (Accession Nos. AY I 86194 and DQ205516). Utilizing the teachings of the present application, the skilled artisan would be able to use any of the aforementioned sequences, or any other publicly available CMV sequence to prepare chimeric ClvIVs or to otherwise genetically modify a CMV.
It has been demonstrated in accordance with the present invention that laboratory strains of CMV that have been passaged repeatedly in fibroblasts can be successfully conditioned by propagation on the selected cell line. For instance, as described in the Example herein. BADrUL131. a BAC clone of the repeatedly passaged AD169 HCMV
strain in which the UL131 ORF has been repaired, was introduced by electroporation into cultured human foreskin fibroblasts. and the resulting virus preparation was amplified once in the epithelial cell line ARPE-19. Thus, various embodiments of the invention comprise the use of CMV (or the genomes of CMV) that has been passaged in a cell type that is different from the cell type selected for the conditioning step. For example. a CMV strain can be passaged multiple times in fibroblasts, then amplified in epithelial cells and thereafter used to produce a vaccine. It will be appreciated that the CMV can be amplified/propagated for one or more rounds in the selected cell type.
In preferred embodiments, the methods of the invention are used to produce live attenuated CMV for use as a vaccine. Methods to attenuate viruses are known in the art.
Preferably, attenuated CMV exhibit a diminished capacity for infectivity, and/or pathogenicity. including latency and activation, yet remain capable of inducing an immune response that treats or protects the host against CMV infection. Examples of attenuated CMV strains include, but are not limited to, laboratory strains, such as AD169 and Towne, which replicate almost exclusively in fibroblasts. Such attenuated strains, engineered if necessary to produce the requisite surface protein or protein complexes for appropriate tropism, can be grown epithelial cells or in fibroblasts and thereafter epithelial cells as discussed above, for use in the vaccine composition of the invention.
Serial passage in cultured cells, particularly fibroblasts, can be used to attenuate CMV. Repeated passaging of virally-infected host cells is carried out in vitro until sufficient attenuation of the virus is achieved. Passaging may be conducted under specific environmental conditions, such as modulated temperature. pH, humidity, in order to select for viruses with reduced infectivity or pathogenicity. If this method of attenuation is used, the serially passaged virus is then amplified in the selected cell type for one or more passages to produce the CMV to be used in the vaccine compositions of the invention.
Mutagenesis can also be employed to attenuate a virus. For example, CMV
virions can be exposed to ultraviolet or ionizing radiation or chemical mutagens.
according to techniques known in the art. In addition to their use to produce chimeric viruses.
recombinant techniques can also be used to produce attenuated CMV virions. For instance, site-directed mutagenesis, gene replacement, or gene knockout techniques can be used to derive virus strains with attenuated infectivity, pathogenicity or latency. An example of modifying a CMV by knockout mutagenesis is set forth in WO/2007/038316, which describes CMVs with genomes deleted in one or more latency-promoting genes, displaying an altered ability to enter or maintain a latent state.
In other embodiments. CMV isolated from the selected cell cultures are inactivated or killed and used in vaccine compositions. Methods of inactivating or killing viruses, e.g., with a chemical such as formalin, are well known in the art. It will be understood by the skilled artisan that the killed or inactivated CMV will comprise all or a substantial portion of the components of the viral particle, such that the diversity generated by the amplification in the selected cell type is maintained in the vaccine composition.
The methods of the invention can be used to create combinations of CMVs propagated in different selected cell types. thereby conferring an additional level of diversity to the vaccines that are produced. In one embodiment, a single CMV isolate or strain is used to infect two or more different cultured cell lines of different types. e.g., retinal epithelial cells and endothelial cells. The CMV produced by amplification in the respective cell types is then combined for use in a single vaccine. In another embodiment, two or more different clinical isolates or strains of CMV are used to infect a single selected cell line, and the multi-strain or multi-isolate CMV population produced by amplification in that cell type is used to produce a vaccine. In yet another embodiment. multiple isolates or strains are used to infect two or more different cultured cell lines of different types. and the CMV
populations produced by amplification in the respective cell types are combined for use in the vaccine.
Another aspect of the invention features kits for producing CMV vaccine materials in accordance with the methods described above. The kits comprise in separate containers in a single package or in separate containers in a virtual package. as appropriate for the use and kit component, aliquots of cell lines of one or more selected cell types, as well as one or more CMV isolates or strains, or vectors carrying the genomes of such CMV strains, to be introduced into and amplified in the selected cultured cell lines. Such kits also typically contain instructions, or links to instructions, for how to carry out the various steps of the method. Optionally, kits can also comprise culture medium and other reagents suitable for carrying out the cell culture and virus manipulations.
Vaccine compositions and methods of use:
Another aspect of the invention features an immunogenic composition (referred to interchangeably herein as a vaccine composition) comprising a cytomegalovirus (CMV) population or virion components thereof. admixed with a suitable pharmaceutical carrier or adjuvant, wherein the CMV is obtained via propagation in a selected cell type, for instance, an epithelial cell culture. As mentioned above, CMV vaccines have heretofore typically been prepared using CMV propagated in fibroblasts. However, it has been demonstrated in accordance with the present invention that propagation in epithelial cells yields virus that differs from fibroblast-propagated virus in many different ways. Virus produced in epithelial cells preferentially fuses with the plasma membrane, whereas fibroblast-derived virus mostly enters by receptor-mediated endocytosis. In addition, epithelial cell-generated virions had higher intrinsic -fusion from without- activity than fibroblast-generated particles, which influences the kinetics of infection. Furthermore, the two virus preparations trigger different cellular signaling responses, as evidenced by markedly different alterations in the transcriptional profile of infected epithelial cells.
In particular, CMV produced by propagation in epithelial cells have one or more of the following features, as compared with an equivalent strain or isolate of the virus produced by propagation in fibroblasts. First, as mentioned above, they can be distinguished by their entry into the host cells by fusion with host cell plasma membranes. CMV produced on epithelial cells also display greater virion-mediated cell-cell fusion of the host cells as compared with an equivalent CMV population isolated from cultured fibroblasts. as well as accelerated virus growth in the host cells as compared with an equivalent CMV population isolated from culture fibroblasts. In addition, they elicit a subdued cellular response as compared with equivalent CMV propagated in fibroblasts. At 10 hours post-infection about two-thirds fewer genes (-50 versus ¨150 genes) exhibit a 2.5 fold or more change in expression level. In addition.
epithelial-grown CMV can be characterized by the particular profile of host genes whose expression is changed (increased or decreased) following infection. These gene expression profiles are detailed in the Example. and can involve a change in expression of one or more genes represented by GenBank Accession Nos: AK094860. NM_145023, NM_133492.
NM 001039580. NM 001004301, NM 001034 A1369525, AK123066, NM 005345.
_ NN1_020731. BC071797. NM 003414, NM 000800. NM 138467. AK090803. AL133118.
NM 001165, BG001037. NM 024861. NM 001043, NM 016/39, NM 001018084.
NM 001037442, NM 017600. NM 022097, NM 175868, NM 032266, NM 003841, NM 005039. NN1_145051. NM 004294, AW856073, NM 024050, AF085968, NM 080927.
NM _022115. AK056703, NM 000808. NM 012377, NM _006793. NM 031466, NM 005185, NM 139173, BX360933, NM 016125, NM 002104, NM 032188, NM 004185, NM 004843 or NM 173550.
In this aspect of the invention, as in the foregoing aspects of the invention.
CMV or a combination of CMVs amenable to development as a vaccine is suitable for use as a source of the aforementioned CMV population. as long as they can be grown in at least one epithelial cell line or another selected cell type. In one embodiment, the CMV is HCMV or an HCMV-like virus. In another embodiment, the CMV originates from another primate, including but not limited to chimpanzee and rhesus monkey, as described above. The CMV can be an unmodified virus from a selected source, or it can be a chimeric virus produced by genetic modification or combination of elements from two or more different CMV strains or isolates.
as described above.
In preferred embodiments. the vaccine compositions comprise live attenuated CMV, which can be produced by the methods outlined above, all familiar to the skilled artisan. In other embodiments. CMV isolated from the selected cell cultures are inactivated or killed and used in vaccine compositions.
The vaccine compositions can comprise combinations of different strains or isolates of CMV, which can be propagated on a single epithelial cell cultures or on a number of different epithelial cell cultures, or on cells of another cell type, to generate additional diversity.

