专利摘要:
Figure 1 The present invention relates to a recombinant human cytomegalovirus (CMV) protein dimer complex comprising the CMV gH protein or one of its complex-forming fragments, and CMV UL116 or a complex-forming fragment thereof. Nucleic acids encoding said gH / ULH6 dimer complex, host cells for recombinant expression of said gH / ULH6 dimer complex, and the use of said gH / UL116 dimer complex for use as an antigen are also provided herein. vaccine. Figure 1
公开号:BE1023077B1
申请号:E2016/5051
申请日:2016-01-22
公开日:2016-11-17
发明作者:Luca Bruno;Stefano Calo;Andrea Carfi;Mirko Cortese;Marcello Merola
申请人:Glaxosmithkline Biologicals S.A.;
IPC主号:
专利说明:

CMV ANTIGEN AND USES Field of the invention
The present invention relates to a cytomegalovirus (CMV) antigen suitable for vaccine uses.
Context
Human cytomegalovirus (HCMV) causes persistent and widespread human infections that vary with the age and immunocompetence of the host. Primary infection of hosts with a functioning immune system is associated with mild symptoms, although it progresses with fever, hepatitis, splenomegaly and mononucleosis. In contrast, when primary infection or reactivation occurs in immunocompromised or immunodeficient hosts, the latter often experience life-threatening diseases, including pneumonia, hepatitis, retinitis and encephalitis (Sinclair and Sissons, J. Gen. Virol 87: 1763-1779, 2006). HCMV infection has been recognized for its association with three different populations: neonates with immature immune systems; transplant recipients with compromised immune systems due to the use of drugs and HIV-infected patients with compromised immune systems due to the decline of CD4 + T cells.
HCMV can be particularly devastating in newborns, causing abnormalities in neurodevelopment. In industrialized countries, intrauterine viral infection is the most common. Estimates suggest that between 0.6% and 0.7%, depending on the seroprevalence of the population examined, all neonates are infected in utero (Dollard et al., Rev. Med., Virol., 17 (5)). ): 355-363, 2007). In the United States alone, this corresponds to approximately 40,000 new infections per year. Around 1.4% of CMV intrauterine infections result from transmission by women with established infection. A new maternal infection occurs in 0.7 to 4.1% of pregnancies and is transmitted to the fetus in about 32% of cases. Around 90% of infected infants are asymptomatic at birth and most will develop serious sequelae of infection over several years, including mental retardation and hearing loss. Other infected children have symptomatic HCMV disease with symptoms of irreversible central nervous system involvement such as microencephalitis, encephalitis, epileptic seizures, deafness, upper motor neuron disorders, and psychomotor retardation (Kenneson et al. Rev. Med Virol., 17 (4): 253-276, 2007). In summary, in the United States, 8,000 children a year develop a neurological disease associated with the virus. Congenital infection is the main driving force behind efforts to develop a vaccine against HCMV.
Efforts to develop an HCVV vaccine began more than 40 years ago. Over the years, a number of HCMV vaccines have been evaluated, including a whole virus vaccine, chimeric vaccines and subunit vaccines. The whole virus vaccine prevented neither infection nor viral reactivation in immunized adult women, nor did it increase protection against disease compared to HIV-positive individuals (Arvin et al., Clin Infect Dis 39 (2), 233-239, 2004). Each of the chimeric vaccines was well tolerated, but concerns about the potential risk of establishing a latent infection hampered the progression of these vaccines. The subunit vaccine approach, based on the assumption that immunity is directed against a limited number of dominant antigens, has shown poor efficacy to date. These results suggest that an effective vaccine may need to be directed against multiple antigens expressed at different stages of viral replication.
The glycoproteins of the CMV envelope, gB, gH, gL, gM, gN, UL128, UL130, and UL131, represent attractive candidate vaccines because they are expressed on the viral surface and can trigger protective humoral immune responses that neutralize the virus. . However, there is a need to develop CMV antigen variants suitable for immunization. summary
As disclosed and exemplified herein, the inventors have discovered a new human cytomegalovirus (CMV) protein dimer complex formed by the CMV, gH and UL116 proteins.