Furthermore, live attenuated CMV can be combined with killed or inactivated CMV. or with immunogenic components of CMV to produce a combination vaccine, e.g., live attenuated CMV combined with heat killed CMV, or combined with material for a subunit vaccine, or a combination of all three types of materials. Examples of immunogenic CMV
polypeptides and complexes suitable for subunit vaccines are described in WO 2007/146024 entitled "Cytomegalovirus Surface Protein Complex for Use in Vaccines and as a Drug Target."
The vaccine composition can further comprise one or more adjuvants. Adjuvants can be any substance that enhances the immune response to the antigens in the vaccine. Non-limiting examples of adjuvants suitable for use in the present invention include Freund's adjuvant, incomplete Freund's adjuvant, saponin, surfactants such as hexadecylamine, octadecylamine, lysolecithin, dernethyldioactadecyl ammonium bromide. N,N-dioctadecyl-N'-N-bis (2-hydroxyethylpropane diamine). methoxyhexa-decyl-glycerol. pluronic polyols.
polyanions such as pyran, diethylaminoethyl (DEAE) dextran, dextran sulfate, polybrene, poly IC, polyacrylic acid. carbopol, ethylene maleic acid, aluminum hydroxide, and aluminum phosphate peptides, oil or hydrocarbon emulsions, and the like.
Vaccines can be formulated in aqueous solutions such as water or alcohol, or in physiologically compatible buffers such as Hanks' solution. Ringer's solution.
or physiological saline buffer. including PBS. Vaccine formulations can also be prepared as solid form preparations which are intended to be converted, shortly before use. to liquid form preparations suitable for administration to a subject. for example. by constitution with a suitable vehicle, such as sterile water, saline solution, or alcohol. before use.
The vaccine compositions can also be formulated using sustained release vehicles or depot preparations. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
Thus. for example. the vaccines may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example. as a sparingly soluble salt. Liposomes and emulsions can be used as delivery vehicles suitable for use with hydrophobic formulations.
Sustained-release vehicles may. depending on their chemical nature, release the antigens over a range of several hours to several days to several weeks to several months.
The vaccine compositions may further include one or more antioxidants.
Exemplary reducing agents include mercaptopropionyl glycine, N-acetylcysteine, P-mercaptoethylamine, glutathione, ascorbic acid and its salts, sulfite, or sodium metabisulfite, or similar species. In addition, antioxidants can also include natural antioxidants such as vitamin E. C, leutein, xanthine. beta carotene and minerals such as zinc and selenium.
Vaccine compositions may further incorporate additional substances to function as stabilizing agents. preservatives, buffers, wetting agents, emulsifying agents. dispersing agents, and monosaccharides. polysaccharides, and salts for varying the osmotic balance.
The vaccines can further comprise immunostimulatory molecules to enhance vaccine efficacy. Such molecules can potentiate the immune response. can induce inflammation, and can be any lymphokine or cytokine. Nonlimiting examples of cytokines include interleukin (IL)-1, IL-2. IL-3, IL-4, IL-12, IL-13. granulocyte-macrophage colony stimulating factor (GNICSF). macrophage inflammatory factor, and the like.
Vaccines can be formulated for and administered by infusion or injection (intravenously, intraarterially. intramuscularly, intracutaneously, subcutaneously, intrathecally, intraduodenally. intraperitoneally. and the like). The vaccines can also be administered intranasally, vaginally, rectally, orally. topically. buccally, transmucosally, or transdermally.
An effective antigen dosage to treat against CMV infection can be determined empirically, by means that are well established in the art. The effective dose of the vaccine may depend on any number of variables, including without limitation, the size, height.
weight, age, sex, overall health of the subject, the type of formulation, the mode or manner or administration, whether the virus is active or latent. whether the patient is suffering from secondary infections, or other related conditions.
Vaccine regimens can also be based on the above-described factors. Vaccination can occur at any time during the lifetime of the subject, including development of the fetus through adulthood. Supplemental administrations, or boosters. may be required for full protection. To determine whether adequate immune protection has been achieved, seroconvcrsion and antibody titers can be monitored in the patient following vaccination.
The following example is provided to describe the invention in more detail. It is intended to illustrate. not to limit, the invention.
EXAMPLE
Human Cvtomegalovirus Uses Two Distinct Pathways To Enter Retinal Pigmented Epithelial Cells The experimental results described in this example demonstrate that HCMV
produced in two different cell types enters epithelial cells via different pathways.
Virions generated in epithelial cells preferentially enter via fusion at the plasma membrane.
whereas virions from fibroblasts enter by pH-dependent endocytosis. The two virus preparations induced markedly different cellular responses.
Materials and Methods Biological reagents. Human foreskin fibroblasts (HFFs) at passage 10 to 15 were maintained in medium with 10% newborn calf serum. Human MRC-5 embryonic lung fibroblasts and ARPE-19 retinal pigmented epithelial cells (American Type Culture Collection) at passage 24 to 34 were maintained in medium with 10% fetal bovine serum.
Human renal proximal tubular epithelial cells (hRPTECs) (Cambrex) were grown in medium with 10% fetal bovine serum and used at passage 4 to 5.
BADwt is derived from a BAC clone of the AD169 HCMV strain: BADrUL131 (19, 21) is a derivative of BADwt in which the UL131 ORF has been repaired; BFXwt is derived from a BAC clone of the VR1814 clinical FICIVIV isolate. Viruses were prepared by electroporation of BAC DNAs into HFFs, and the resulting virus preparation was amplified once in ARPE-19 cells or HFFs. unless otherwise specified. Cell-free virions were partially purified by centrifugation through a sorbitol cushion and resuspended in serum-free medium.
Virus titers were determined by plaque assay on MRC-5 cells. Neutralization of BADrUL131 was assayed by plaque reduction assay (19), by using purified anti-pUL130 monoclonal antibody (3E3) (19).
Anti-1E1 monoclonal antibody 1B12 was described previously (21). Rabbit anti-Sp100 polyclonal antibody (Chemicon) was used to visualize the NDlOs.
Electron microscropy. ARPE-19 cells were exposed to virus at 4 C for 1 h, unbound virus was removed by two washes with cold PBS, growth medium (37 C) was added for 15 min. cells were rinsed with phosphate-buffered saline (PBS). fixed and processed for electron microscopy, and examined with an FEI Tecnai-T12 microscope at 80 kv.
Assay for the dependence of infection on endosome acidification. ARPE-19 cells were pretreated with NH4C1 or Bafilomycin AI (BFA) (Sigma) for 1 h at 37 C, followed by infection in the continued presence of the inhibitor. 16 h later. cultures were fixed in 2%
paraformaldehyde and permeabilized with 0.10A Triton X-100. IE1 was identified by immunofluorescence using monoclonal antibody 1B12 (21)plus Alexa 546-conjugated secondary antibody and nuclei were stained with DAPI. Inhibition was calculated as the percentage of 1E1-expressing drug-treated relative to untreated cells.

Analysis of the fusion activity of virion proteins. To assay "fusion from without".
ARPE-19 cells were grown 10 90% confluence and infected. After 1 hat 37 C, the inoculum was removed and medium containing 200 .1g/m1 of phosphonoformic acid (PFA) was added to inhibit viral DNA synthesis. Fusion was monitored by visual inspection for syncytium formation.
A luciferase reporter assay was adapted to quantitatively analyze virion fusion activity. Reporter and effector ARPE-19 cells were prepared by electroporation (90-95%
efficiency) with a plasmid canying a luciferase gene under the control by a T7 promoter and a pcDNA3-T7 polymerase plasmid. respectively. At 24 h post transfection, the cells were mixed at a 1:1 ratio, and incubated at 37 C for an additional 16 h. The mixed populations were then exposed to HCMV virions at 4 C for 1 h, after which the monolayer was washed twice with cold PBS followed by addition of buffers (PBS with 10 mM 2-(N-morpholino)ethanesulfonic acid and 10 mM HEPES) with a final pH ranging of 4.5 to 8.
After 3 min at 37 C. the buffers were removed, and normal growth medium was added. At 6 hpi. the cells were lysed, and luciferase activity was assayed using a luciferase reporter assay system (Promega).
Assay of cellular transcriptional responses. Confluent ARPE-19 cells were serum starved for 24 h, followed by mock infection or infection. Total RNA was extracted at 6 or hpi by using Trizol (Invitrogen), and purified with an RNeasy column (Qiagen).
The RNA samples were amplified and labeled (cyanine-3) with the Agilent low RNA
input fluorescent linear amplification kit. To control for chip to chip variation, a reference RNA
(Clontech) was labeled (cyanine-5) and co-hybridized with the probes prepared from mock or HCNIV-infected cells. The hybridization was performed in duplicate with Aligent Human 44K oligonucleotide arrays. Arrays were scanned using an Agilent scanner at 5 micron resolution, and images were analyzed with Agilent Feature Extraction software to determine the intensities of fluorescent signals for hybridized spots and for background subtraction.
Agilent GeneSpring GX software was used for normalization and quantification of relative RNA changes.
Results Fibroblast-derived virions activate immediate-early gene expression in ARPE-19 cells with slower kinetics than epithelial cell-derived virions. The AD169 HCMV strain (BADivt) replicates poorly in ARPE-19 epithelial cells due to a mutation in its UL131 gene (10. 21). Repair of the mutation in AD169, producing BADrUL131, restores epithelial cell =

tropism (21) by allowing production of a gH/gL/pUL128/pUL130/pUL131 virion glycoprotein complex that is required for successful entry into these cells (19. 20).
BADrULI31 grown in ARPE-19 epithelial cells (epiBADrUL131) initiates its program of gene expression in epithelial cells more rapidly than BADrUL131 grown in HFF
fibroblasts (fibroBADrUL131) (Fig. IA). When ARPE-19 cells were infected with epiBADrUL131. ¨17% of the cells expressed detectable 1E1 protein at 6 h post infection (hpi). 1E1 expression was accompanied by disruption of NDlOs in the nucleus.
In contrast, infection withfibroBADrUL131 led to 1E1 expression in only 2.8% of ARPE-19 cells at 6 hpi. The number of 1E1-expressing cells, however, increased with time. There was no significant difference in the percentage of 1E1-expressing ARPE-19 cells at 24 hpi with virus produced in the two cell types (Fig. 1B).
Virions produced in HFFs versus ARPE-19 cells enter ARPE-19 cells via distinct pathways. An electron microscopic examination of virus entry was performed to determine if the different kinetics of 1E1 accumulation for ARPE-19 cell-derived virus versus HF17-derived virus resulted from an event prior to the onset of viral gene expression. ARPE-19 cells incubated with epiBADrUL131 orfibroBADrULI31 were permitted to attach at the cell surface at 4 C. and cultures were shifted to 37 C for 15 min to allow internalization before processing for microscopy. For each sample, 40-50 cells were examined, with at least 90%
of the cells showing either intact virions or capsids. The number of virus particles in each cell varied from 2-8. with most cells showing 2- 3 particles.
In epiBADrUL131-infected ARPE-19 cells, virions were found almost exclusively at the cell surface. with about 97% of the virions at the apical membrane. Some particles were close to the cells but the section did not reveal evidence of contact (Fig.
2A, panel a). and others were captured in the process of fusion at the plasma membrane (Fig. 2A.
panels b and c). Capsids beneath the inner surface of the membrane were observed rarely: in fact, only two examples were identified (Fig. 2A, panels d and e). No enveloped virions were found inside the cells. This result indicates that ep1BADrUL131 enters the ARPE-19 cells by fusion with the plasma membrane. In contrast, fibroBADrUL131-infected cells contained virions at the cell membrane (-65% of total) and inside the cell within vesicles (-35% of total) (Fig.
2B). The particles within vesicles were enveloped. indicating they entered by endocytosis.
Entry of the BFXwt clinical isolate propagated in fibroblasts was also examined. This clinical isolate accumulated in vesicles within ARPE-19 cells (Fig. 2C), supporting the validity of BADrUL131 as a model for cell entry by a clinical isolate of HCIvIV.