Accordingly, the invention relates to a recombinant human cytomegalovirus (CMV) protein dimer complex, comprising the CMV gH proteins or a complex-forming fragment thereof, and CMV UL116 or a fragment thereof forming a complex.
Immunogenic compositions comprising a dimer complex gH / UL116 are also provided herein. The immunogenic complex may further comprise a CMV protein or an additional CMV protein complex. Suitable CMV antigens that can be combined with the gH / ULH6 dimer complex include, for example: gB, gH, gL, gO, gM, gN; UL128, UL130, UL131, RL10, RL11, RL12, RL13, UL4, UL5, UL10.5, UL80.5, UL119, UL122, UL133, UL138, UL148A, UL1, UL7, UL9, UL16, UL18, UL20, UL40, UL41A, ÜL42, ÜL47, UL111A, UL122, UL132, UL136, UL141, one of their immunogenic fragments, or a combination thereof. Suitable CMV protein complexes that can be combined with the gH / UL116 dimer complex include, for example, the pentamer complex gH / gL / UL128 / UL130 / UL131, the gH / gL complex, the trimer complex gH / gL / gO, or one of their combinations.
Nucleic acids encoding the CMV gH / UL116 dimer complex, as described herein, are also provided herein. The nucleic acid may be a single nucleic acid construct (e.g., a single vector) encoding both gH and Ullo. The nucleic acid may also be a combination of nucleic acid constructs (e.g., a first vector encoding gH and a second vector encoding UL116). The nucleic acid or nucleic acids can be used in the form of a nucleic acid-based vaccine (e.g., a self-replicating RNA molecule encoding the gH / ULH6 dimer complex). The nucleic acid can also be used for the recombinant production of the gH / UL116 dimer complex described herein. The invention also provides a host cell comprising the nucleic acids described herein. The host cells can be used to recombinantly express the gH / UL116 dimer complex. Preferably, the dimer gH / UL116 complex may be secreted from the host cell. Preferred host cells are mammalian host cells such as CHO cells or HEK-293 cells. The invention also provides a cell culture comprising the host cell described herein. Preferably, the culture has a size of at least 20 liters, and / or the yield of the gH / UL116 dimer complex is at least 0.1 g / l. The invention also provides a method of inducing an immune response against cytomegalovirus (CMV), comprising administering to a subject in need of an immunologically effective amount of the dimer gH / UL116 complex described herein. The invention also provides a method of inhibiting the entry of cytomegalovirus (CMV) into a cell, comprising contacting the cell with the gH / λ L116 dimer complex described herein.
It is also proposed uses of the compositions described herein for the induction of an immune response against cytomegalovirus (CMVj, and the use of the compositions described herein in the manufacture of a medicament for inducing an immune response against Cytomegalovirus (CMV) The invention further relates to a method of forming the dimer complex gH / UL116 described herein, comprising administering one or more nucleic acids encoding gH and UL116 (or one of their complex-forming fragments) to a cell, and maintaining the cell under conditions suitable for the expression of said gH and UL116 (or of one of their complex-forming fragments), in which the gH dimer complex The cell can be in vivo or in vitro For in vitro purposes, any suitable host cell (e.g., a bacterial host or a eukaryotic cell line) can be used. In vivo, the cell can be an epithelial cell, an endothelial cell, or a fibroblast.
Brief description of the drawings
Figure 1 shows the position of UL116 in the genome of HCMV TR and the conservation of the sequence among HCMV strains adapted to the laboratory and clinical. (A) ORF map of the TR BAC clone used in the examples. The arrows indicate the relative orientations of the repeated and unique ORF blocks. The ull16 gene, in bold, is located between the u1115 (gL) and u117 genes on the antisense strand. (B) T-Coffee Multiple Alignments of Primary Amino Acid Sequences Showing 98% Conservation Degree of the u1116 Gene from a Coherent Group of Human CMV Strains (Top to Bottom, SEQ ID NO: 6, 7, 8 , 9, 10, 11, 12, 13, 14, 15, 16, and 17). The asterisk indicates the 14 predicted N-glycosylation sites. SP indicates the predicted N-terminal signal peptide.