Infection of ARPE-19 cells by fibroblast- but not epithelial cell-derived virus is p1-1 dependent. Many viruses that enter cells by endocytosis (1.4, 10) require acidification of endosomes for the virion envelope to fuse with the endosomal membrane and release the capsid into the cytoplasm. NH4C1. which buffers endosomal pH. and bafilomycin Al (BFA).
which blocks the endosomal ATPase proton pump, were tested for their effect on infection of ARPE-19 cells. After pretreatment with either agent. cells were infected and cultured in drug-containing medium for a further 1611. Successful infections were scored by assaying for IEl-positive cells. Consistent with the ultrastructural analysis described above, pretreatment with either agent had only a modest effect on ep1BADrUL131 infection (Fig.
3A). In contrast, both agents inhibited 1E1 expression afterfibroBADrUL131 infection in a dose dependent manner. indicating that the entry of fibroblast-generated virus was dependent on endosomal acidification. The fact that the agents had little effect on entry by epiBADrUL131 shows that the inhibition offibroBADrUL131 did not result from toxicity.
It was next determined whether virus grown in other types of epithelial cells and fibroblasts display the same properties as ARPE19- and HFF-derived virions.
Virus stocks from laPTEC epithelial cells and MRC-5 fibroblasts were used to infect ARPE-19 cells after treatment with NH4C1 or BFA. and they responded to the inhibitors exactly as did virus grown in ARPE-19 cells or HFFs (Fig. 3B, left panel). Thus. BADrUL131 produced in two different fibroblasts was substantially more sensitive to the inhibitors than virus produced in two different epithelial cell lines.
The effect of endosomal pH on entry of the BFXwt clinical isolate into ARPE-19 cells was also assayed (Fig. 3B. right panel). NI-14C1 or BFA significantly reduced the number of lEl-positive ARPE-19 cells produced by infection with fibroblast-generated BFX114, but only a slight inhibition was observed after infection with epithelial cell-derived BFXwt.
Virions produced in epithelial cells have higher intrinsic fusion activity than virions from fibroblasts. As is the case for other herpes viruses. HCMV
clinical isolates promote cell-cell fusion that can be detected as early as 3-5 hpi. The rapid production of syncytia without de novo synthesis of virus envelope proteins indicates that it is promoted by -fusion from without", a process by which enveloped virions directly fuse target cells. Since BADrUL131 produced in epithelial cells versus fibroblasts enters epithelial cells differently, the possibility that they would exhibit different -fusion from without"
activities was tested.
Mock-infected ARPE-19 cells exhibited no syncytia (Fig. 4A). and syncytia were rarely found after infection withfibroBADrUL131 (Fig. 4B). In contrast, after exposure to epiBADrUL131, cell-cell fusion was detected as early as 6 hpi, and 20-30% of the nuclei were aggregated in syncytia by 24 hpi (Fig. 4C). Cells were treated with PFA, which blocks progression to the late phase of infection, so the fusion must have been induced by epiBADrUL131 particles and not by newly expressed virion proteins.
A luciferase reporter assay was used to quantify the fusion activity of viral particles as well as the effects of pH on fusion from without. Reporter and effector cells received a plasmid containing a luciferase gene driven by a T7 promoter or a T7 RNA
polymerase expression plasmid. respectively. The two ARPE-I9 derivatives were mixed, and infection-dependent fusion was quantified by assaying luciferase expression. epiBADUL131 consistently induced higher fusion activity thanfibroBADrUL131 (Fig. 4D). At pH 7-8, the activity offibroBADrUL131 was ¨3-fold lower than that of epiBADrUL131. When the cells were treated with low pH buffers after virus adsorption, both virus preparations mediated modestly enhanced fusion. BADwt did not induce fusion in this assay.
The mode of entry does not alter LICIVIV cell tropism. As discussed above, there is precedent for a herpesvirus to favor entering a specific cell type depending on the cell in which the infecting virus was produced. This phenomenon is different than the one that was observed as described above, i.e.. HCMV preparations from different cell types enter epithelial cells by different mechanisms. Nevertheless, it remained possible that the different entry mechanisms would impact on the efficiency of replication and yield, resulting in a tropic effect. Therefore, experiments were conducted to determine whether the mode of entry influenced HCMV plaque production on epithelial cells as compared to fibroblasts (Table 1).
Stocks of BADrUL131 were produced in ARPE-19. liRPTEC. HFF or MRC-5 cells and assayed for plaque formation on ARPE-19 or MRC-5 cells (Table 1). Although slightly more plaques were produced on ARPE-19 than MRC-5 cells, neither epithelial cell-nor fibroblast-derived virus preferentially generated plaques on one cell type compared to the other.
TABLE 1. Titration of epithelial cell derived or fibroblast derived BADrUL131 in ARPE19 and MRCS cells (x105) Source of Target cells Ratiob replication" ARPE-19 MRCS
ARPE-19 8.8 3.4 2.6 hRPTEC /.9 1.9 1.5 NIRC5 4.3 2.7 1.6 HFF 6.8 2.7 2.5 "2x105 ...
pi-u of BADrUL131 originally titrated in IIFFs were used to infect ARPE-19 or MRCS cells.
b .
Ratio of ARPE-19 titer in relation to MRCS titer.
pUL130-specific antibody blocks ARPE-19 infection by both epithelial- and fibroblast-derived virus. A pULI30-specific antibody, which neutralizes HCMV
infection of epithelial cells (19). was able to block ARPE-19 infection by either mode of entry (Fig.
5A). It inhibited infection by both viruses in a dose dependent manner, although epiBADrUL 131 was somewhat more sensitive to neutralization thanfibroBADrUL131. The ability of the antibody to inhibit both modes of entry reinforces the conclusion that the pUL130-containing complex functions whether fusion occurs at the plasma membrane or the endosomal membrane.
It has been reported previously that the gH/gL/pUL128/pUL130/pUL131 complex is dispensable for HCMV to be internalized by endothelial or epithelial cells, because laboratory strains lacking this complex are efficiently endocytosed (10).
However.
subsequent fusion with endosomal membrane and escape into the cytoplasm requires the complex. Consistent with these earlier results, the antibody to pUL130 did not block binding or internalization of epiBADrUL131,fibroBADrULI31 or BADivt when assayed on ARPE-19 cells (Fig 5B). However, the total amount of internalized fibroblast-derived virus was lower than that of the epithelial cell-derived virus. This might reflect a reduced rate of internalization, which would be consistent with the delay in onset of tEl expression by the fibroblast-derived virus (Fig. 1).
ep1BADrUL131 andfibroBADrUL131 induce different transcriptional responses in ARPE-19 cells. Like many other viruses, HCMV modulates cellular signaling pathways during entry. One consequence of the altered intracellular signaling is a dramatic change in the cellular transcriptome, which results substantially from contact of virion glycoproteins with the host cell.
Accordingly. the impact of the two entry pathways on the transcriptional response of ARPE- 1 9 cells was investigated. Cells were mock infected or infected with epiBADrUL131 orfibroBADrUL131, and total RNA was purified 6 or 10 h later. Relative RNA
levels were analyzed by using microarrays, and infected-cell RNAs whose levels changed by a factor of >2.5 relative to mock-infected controls were identified (Tables 2-5). The distributions of RNAs with increased or decreased expression are depicted by Venn diagrams in Fig. 6A.

1 Table 2. Differentially transcribed genes from epiBADrUL131-infected ARP19 cells at 6 h after infection _ Genbank Fold Change Gene Name NM_020904 7.218 PEPP1 .kK.124132 5.97 L0C340286 AK074031 4.89 SLIM; FLJ34715 NM_058188 4.658 PRED54; MGC149386; MGC149387 NN1_022047 4.578 1BP
NM 020436 3.172 DRRS; HSAL4; ZNI7797; MGC133050; WI 112F19.1 NM_00 I 165 3.049 Alp!; API2; MIIIC; CIAP2: HAIP1; HIAP I; MALT2;