Figure 2 shows the kinetics of expression of UL116 and the addition of carbohydrates in fibroblasts infected with human CMV. (A) Uninfected (pseudo-infected) and TR-UL116-Flag infected HFF-1 cells at a multiplicity of infection (MOI) of five were harvested at the indicated times after infection (p.i.). Equivalent amounts of cell lysates were subjected to SDS-PAGE under reducing conditions and analyzed by immunoblotting with anti-Flag antibody. Antibodies to IE1 of HCMV and protein pp28 as indicated on the left side of the figure. Detection of actin was used as a protein loading control. (B) Five-day TR-UL116-Flag infections of HFF-1 cells were performed in the presence (right lane) or absence (left lane) of phosphonacetic acid (PAA), an inhibitor of late-phase protein expression of HCMV. The lysates were prepared from the infected cells and the expression of UL116 was detected by Western blot analysis using anti-Flag antibody. (C) The lysate of TR-UL116-Flag-infected (Hf-1) whole HFF-1 cells from a single vial was divided into three aliquots for glycosidase digestion (left lane, untreated control; middle lane, endoglycosidase H digestion, right lane, PNGase F). Proteins were separated on SDS-PAGE under reducing conditions and UL110 was revealed by immunoblotting using anti-Flag antibody.
Figure 3 shows the location of UL116 in the virion envelope. Western blot analysis was performed on purified virions from viruses expressing Flag-tagged UL116. The total virion lysate (V), the envelope fraction (E) and the integument / coat fraction (T) were probed for the indicated antigens.
Figure 4 shows the interaction of UL116 with gH in transfected KEK 293T cells. (A) Representation of the detection of UL116 by FACS analysis on non-permeabilized KEK293T cells. Cells were transfected with expression vectors for UL116, gH, and gB both alone and in combination as shown in the figure. Forty-eight hours after transfection, the cells were stained at 4 ° C with mouse polyclonal anti-UL116 sera at different dilutions. The excess probe was removed by washing in PBS and then the cells were fixed and stained with an anti-mouse antibody conjugated to AlexaFluor-488. Fluorescent-positive cells by Alexafluor-488 were found only in UL116 / gH cotransfection configuration and compared to single transfectants for UL116, the gH, gB with the gB / UL116 pair being used as the negative witnesses. The experiments were performed in triplicate to provide a statistically consistent set of data. (B) Representation of the coimmunoprecipitation of UL116-gH. Lysates from the transiently expressing HEK293T cells UL116-his / gH-myc, UL116-his, gB and UL116-his / gB were subjected to parallel immunoprecipitation experiments (used antibodies specified on the left of the figure). with magnetic beads covalently linked to both anti-His and anti-myc. Total lysates (entry) and eluted samples were separated by SDS-PAGE and analyzed by immunoblotting for both His markers. and myc (shown at the bottom of the figure).
Figure 5 shows the interaction of ÜL116 with gH in infected HFF-1 cells. Co-immunoprecipitation (Co-IP) complexes of gH. Cell lysates were prepared from HFF-1 cells infected separately with HCMV-TR-UL116-Flag and wild type TR (wt) (5 days p.i.). The complexes were captured using the anti-gH MSL-109 human monoclonal antibody and magnetic A / G protein beads. The total and eluted samples were immunoblotted using anti-Flag mAb (top panel), anti-gH rabbit serum (middle panel), and anti-gL rabbit serum (panel). bottom).
Figure 6 shows a multi-stage growth curve analysis of the ORF X-Flag TR reconstituted virus and the parental HCMV TR strain. HFF cells seeded in six well plates (5 x 105 cells / well) were infected with an MOI of 0.1. At the indicated time points (days after infection), the supernatants from the infected cultures were harvested, and the total PFUs of the infectious viruses in the culture supernatants were determined by plaque technique on HFF cells. Titers at time point 0 represent input inocula and each data point represents the average of three independent wells.
Detailed Description 1. Protein Complex qH / UL116
As disclosed and exemplified herein, the inventors have discovered a new human cytomegalovirus (CMV) protein dimer complex formed by the CMV gH and UL116 proteins.
Accordingly, in one aspect, the invention relates to a human cytomegalovirus (CMV) protein dimer complex, comprising the CMV gH protein or a complex-forming fragment thereof, and CMV ULll6 or one thereof. of its fragments forming a complex. Although gH, UL116 and several other CMV proteins described herein are sometimes referred to as glycoproteins, this nomenclature should not be taken to mean that these proteins must be glycosylated when used with the invention.