NM 001039580 3.011 ASAP; FLJ2I159 NN1_000364 2.91 CMH2; TnTC; cTnT; CMPD2; MGC3889 NM 005031 2.866 PLM; MGC44983 L08436 2.875 CLP; FLJ43657; MGC19733 NM_145867 2.768 MGC33147 AL133118 2.731 AL133118 NM 001034 2.706 R2; RR2m NM 020943 2.674 KIAAI 604 BC039151 2.67 PABPC1L; FLJ42053; LIJ1069P2.3 NM 031217 2.659 DKFZP434G2226 NN4_003425 2.61 KOX5; ZNF13 NM 000499 2.58 A1111; AHRR; CP11; CYP1; P1-450; P450-C; P450DX
NN1_182751 2.578 CNA43; PR02249; MGC126776 NNI_1 44620 7.577 MGC14816; DKFZp31301122 NM_020359 0.4 PLSCR2 AF085968 0.396 AF085968 NM_053064 0.388 GNG2 NM 005039 0.38 PM; PMF; PMS; Ps 1; Ps 2; PRB I L; PRB I Ivl NM 152525 0.373 FLJ25351; FLJ40332 AK125975 0.365 FLJ43987 NM 175868 0.365 MAGE6; MAGE3B; MAGE-3b; MGC52297 NNI_O 17600 0.358 DKFZp434M0331 NM 006650 0.355 CPX2; 921-L; CPX-2; MGC138492 NM 004294 0.343 RF1; NITTRFI: MGC4772I
NN1_006434 0.343 CAP; FLAF2; R85FL; SH3D5: SORB I
NM_031466 0.339 NIBP: Ti; IBP; MGC4737; MGC4769; KIAA1882 NM_000808 0.324 MGC33793 NN1_012377 0.324 0R7C3: 0R19-18; CIT-HSP-87M17 NN1_001018084 0.31 NN1_001018084 NM 024050 0.304 DDAI: PCIA I; MGC2594 NN1_005185 0.299 CLP
NM_022115 0.272 PFM15; ZNF298; C2lor183 NM 016125 0.259 L0051136; MGC111090 NN1_004843 0.256 CRL I; TCCR; WSX1; 1L27R; zcytorl NM 004334 0.242 CD157 NM_004185 0.233 WNT13; XWNT2 BX360933 0.229 SLC25A5 NM_032188 0.777 NIOF; hM0F; FLJ14040 NM_1 73550 0.271 FLJ39267; FLI46740; MGC50805 NM_002104 0.162 TRYP2 Microarray targets that hybridized with labeled RNA from epiBADrUL131-infected ARPE-19 cells were compared to mock-infected cells, and probe sets whose levels varied by 22.5 fold are listed. The Genebank desienation, fold change and gene name are listed.
Table 3. Differentially transcribed genes from fibroBADrUL131-infected A R1319 cells at 6 h after infection Genbank Fold Change Gene name NN1_183040 15.03 SDY; DBND; HPS7; My031; F1130031; NIGC20210;

NM_001165 12.48 AIP I; API2; MIHC; CIAP2: HAIP1; HIAPI; NIALT2;

NM 002852 11.21 TSG-14; TNFAIP5 NM 006509 7.008 I-REL
NM 139314 6.679 NL2; ARP4; HAF; PGAR; HFARP; pp1158; ANGPTL2 NM 002982 5.977 HC11; NICAF; MCP1; MCP-1; SCYA2; GDCF-2 NN1_025169 5.938 ZFP: ZNF64: ZKSCAN7; FLJ12738 NM 033066 5.144 DLG6; ALS2CR5 NM 001946 4.971 NIKP3; PYST1 NN1_000212 4.92 CD6I; GP3A; GPIlla NM 001673 4.214 TS1 I
NM_004464 4.183 HBGF-5; Smag-82 NN1_021101 4.072 CLD1: SEMPl; ILVASC
NM_006851 4.07 GLIPR; RTVP I; CRISP7 AK094860 3.913 AK094860 NM_052875 3.667 Pep8b; MGC10485 NNI_005347 3.648 BIP; MIF2; GRP78; FLJ26106 NM 022842 1592 CD318; TRASK; SIMA135 U16307 3.36 GLIPR; RTVPI: CRISP7 NM 000800 3.335 AFGF; ECGF; FGFA; ECGFA; ECGFB
NNI_000800 3.306 HBGF1; GLI0703; ECGF-beta; FGF-alpha NM 198833 3.257 P18; CAP2 NN1_002053 3.21 GBP1 NM 058179 3.161 PSA; EPIP; PSAT; MGC1460 NM 001004301 3.131 FLJ16542; FLJ34141 NM_180989 3.117 ITR
NN1_000640 3.116 IL-13R; IL I3BP: CD213A2 NN1_002658 3.09 ATF; UPA: URK: u-PA
NNI_018284 3.076 FL110961; DKFZp686E0974; DKFZp686L15228 NN1_000201 3.02/ BB2; CD54; P3.58 NM 005923 3.007 ASK1: MEKK5; MAPKKK5 NNI_018836 3.001 NIOT8; SHREW1; SI IREW-1; RP3-426F10.1 NN1_004556 2.971 IKBE
NM 022044 2.955 SDF2L1 NM_006611 2.954 Ly49; KLRAti; LY49L; Ly-49L; NIGC126520;

NNI_014314 2.935 RIG-I; F1.113599; DKFZp434J1111; DKFZp686N19181 NM 003897 2.906 DIF2; IEXI; PRGI; DIF-2; GLY96; IEX-1; IEX-1L
NM_006417 2.899 p44; MTAP44 NM_006187 2.877 p100; MGC133260 NR_002186 2.876 DKFZp58611420 NN1_033036 2.872 GAL3ST2: GAL3ST-3: MGC142112; MGCI42114 NM 014331 2.86 xCT: CCBR I
NM 003786 2.831 MLP2; NIRP3; ABC31: MOAT-D; cMOAT2; EST90757 NM_001511 2.829 GRO1; GROa; NIGSA; NAP-3; SCYBI; MGSA-a; MGSA alpha NM_000189 /.8/7 1-1K11; HXK2; DKFZp686M1669 NM 00190! /.8/1 CCN2; NOV2; HCS24; IGFBP8; NIGC102839 NM 031217 2.811 DKFZP434G2226 NM 002849 2.766 PTPRQ; EC-PTP; PCPTP1; PTP-SL; PTPBR7 NM_019891 2.764 ERO I LB
NM 002234 2.745 141(2; HCK1; PCN1: HPCN1; KV1.5; NIGC I 17058; MGC I

NNI_198569 2.739 DREG; VIGR: PS ITP2 NNI_020799 2.726 AMSH-FP; AMSH-LP; ALMalpha; FLJ3 1524; KIAA1373; etc NM_014632 /.7/6 KIAA0750; MICAL2PV I; MICAL2PV2 NM 182920 2.721 FLJ42955; KIAA1312 NM_003483 2.715 BABL; LIPO: HMGIC: HMGI-C
NMI 33492 2.706 ACERI; MGC138327; MGC138329 CR598364 2.633 ENST00000370238 NM 000970 /.62 TXREB I ; SHUJ1JN-2; TAXREB107 NM 005444 2.617 RCDI; CNOT9; RCD I +
NNI_194303 2.614 NM_194303 NNI_015359 2.612 ZIP14; cig 1 9: LZT-Hs4; KIAA0062 NNI_016354 2.608 POAT: OATPI: OATP-E; OATP4A I ; OATPRP I ; SLC21Al2 NN1_015009 2.607 LNX3; SEN1ACAP3 AK 124941 /.60/ AK124941 NM 001548 /.602 G I OP 1 ; IF156; ISG56; IF1-56: IFNAI I ; RNM561;

NNI_145023 2.597 FL132762; DKFZp686N0559: RP11-479G22.1 NNI_023070 2.592 FLJ34293: RP 1 1-656D10.1 NM_001902 2.584 MGC947 1 NM 004233 2.563 BL1 I; HB15 NM 020683 /.562 A3AR; AD026; bA552N111.5; RP I 1-552M11.7 NM 031938 2.56 FL.134464; B-DIOX-II
NM_152649 2.55 FLJ34389 BC048263 2.543 L0C146909 XN1_210365 2.527 L0C284288 NNI_007107 2.515 TRAPG; SSR gamma NM 002837 2.513 PTPB; HPTPB: FLJ44133; MGC59935; FIPTP-BETA;
NM_172345 2.505 NM 172345 NM 002609 0.4 JTK I 2; PDGFR; CD140B; PDGFR I ; PDGF-R-beta NM_198353 0.4 KCTD8 NM 003558 0.394 NISS4; STNI7 NM_001010911 0.392 bA418C1.3 NNI_017644 0.391 DREI: F1125796 NM 052892 0.388 FLJ45333; DKFZp686J19100 NM_I 75868 0.387 MAGE6; MAGE3B; MAGE-3b; MGC52297 NM 007282 0.38 RZF; MGCI3689 NNI_005185 0.38 CLP
NM_021990 0.378 GABRE
AK055156 0.375 FLJ30594; MGCI20893: DKFZp761K2322 AF085968 0.375 AF085968 NNI_019555 0.371 GEF3; STA3: XPLN; MGC118905; DKFZP434F2429 NM 004294 0.368 RF1; MTTRF1; MGC47721 NNI_173039 0.365 AQPX1 BU943730 0.364 BU943730 NM_017600 0.364 DKFZp434N10331 NM 007282 0.36 RZF; N1GC13689 AL713743 0.357 FLJ42875; MGC35434; DKFZp761G0122 NM_007314 0.347 ARG; ABLL
AK056190 0.345 WHRN; CIP98; USH2D; KIAA1526; RP I 1-9M16.1;

NM_000372 0.345 OCA1A; OCAIA
BC015929 0.338 RVR; 13D73: FIZF2: EAR-1r; Hs.37288 NM 012377 0.328 0R7C3; 0R19-18: CIT-EISP-87M17 NM_1 38440 0.324 SLITL2 NM 001018084 0.317 NN1_001018084 NNI_000808 0.311 MGC33793 NM 033260 0.31 FIFH1 NM 022160 0.309 DNI0; MGC163307; MGC163309 BC018597 0.308 BC018597 NM_ I 98404 0.305 bA32 IC24.3 NM 024050 0.303 DDA1; PCIAI: N1GC2594 NN1_031466 0.299 NIBP; T1; IBP; MGC4737; NIGC4769; KIAA1882 NM_016831 0.287 GIGI3 NM_022115 0.251 PFM15; ZNI:298; C2lor183 NM 016125 0.249 L0051136; MGC111090 NN1_002104 0.141 TRYP2 NM_013261 0.236 LENI6; PGC1; PGC1A; PGC-1 ; PPARGCI; PGC-1(alpha) NM_002167 0.211 HEIR-1 NM 032188 0.204 MOF;11M0F; FLJI4040 BX360933 0.197 SLC25A5 NM 003862 0.194 ZFGF5; FGF-18 NM 173550 0.148 FLJ39267; FLJ46740; N1GC50805 NN1_004185 0.135 WNT13; XWNT2 I Microarray targets that hybridized with labeled RNA from tibroBADrUL131-infected ARPE-I9 cells were compared to mock-infected cells, and probe sets whose levels varied by >2.5 fold are listed. The Genebank I designation, fold change, and cene name are listed.
Table 4. Differentially transcribed genes from ep1B.kDrULI31-infected ARP19 cells at 10 Ii after infection Genbank Fold Change Gene Name AK094860 5.688 AK094860 NNI_145023 4.19 FLJ32762; DKFZp686N0559; RP11-479G22.1 NNI_133492 3.456 ACER1; N1GC128327; MGC138329 NN1_001039580 3.352 ASAP; FLJ21159 NM 001004301 2.982 FLJ 16542; FLJ34141 NM _001034 2.911 Ft2; RR2M
M369525 2.764 A1369525 AK123066 2.753 AK123066 NM 005345 2.729 HSP72; HSPA1B; HSP70-1 NM 020731 1.711 AHH; A1111R; KIAA1234 BC071797 2.631 BC071797 NM 003414 2.609 HZF2 NM 000800 2.576 AFGF; ECGF: FGFA; ECGFA; ECGFB; HBGF I ; GLI0703;
etc NM 138467 2.571 Clorfl 71; FLJ40918 AK090803 2.557 SRrp35; FLJ14459; FLJ33484; FLJ41221; RP11-63L7.3 AL133118 /.5/9 AL133118 NM_001165 2.508 AIP1; API2; MINC; CIAP2; HAIP1; HIAP I ; MALT2;

BG001037 0.392 TXNRD I
NM 024861 0.388 FLJ22671; MGC150431: MGC150432 NM 001043 0.385 NET; NAT 1; NET!: SLC6A5 NN1_016239 0.384 DFNB3; NlY015; DKFZp686N18198 NM 001018084 0.383 NM 001018084 NM 001037442 0.381 RIPX; KIAA0871 NM_017600 0.377 DKFZp4341V10331 NM 022097 0.369 L0063928 NM 175868 0.356 MAGE6; MAGE3B; MAGE-3b; NIGC52297 NN1_032266 0.342 DKFZp434G118; DKFZp781D2023 M1_003841 0.342 LIT; DCR I; TRID: CD263; TRAILR3; MGC149501;

NM 005039 0.339 PM; PMF; MIS; Ps 1; Ps 2; PRB I L; PRBINI
NNI_145051 0.339 MGC4734; FLJ31197 NN1_004294 0.336 RF I ; MTTRFI: MGC47721 AW856073 0.335 AW856073 NM 024050 0.327 DDA I ; PCIA1; MGC2594 AF085968 0.327 AF085968 NM 080927 0.318 ESDN; CLCP1 NM 022115 0.317 PFN115; ZNF298; C2lorf83 AK056703 0.309 L0C219731 NNI_000808 0.301 MGC33793 NM 012377 0.299 0R7C3; 0R19-18: C1T-HSP-87M17 AOP1; NIERS; AOP-1: SP-22; PR01748; MGC24293;
NNI_006793 0.298 MGC104387 NM 031466 0.289 NIBP; T1; 113P; MGC4737; MGC4769; KIAA1882 NN1_005185 0.286 CLP
NN1_139173 0.286 N1GC131641 BX360933 0.28 SLC25A5 NN1_016125 0.269 L0051136; MGC111090 NNI_002104 0.251 TRYP2 NM 032188 0.248 NIOF; hM0F; FLJ14040 NM_004185 0.245 WNT13; XWNT2 NM_004843 0.237 CRL1; TCCR: WSX I; 1L27R; zcytorl NNI_173550 0.208 FLJ39267; FLJ46740; MGC50805 Microarray targets that hybridized with labeled RNA from epiBADrUL131-infected ARPE-19 cells were compared to mock-infected cells, and probe sets whose levels varied by _?-'2.5 fold are listed. The Genebank designation, fold change and gene name are listed.
Table 5. Differentially transcribed genes fromfibroBADrUL131-infected ARPI9 cells at 10 h after infection Genbank Fold Change Gene Name AK094860 11.26 AK094860 NN1_033066 8.751 DLG6; ALS2CR5 NM 145023 6.529 FU32762; DKFZp686N0559; RP11-479G22.1 NM_152377 4.463 FLJ44073; MGC34837 NM 002310 4.386 SWS; SJS2; STWS: CD118 NR_001279 4.051 L0C164380; MGC26611; MGC26924 NM 005345 4.008 HSP72; HSPA 1: HSPA I B; I ISP70-1 NM_0064 17 3.879 p44; NITAP44 NM_017638 3.783 p28b; FLJ20045 NN1_001165 3.695 AIPI ; API2; MIHC; CIAP2; HAIP I; HIAP I ; N1ALT2;

NM 000640 3.659 IL-13R: IL I 3BP; CD213A2 Mk/1_001673 3.313 TS11 NM 002526 3.274 NT; eN; NTS; NTE: eNT; CD73: E5NT
NM 003786 3.244 NILP2; NIRP3; ABC31; MOAT-D; eMOAT2; EST90757 NM 005527 3.229 hum70t; EISP7O-HOM
NM 018372 3.7/4 RIF I; FLJ11269; RPI1-96K19.1 NM 133492 3.16 ACER1; N1GC138327; MGC138329 NM 033160 3.101 FLJ32813; MGC35232; DKFZp572C163 DB318210 3.094 DB318210 NM_182751 3.093 CNA43; PR02249; N1GC 126776 NN1_180989 3.089 ITR
NN1_005345 3.048 HSP72; HSPAl; HSPA1B; HSP70-1 NM 005345 3.029 HSP72; HSPA I; HSPA I B; HSP70-1 NM_0002 12 2.993 CD61; GP3A; GPIlla NM 145867 2.991 N1GC33147 NN1_021813 2.976 BACH2 NM 006187 2.943 p100; MGC133260 CR594200 2.942 L00643837 NM 012419 2.941 RGSZ2; RGS-17; IIRGS17 AF038194 /.923 AF038194 NM 018664 2.921 SNFT; BATF3; JUNDM I
NM 017577 2.907 FLJ35862; FLJ40464 NM 144633 2.887 ELK; ELKI; elk3; Kv12.1 NM_144620 2.86 MGC14816; DKFZp31301122 NM_001004301 2.859 FLJ16542; FLJ3414 I
NM 002852 2.847 TSG-14; TNFAIP5 NM 007107 2.839 TRAPG; SSR gamma NN1_032778 2.836 MDIG; N052: MINA53; FLJ14393; DKFZp76201912 NN1_032523 2.828 ORP6; FLJ36583: MGC59642 NM_005515 2.808 HB9; SCRA I; HOXHB9 NM 002201 2.804 CD25; I I ENI45 NM_152649 2.799 FLJ34389 NM 033036 2.794 GAL3ST2; GAL3ST-3; MGC142112; MGCI42114 NN1_006509 2.791 1-REL
NM 004233 2.789 BL11;11B15 NM_180989 2.77/ 1TR
NM_020988 2.735 GNAO; G-ALP1-LA-o; DKFZp68600962 U16307 2.687 GLIPR; RTVP I; CRISP7 NN1_003706 /.67/ CPLA2-gamma: DKFZp586C0423 NM_153689 2.662 FLJ38973 NM 000800 2.653 AFGF; ECGF; FGFA; ECGFA; ECGFB; HBCIFI; GLI0703; etc BC043212 2.643 L0C402125 NN1_002670 2.619 I-PLASTIN
NM 152408 2.614 FLJ35779; MGC120442; MGC120443; NIGC120444 NM 198951 2.613 TG2; TGC
NM 012329 2.595 NINIA; PAQR I I
NM 001009954 2.589 FLJ20105; MGC131695 NM 032228 2.585 FAR1; FLJ22728; FLJ33561 A1369525 2.584 A1369525 NM 004170 2.583 EAAC I ; EAAT3 NM 002930 2.571 RIN; RIBA; ROC2 AK023856 2.569 L0C339803 NM_024525 2.558 FLJ22584 NM_152649 2.553 FLJ34389 NM 181795 2.552 PRKACN2; FLJ23817 BC039151 2.549 PABPC IL: 11.142053; (1.1 I 069P2.3 NM_006547 2.525 INIP3; KOC1; IMP-3; VICKZ3; DKFZp686F1078 NM 000641 2.5-71 AGIF; IL-11 NM_I45306 2.506 C 1 Oorf35 AK021804 0.398 AK021804 NM 007211 0.397 HoJ-1; Cl 2orf2 NM 203434 0.397 MGC70833; bA247Al2.2 NM 000362 0.397 SFD; K222; K222TA2; FISMRK.227 AK056703 0.395 LOC219731 NM 003558 0.395 NISS4; STNI7 NM 016831 0.394 0I013 NM_024861 0.394 FLJ22671; MGC150431; MGC150432 BF5 14513 0.393 BF514513 NR_002819 0.392 MALAT- I
NM 002609 0.391 JTK12; PDGFR; CD140B; PDGFRI; PDGF-R-beta NM 018027 0.391 FRMD4; FLJ10210; KIAAI294; bA295P9A
NM_001010911 0.39 bA418C1.3 AW444553 0.389 FAM84B
AK056190 0.388 WHRN; CI P98; USH2D; KIAA1526; RPI 1-9M16.1 NM 175868 0.385 MAGE6; MAGE3B; MAGE-3b; MGC52297 AB051431 0.385 K1AA1644; MGC125851; MGC125852 NM 001003683 0.384 HCAM1; I ISPDE I A; MGC26303 NM 004294 0.384 RF I; NITTRF1; MGC47721 NM 006516 0.383 GLUT; GLUTI; MGCI41895; MGC141896 BX104999 0.382 13X104999 AL713743 0.381 FLJ42875; MGC35434; DKFZp761G0122 NM 000322 0.381 RDS: RP7; rd2; AVMD; FRPH; AOFNID; TSPAN22 NM 007314 0.381 ARG; ABLL
NM 018371 0.376 CliGn; FLJI1264; beta4GaINAcT
NR_002802 0.376 TncRNA
DB527271 0.376 DB527271 NNI_006393 0.374 LNEBL; bA56H7.1; NIGC 119746: NIGC119747 NM_013989 0.372 , 5DII=, TXDI2 NM 017600 0.37 DKFZp434N1033 I
BC011595 0.369 NMB; I IGFIN
AF085968 0.366 AF085968 BC018597 0.365 BC018597 NM_014729 0.362 TOX1; KIAA0808 NM_001003940 0.362 FLJ00065 NM 000372 0.358 OCA I A; OCAIA
NM 019555 0.358 GEF3: STA3; XPLN; MGC118905; DKFZP434F2429 NNI_022115 0.357 PFM15; ZNF298; C21 orl83 NM_I98353 0.355 KCTD8 NM_032434 0.355 KIAA1805; MGC111046 AK055386 0.355 AK055386 NM 006933 0.352 SMIT; SMIT2 CR622110 0.35 CR622110 AW856073 0.347 AW856073 NM 015074 0.345 KLP; CMT2; CMT2A; CNIT2A1; HMSNII
NM 032866 0.342 JACOP; FLJ14957; KIAA1749; MGC138254 NN1_012377 0.342 0R7C3; OR19-18; CIT-11SP-87M17 NM 005261 0.341 KIR; MGC26294 AK023391 0.339 AK023391 NM 002214 0.339 ITGB8 NNI_I 82728 0.339 LAT2: LPI-PC1 NM 024050 0.338 DDA I; PCIAl; MGC2594 NM 005185 0.338 CLP
NM 016613 0.337 AD021; AD036: FLJ38155; DKFZp434L142 NM 000782 0.336 CP24; CYP24; MGC126273; MGC126274; P450-CC24 NNI_001624 0.335 ST4 NM 007282 0.333 RZF; MGC13689 NN1_001037442 0.327 RIPX; KIAA0871 N1%1_004318 0.32 BAH; HAAH; JCTN; junctin; CASQ2BP1 BU943730 0.32 BU943730 NM_205849 0.319 FLJ40182 NM_000808 0.315 MGC33793 NNI_033260 0.313 HFHI
NNI_000916 0.309 OT-R
NM 032188 0.309 MOF; hM0F; FLJ14040 BX360933 0.306 SLC25A5 NM 014351 0.304 NST; BRSTL I; SULTX3; BR-STL-1: NIGC40032; DJ388M5.3;
etc NNI_002167 0.3 HEIR-1 NM 001033086 0.298 c1J631M13.5: RP11-189J1.1 NM 000372 029/ OCA1A; OCALA
NNI_001002926 0.289 TWISTNB
AK094143 0.288 CI 4orf78; KIAA2019 NM_004466 0.287 GPC5 NM_031466 0.276 NIBP; T1; IBP; MGC4737; MGC4769; K1AA1882 NM_013261 0.271 LEM6; PGC1; PGC I A; PGC- I v; PPARGC I ; PGC-1(alpha) NM 000693 0.267 ALDH6; RALDH3; ALDHIA6 NM 016125 0.26 L0051136; MGC111090 AK1-24390 0.23 AK124390 NM_002104 0.228 TRYP2 NM 005341 0.209 HKR3; pp9964 NM 173082 0.202 FLJ27258; FLJ37625; FLJ450 I 2 NM 173550 0.191 FLJ39267; FLJ46740; MGC50805 NN4_004185 0.133 WNTI3; XWNT2 NM_021727 0.0415 CYB5RP; LLCDL3 Microarray targets that hybridized with labeled RNA from fibroBADrUL131-infected ARPE- 19 cells were compared to mock-infected cells, and probe sets whose levels varied by ?-'2.5 fold are listed. The Genebank designation, fold change and gene name are listed.
At 6 h after epiBADrUL131 infection, the levels of 47 RNAs were changed as compared to mock-infected cells, and 121 RNAs were altered infibroBADrUL131-infected versus mock-infected cells. The set of modulated RNAs was substantially different for the two viruses: only 19 RNAs were altered after infection with either ep1BADrUL131 or fibroBADrUL131. Although there might be several instances in which a gene was altered by one virus by a factor of 22.5-fold, while the other virus induced a more modest alteration that fell below the cut-off, inspection of the data revealed that this was not common. At 10 hpi.
the number of host cell RNAs modulated by epiBADrU L131 increased only slightly (50 RNAs), whereas a more substantial increase was observed forfibroBADrUL131 (153 RNAs). At the later time, the number of RNAs modulated by both viruses increased to a limited extent (28 RNAs). The microarray results were confirmed by real time RT-PCR for one RNA that was not altered and six RNAs that were altered by infection (Fig.
6B).
To further compare the modulation of RNA levels byfibroBADrUL131 versus epiBADrUL131. the array results were filtered using a gene list comprised of four Gene Ontology groups: host-pathogen interaction (GO:0030383). cell communication (GO:0007154). viral life cycle (GO:0016032) and cell-cell signaling (GO:0007267). Nearly one third of the mRNAs (70 of 222) that were regulated greater than 2.5 fold in fibroBADrUL131-infected ARPE-19 cells were present in the combined grouping (Table 6).
In marked contrast, only one of 86 RNAs induced by ep1BADrUL131 was found in these four Gene Ontology groups. The two virus preparations generated substantially different transcriptional responses upon infection of epithelial cells.
Table 6. libroBADrUL131-modified cellular RNA levels Fold Change Fold Change Genbank 6hpi 10hpi Gene Name NM 006509 7.008 2.791 1-REL
NL2; ARP4; FIAF; PGAR; 1-1FARP; pp1158;
NN1_139314 6.679 2.067 ANGPTL2 NN1_002982 5.977 nc HCI1; MCAF; MCP1; MCP-1; SCYA2.; GDCF-2;
etc NM 000212 4.9/ 2.993 CD61; GP3A; GPIIIa NM 002310 nc 4.386 SWS; SJS2; STWS; CD118 NN1_004464 4.183 2./64 HBGF-5; Smag-82 NM_021101 4.072 nc CLD1; SEMP1; ILVASC
NIN1_006851 4.07 2.364 GLIPR; RTVP1; CRISP7 NM 005347 3.648 nc BIP; MIF2; GRP78: FLJ26106 U16307 3.36 2.687 GLIPIR: RTVPI: CRISP7 NM 000800 3.335 2.115 AFGF: ECGF: FGFA; ECGFA; ECGFB: HBGF1;
etc NM 002526 nc 3.274 NT; eN; NT5; NTE; eNT; CD73; E5NT
NM 005527 nc 3.229 hum70t; IISP7O-HONI
NM 002053 3.21 nc GBPI
NM 180989 3.117 nc ITR
NM 000640 3.116 3.659 1L-13R; 11.13BP; CD213A2 NM_002658 3.09 nc ATF; UPA; URK; u-PA

NM_I 80989 nc 3.089 ITR
NNI_018284 3.076 2.185 FLJI0961; DKFZp686E0974; DKFZp686L15228 NM 005345 nc 3.048 FISP72; HSPAI; HSPA I B; I ISP70-1 NM 000201 3.011 nc BB2; CD54; P3.58 NNI_004556 2.971 nc IKBE
NM_006611 2.954 2.263 Ly49; KLRAii; LY49L; Ly-49L; MGC126520;
etc RIG-I; FLJ 13599; DKF2p4341J 1111:
NNI_014314 2.935 2.071 DKFZp686N19181 NM 003897 2.906 nc DIF2; IEX I; PRG I ; DIF-2; GLY96; IEX-1; 1EX-NM 006417 2.899 3.879 p44; MTAP44 NM 144633 nc 2.887 ELK; ELK 1 ; elk3: Kv12.1 NM 006187 2.877 2.943 p100; MGCI33260 NM 032778 nc 2.836 MDIG; N052; MINA53; FLJ14393;
DKFZp76201912 N1LP2; NIRP3; ABC31; MOAT-D; cNIOAT2;
NNI_003786 2.831 3.244 EST90757 NM_001511 /.8/9 nc GRO I ; GROa; MGSA; NAP-3; SCYB1; MGSA-a; etc NM 001901 /.8.71 nc CCN2; NOV2; HCS24; IGFBP8; MGC102839 NM 002849 2.766 nc PTPRQ; EC-PTP; PCPTP I ; l'TP-SL; PTPBR7 NNI_002234 2.745 nc HK.2; FICK I ; PCN1: HPCNI; KV1.5; MGC
117058; etc NM 198569 2.739 2.182 DREG; VIGR; PS1TP2 NM 000970 2.60 tic TXREB I; SHUJUN-2; TAXREBI07 NM_I 98951 nc 2.613 TG2; TGC
NM 015359 /.61/ nc ZIP14; cig19; LZT-Hs4; KIAA0062 NNI_001548 7.60/ nc GIOPI: IF156; ISG56; IF1-56; IFNAII; RNM561;
etc NM_012329 nc 2.595 NIMA; PAQR11 NM 002930 nc 2.571 IUN; RIBA; ROC2 NM 004233 2.563 2.789 BL1 I: HBI5 NM 020683 2.562 nc A3AR: AD026; bA552M11.5; RP11-552M I 1.7 NM_I8 I 795 nc 2.552 PRICACN2; FLJ23817 NN1_006547 nc 2.525 IMP3; KOCI; IMP-3: VICKZ3; DKFZp6861:1078 NM 000641 nc 2.521 AGIF; IL-11 NM 172345 2.505 nc NM _172345 NM 012419 2.359 2.941 RGSZ2; RGS-17; taGS17 NM 020988 2.188 2.735 GNAO; G-ALPHA-o; DKFZp68600962 NM_00220 I 2.028 2.804 CD25; FIEM45 NM 004318 0.495 0.32 BAH; HAAH; JCTN; junctin; CASQ2BP I
NM 013989 0.492 0.372 D2: 5D11; SelY; TXDI2 NNI_005261 0.468 0.341 KIR: MGC26294 NM_000916 0.434 0.309 OT-R
NM 014351 0.408 0.304 NST: BRSTL I; SULTX3; BR-STL-1; MGC40032;
etc NM 002609 0.4 0.391 JTK12; PDGFR; CD140B; PDGFR1; PDGF-R-beta NM_0072 I 1 tic 0.397 HoJ- 1 ; Cl2or12 NN1_001003683 nc 0.384 HCANI I ; liSPDE IA; MGC26303 NM 006516 nc 0.383 GLUT; GLUT I; MGC141895; MGC141896 NN4_000322 nc 0.381 RDS; RP7; rd2; AVMD; PRPI I; AOFN1D;

NNI_021990 0.378 0.49 GABRE
NM 018371 'lc 0.376 ChGn; FLJI1264; beta4GaINAcT
NM 019555 0.371 0.358 GEF3; STA3; XPLN; MGC I 18905;
DKFZP4341:2429 NM_007314 0.347 0.381 ARG; ABLL
NM_002214 nc 0.339 ITGB8 NM 182728 tic 0.339 LAT2; LPI-PC I
BC015929 0.338 0.453 RVR; BD73; HZF2; EAR-1r; I Is.37288 - 34.

NM_016831 0.287 0.394 GIG13 NM 013261 0.236 0.271 LEM6: PGC1; PGC I A; PGC-1v; PPARGC1; etc NNI_003862 0.194 nc ZFGF5; FGF-18 Four GO groups were combined: host-pathogen interaction (GO:0030383), cell communication (GO:0007154), viral life cycle (GO:0016032) and cell-cell signaling (GO:0007267). The set of 9276 genes was used to filter array results from libroBADrUL131-infected ARPE-19 cells.
Genbank identifiers and gene names are shown along with the fold induction or repression at 6 and 10 hpi.
Probe sets that did not change by ?2.5 compared to mock-infected cells are designated by "nc" for no change.
Discussion ARPE-19 epithelial cells can be infected by HCMV through two different routes:

fusion at the plasma membrane or endocytosis followed by fusion at the endosomal membrane.
Both modes of entry initiate a productive infection. The route of entry depends on the cell type in which the virus was propagated. HCM V from epithelial cells enters by the former route.
and virus grown in fibroblasts follows the latter path. This conclusion follows from ultrastructural analysis and differential sensitivity of infection to agents that block acidification of endosomes. The observation that virus grown in epithelial cells has greater "fusion from without- activity than does virus produced in fibroblasts reinforces the view that the two virus preparations interact with ARPE-19 cells in a fundamentally different manner.
Importantly, both modes of entry require pUL130 function because pUL130 antibody neutralized infection by virus produced from either source. The gH/gL/pUL128/pUL130/pUL131 complex functions at the ARPE-19 plasma membrane if the infecting virus has been produced in epithelial cells and at the endosomal membrane if the virus was grown in fibroblasts.
Neutralized virus in the endosome fails to escape and presumably suffers the same fate as AD169. which lacks the pUL130-containing complex and accumulates in epithelial cell endosomes without initiating a productive infection (10).
Virus grown in fibroblasts induces 1E1 protein accumulation in ARPE-19 cells after a delay relative to virus from epithelial cells. suggesting that some aspect of entry by endocytosis proceeds more slowly than entry by fusion at the plasma membrane.
Many virions are evident in endosomes, but no capsids were seen in the cytoplasm after entry of fibroblast-generated virus: and capsids were found rarely in the cytoplasm of cells infected with epithelial cell-produced virus. Apparently, virions linger for a time in endosomes. but once a capsid is freed of its envelope and reaches the cytoplasm. it is rapidly disassembled.
How are HCMV virions produced in the two cell types different? It appears different -fusion from without- activities provide an indication. Not only did epiBADrUL131 induce fusion more efficiently than fibroBADrU L131, but lowered pH enhanced the activities of both virus preparations. Without intending to be bound or limited by any explanation of mechanism, it is possible that fusion of membranes requires a threshold of fusion activity.
The ability of pULI30 antibody to neutralize both virus preparations indicates that both depend on the gH/gL/pUL128/pUL130/pUL131 complex for fusion, so experiments were devised to the hypothesis that the viruses contain different amounts of the complex. Several of its constituents were assayed, and it was found that a slightly higher ratio (-2-fold) of gH
/gL/pUL128/pUL130/pUL131 to gH/gL/g0 were present in epiBADrUL131 particles than in fibroBADrUL131 particles. The levels of gB, pp28 and pp65 were similar in the two virion preparations.
There is precedent in EBV for production of viruses with different relative amounts of a 01 complex: particles produced by B cells are deficient for gH/gL/gp42 (18).
However.
other factors may be involved. Perhaps a constituent of the complex that was not assayed is altered. Alternatively, the ratio of the gH complex to one or more additional virion glycoprotein complexes might modify fusion activity. Finally, it may be that an unidentified cell protein, supplied to the virions when they are produced within epithelial cells or fibroblasts, might alter the complex.
Are there physiological consequences to the two modes of entry? epiBADrUL131 andfibroBADrUL131 induced markedly different cellular transcriptional responses after infection of ARPE-19 cells. Assuming that the difference is indeed due to virions or virions plus specifically associated cellular factors. the microarray experiment demonstrates a strikingly different transcriptional response to infection. Endocytosis is intimately involved in the regulation of signaling by cell surface molecules. As a consequence. a virus might modulate cell signaling, and the cellular transcriptome, differently if it enters by fusion at the plasma membrane versus endocytosis. The differences in cell signaling likely have physiological consequences that are not detected in cultured cells. such as effects on virus spread, immune evasion, or virulence.
References:
1. Miller. N. & Hutt-Fletcher, L. M. (1992)J Virol 66, 3409-14.
2. Nemerow. G. R. & Cooper, N. R. (1984) Virology 132, 186-98.
3. Nicola. A. V., Hou. J., Major, E. 0. & Straus, S. E. (2005) J Virol 79, 7609-16.
4. Nicola, A. V., McEvoy, A. M. & Straus, S. E. (2003)J Virol 77, 5324-32.
5. Milne, R. S., Nicola, A. V., Whitbeck. J. C., Eisenberg. R. J. & Cohen.
G. H. (2005)J
Virol 79, 6655-63.
6. Wittels, M. & Spear. P. G. (1991) Virus Res 18, 271-90.
7. Plachter, B., Sinzger, C. &. Jahn, G. (1996) Aciv Virus Res 46, 195-261.
8. Compton, T.. Nepomuceno, R. R. & Nowlin, D. M. (1992) Virology 191, 387-95.

9. Bodaghi. B., Goureau. O.. Zipeto, D., Laurent. L., Virelizier. J. L. &
Michelson. S.
(1999)J Inztitzmol 162. 957-64.
10. Ryckman. B. J.. Jarvis. M. A.. Drummond, D. D., Nelson. J. A. &
Johnson. D. C.
(2006)1 Virol 80, 710-22.
11. Wang. X.. Kenyon. W. J.. Li, Q., Mullberg, J. & Hutt-Fletcher, L. M.
(1998) J Virol 72, 5552-8.
12. Li, Q., Turk, S. M. & Hutt-Fletcher, L. M. (1995) J Virol 69, 3987-94.
13. Hutt-Fletcher, L. M. & Lake, C. M. (2001) Curt- Top ilificrobiol Immunol 258, 51-64.
14. Haan. K. M.. Kwok. W. W.. Longnecker. R. & Speck, P. (2000) J Virol 74, 2451-4.
15. Haan. K. M. & Longnecker. R. (2000) Proc Nati Acad USA 97, 9252-7.
16. Li. Q.. Spriggs. M. K.. Kovats. S.. Turk, S. M., Comeau. M. R.. Nepom, B. & Hutt-Fletcher, L. M. (1997) J Virol 71, 4657-62.
17. Wang, X. & Hutt-Fletcher. L. M. (1998)1 Virol 72, 158-63.
18. Borza. C. M. & Hutt-Fletcher, L. M. (2002) Nat Med 8, 594-9.
19. Wang. D. & Shenk, T. (2005) Proc Nati Acad Sci USA 102, 18153-8.
20. Adler. B.. Scrivano. L.. Ruzcics, Z., Rupp. B.. Sinzger. C. &
Koszinowski. U. (2006) J Getz Virol 87, 2451-2460.
21. Wang, D. & Shenk, T. (2005)1 Virol 79, 10330-8.
22. Hahn, G., Revell , M. G.. Patrone, M., Percivalle. E.. Campanini. G..
Sarasini. A., Wagner, M., Gallina, A., Milanesi, G., Koszinowski, U.. Baldanti, F. & Gerna.
G.
(2004)1 Virol 78, 10023-33.
The present invention is not limited to the embodiments described and exemplified above, but is capable of variation and modification within the scope of the appended claims.

Claims (14)

What is claimed is:
1. A method of making an immunogenic composition comprising a population of live attenuated, inactivated or killed cytomegaloviruses (CMVs), or virion components thereof, comprising:
a) passaging a strain or an isolate of CMV having a gene encoding a functional pUL131 protein in a fibroblast;
b) amplifying the strain or isolate of CMV in an epithelial cell to produce an amplified strain or isolate of CMV;
c) harvesting the amplified strain or isolate of CMV to obtain a cell-type conditioned CMV; and d) combining the cell-type conditioned CMV with a pharmaceutical carrier or adjuvant, thereby making an immunogenic composition comprising a population of live attenuated, inactivated or killed CMV or virion components thereof.
2. The method of claim 1, wherein the strain or isolate of CMV is a human CMV strain or isolate.
3. The method of claim 1, comprising amplifying the strain or isolate of CMV in two or more different epithelial cells.
4. The method of claim 1, comprising passaging and amplifying two or more CMV strains or isolates and combining the two or more CMV strains or isolates.
5. An immunogenic composition produced by the method of claim 1.
6. A kit comprising a container in which is contained:
a) one or more strains or isolates of CMV having a gene encoding a functional pUL 1 3 1 protein;
b) fibroblasts; and c) epithelial cells.
7. An immunogenic composition comprising a population of live attenuated, inactivated or killed cytomegaloviruses (CMVs) having a gene encoding a functional pUL131 protein, or virion components thereof, admixed with a suitable pharmaceutical carrier or adjuvant, wherein the CMV population has been passaged in fibroblasts, amplified in epithelial cells, and harvested from the epithelial cells.
8. The immunogenic composition of claim 7, wherein the CMV population has been passaged in human fibroblasts and amplified in human epithelial cells.
9. The immunogenic composition of claim 7, wherein the CMV population is characterized by one or more features in subsequently infected host cells, said features selected from:
a) entry into the host cells by fusion with host cell plasma membranes;
b) greater virion-mediated cell-cell fusion of the host cells as compared with an equivalent CMV population isolated from cultured fibroblasts;
c) accelerated virus growth in the host cells as compared with an equivalent CMV
population isolated from cultured fibroblasts;
d) elicitation of a cellular response involving changes in expression greater than or equal to 2.5 fold of about two thirds fewer genes than a response elicited by an equivalent CMV population isolated from cultured fibroblasts at 10 hours post-infection:
and e) elicitation of a cellular response involving a change in expression of one or more genes represented by GenBank Accession Nos: AK094860, NM_145023, NM_133492, NM_001039580, NM_001004301, NM_001034, A1369525, AK123066, NM_005345, NM_020731, BC071797, NM_003414, NM_000800, NM_138467, AK090803, AL133118, NM_001165, BG001037, NM_024861, NM_001043, NM_016239, NM_001018084, NM_001037442, NM_017600, NM_022097, NM_175868. NM_032266, NM_003841, NM_005039, NM_145051, NM_004294, AW856073, NM_024050, AF085968, NM_080927, NM_022115, AK056703, NM_000808, NM_012377, NM_006793, NM_031466, NM_005185, NM_139173, BX360933, NM_016125, NM_002104, NM_032188, NM_004185, NM_004843 and NM_173550.
10. The immunogenic composition of claim 7, wherein the CMV population comprises two or more different CMV strains or clinical isolates.
I 1. Use of an immunogenic composition comprising a population of live attenuated, inactivated or killed cytomegaloviruses (CMVs) having a gene encoding a functional pUL131 protein, or virion components thereof, admixed with a suitable pharmaceutical carrier or adjuvant, wherein the CMV population has been passaged in fibroblasts, amplified in epithelial cells, and harvested from the epithelial cells, for immunizing an individual against CMV.
12. Use of an immunogenic composition comprising a population of live attenuated, inactivated or killed cytomegaloviruses (CMVs) having a gene encoding a functional pUL131 protein, or virion components thereof, admixed with a suitable pharmaceutical carrier or adjuvant, wherein the CMV population has been passaged in fibroblasts, amplified in epithelial cells, and harvested from the epithelial cells, for the preparation of a medicament for immunizing an individual against CMV.
13. The use of claim 11 or 12 wherein the immunogenic composition is the immunogenic composition as defined in any one of claims 7 to 10.
14. The use of any one of claims 11 to 13, wherein the individual is a human.
CA2701727A 2007-10-10 2008-10-10 Cytomegalovirus vaccines and methods of production Active CA2701727C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US99842607P 2007-10-10 2007-10-10
US60/998,426 2007-10-10
PCT/US2008/079494 WO2009049138A1 (en) 2007-10-10 2008-10-10 Cytomegalovirus vaccines and methods of production

Publications (2)

Publication Number Publication Date
CA2701727A1 CA2701727A1 (en) 2009-04-16
CA2701727C true CA2701727C (en) 2016-12-13

Family

ID=40549581

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2701727A Active CA2701727C (en) 2007-10-10 2008-10-10 Cytomegalovirus vaccines and methods of production

Country Status (15)

Country Link
US (2) US9439960B2 (en)
EP (1) EP2200645B1 (en)
JP (1) JP5409638B2 (en)
KR (1) KR20100084627A (en)
CN (1) CN101820906B (en)
AU (1) AU2008310713B2 (en)
BR (1) BRPI0818656A2 (en)
CA (1) CA2701727C (en)
IL (1) IL204850A0 (en)
MX (1) MX2010003713A (en)
NZ (1) NZ584459A (en)
RU (1) RU2491340C2 (en)
SG (1) SG185291A1 (en)
WO (1) WO2009049138A1 (en)
ZA (1) ZA201002216B (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9439960B2 (en) 2007-10-10 2016-09-13 The Trustees Of Princeton University Cytomegalovirus vaccines and methods of production
US8580276B2 (en) 2009-06-05 2013-11-12 City Of Hope Genetically stable recombinant modified vaccinia ankara (rMVA) vaccines and methods of preparation thereof
TWI570240B (en) 2011-09-09 2017-02-11 默沙東公司 A conditional replicating cytomegalovirus as a vaccine for cmv
WO2013144722A2 (en) * 2012-03-27 2013-10-03 Variation Biotechnologies, Inc. Methods for detection of anti-cytomegalovirus neutralizing antibodies
CN104853771A (en) 2012-07-06 2015-08-19 诺华股份有限公司 Complexes of cytomegalovirus proteins
JP6419695B2 (en) 2012-07-27 2018-11-07 シティ・オブ・ホープCity of Hope MVA vaccine for delivery of UL128 complex and prevention of CMV infection
US9395369B2 (en) 2012-10-19 2016-07-19 Regents Of The University Of Minnesota Guinea pig cytomegalovirus (CIDMTR strain)
CN103937871B (en) * 2013-01-23 2019-07-23 上海市东方医院 The application of SRRP35 gene and expression product in cancer diagnosis and treatment
US20160289303A1 (en) * 2013-11-15 2016-10-06 President And Fellows Of Harvard College Methods and compositions for the treatment of hcmv
US10611800B2 (en) 2016-03-11 2020-04-07 Pfizer Inc. Human cytomegalovirus gB polypeptide
EP3601543A4 (en) * 2017-03-30 2021-01-27 Merck Sharp & Dohme Corp. Addition of nucleases directly to cell culture to facilitate digestion and clearance of host cell nucleic acids
CA3060019A1 (en) * 2017-04-19 2018-10-25 Glaxosmithkline Biologicals Sa Modified cytomegalovirus proteins and stabilized complexes
CA3079828A1 (en) * 2017-11-01 2019-05-09 Merck Sharp & Dohme Corp. Stable formulations of cytomegalovirus
US11629172B2 (en) 2018-12-21 2023-04-18 Pfizer Inc. Human cytomegalovirus gB polypeptide
TWI810589B (en) 2020-06-21 2023-08-01 美商輝瑞股份有限公司 Human cytomegalovirus gb polypeptide

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3959466A (en) * 1974-04-15 1976-05-25 The Wistar Institute Highly attenuated cytomegalovirus vaccine and production thereof
US4058598A (en) * 1974-10-18 1977-11-15 Harold Stern Cytomegalovirus attenuation method and vaccine
US6471965B1 (en) * 1994-07-27 2002-10-29 Bio-Virus Research Incorporated Vaccine containing whole killed herpes viruses to prevent development of atherosclerotic plaque
DE10144906B4 (en) * 2001-09-12 2013-11-28 Novartis Vaccines And Diagnostics Gmbh Process for the large-scale production of vaccines
US7407744B2 (en) * 2003-07-25 2008-08-05 The Regents Of The University Of California Cytomegalovirus gene function and methods for developing antivirals, anti-CMV vaccines, and CMV-based vectors
US7704510B2 (en) * 2006-06-07 2010-04-27 The Trustees Of Princeton University Cytomegalovirus surface protein complex for use in vaccines and as a drug target
US9439960B2 (en) 2007-10-10 2016-09-13 The Trustees Of Princeton University Cytomegalovirus vaccines and methods of production

Also Published As

Publication number Publication date
RU2491340C2 (en) 2013-08-27
CN101820906B (en) 2015-04-22
SG185291A1 (en) 2012-11-29
EP2200645B1 (en) 2016-01-27
IL204850A0 (en) 2010-11-30
BRPI0818656A2 (en) 2015-04-14
US9439960B2 (en) 2016-09-13
US20100285059A1 (en) 2010-11-11
NZ584459A (en) 2012-06-29
AU2008310713A1 (en) 2009-04-16
JP2011500592A (en) 2011-01-06
ZA201002216B (en) 2011-06-29
WO2009049138A1 (en) 2009-04-16
CN101820906A (en) 2010-09-01
MX2010003713A (en) 2010-08-02
EP2200645A4 (en) 2012-07-11
US20170014504A1 (en) 2017-01-19
KR20100084627A (en) 2010-07-27
US10300130B2 (en) 2019-05-28
RU2010117741A (en) 2011-11-20
EP2200645A1 (en) 2010-06-30
AU2008310713B2 (en) 2012-06-14
CA2701727A1 (en) 2009-04-16
JP5409638B2 (en) 2014-02-05

Similar Documents

Publication Publication Date Title
US10300130B2 (en) Cytomegalovirus vaccines and methods of production
JP5926804B2 (en) Conditional replication cytomegalovirus as a vaccine for CMV
JP5822433B2 (en) Cytomegalovirus surface protein complex for use as a drug target in a vaccine
US20130045230A1 (en) Composition containing hcmv particles
Gray Simian varicella in old world monkeys
Donofrio et al. Outcome of bovine herpesvirus 4 infection following direct viral injection in the lateral ventricle of the mouse brain
US20130195918A1 (en) Equine Herpesvirus 1 Vaccine and Vector and Uses Thereof
Shafik An overview of Immunopathogenesis towards COVID-19 infection
Polz-Dacewicz et al. Cytomegalovirus (CMV)-a new prospect for prevention
Langhoff et al. Pneumonitis in human cytomegalovirus infection

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20130718