GH CMV Proteins
The human CMV glycoprotein H (gH), encoded by the ÜL75 gene, is a virion glycoprotein that is essential for infectivity and is conserved among members of alpha-, beta-, and gamma-herpesviruses. Based on the crystal structures of the gH / gL complexes of HSV-2 and EBV, the N-terminal residues of gH form a globular domain at one end of the structure (the "head"), which is involved in interactions with gB and activation of membrane fusion. The C-terminal domain of gH, proximal to the viral membrane (the "tail"), is also involved in membrane fusion. The gH has determinants that are recognized by the TLR2 host factor, and it directly interacts with a heterodimer formed between the TLR2 and TLR1 host factors. TLR2 mediates NF-κΒ activation and inflammatory cytokine responses from cells.
The gH from any CMV strain can be used. By way of example, the gH from the CMV Merlin strain is represented by SEQ ID NO: 1 (GI: 52139248, 742 amino acid residues). The gH from the CMV Towne strain is represented by SEQ ID NO: 2 (GI: 138314, also 742 amino acid residues), and the gH from the CMV strain AD169 (GI: 138313) is represented by SEQ ID NO: 3. The gH from other CMV strains, such as strains VR1814, Toledo, TR, PH, TB40 / e, or Fix (aka VR1814) can also be used.
The gH of the Towne strain has been characterized as comprising: (i) six N-glycosylation sites (at residues 55, 62, 67, 192, 641 and 700); (ii) a hydrophobic signal sequence at its N-terminus (amino acid residues 1 to 23); (iii) an ectodomain (residues 24 to 717) that projects out of the cell into the extracellular space; (iv) a C-terminal cytoplasmic domain (residues 737 to 742. SEQ ID NO: 2 shares 99% and 96% amino acid sequence identity with SEQ ID NO: 1, and SEQ ID NO: 3, respectively Generally, the N-terminal signal sequence of the full-length gH protein is cleaved by a host cell peptidase signal to produce a mature gH protein.As such, to the gH protein expressed by the host cell described herein, it may be missed. the N-terminal signal sequence (e.g., gH is encoded by a nucleotide sequence lacking the coding sequence for the N-terminal signal sequence).
It is also encompassed by the invention, gH complex-forming fragments, such as a fragment of gH lacking the transmembrane domain (TM) (e.g., residues 718-736 of SEQ ID NO: 2), the C-terminal domain (e.g., residues 737 to 742 of SEQ ID NO: 2), the N-terminal signal sequence (e.g., residues 1 to 23 of SEQ ID NO: 2), or one of their combinations. A complex-forming fragment of gH may be any portion or portion of the gH protein that retains the ability to form a complex with another CMV protein. In some embodiments, a complex forming fragment of gH forms part of the gH / UL116 dimer complex. For example, expression of the full-length gH sequence may interfere with the purification of the soluble dimer complex because the TM domain of the gH is hydrophobic. Instead, the gH / UL116 dimer complex may comprise a fragment of the gH in which at least a portion of the TM domain of the gH is deleted.
For example, a fragment of gH comprising the N-terminal signal sequence and the ectodomain of gH, but not the TM domain, may be used. An appropriate fragment of the gH may also comprise a portion of the ectodomain of the gH (e.g., at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%). %, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of the ectodomain sequence of gH), but none, or only a small portion of the TM domain. Alternatively, at the fragment of the gH described here, there may be between 1 and 20 amino acid residues missing (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 , 13, 14, 15, 16, 17, 18, 19 or 20 amino acid residues, or missing residues 1 to 20, residues 1 to 15, residues 1 to 10, residues 2 to 20, residues 2 to 15, residues 2 to 10, residues 5 to 20, residues 5 to 15, or residues 5 to 10) at the N-terminus and / or the C-terminus of the solid ectodomain length. Residues at the C-terminal domains are not thought to be necessary for immunogenicity. An example of an appropriate fragment of the gH described herein is SEQ ID NO: 4, which corresponds to amino acid residues 1 to 715 of SEQ ID NO: 1. Another example of a fragment of the gH described here is represented by SEQ ID NO: 5, which lacks the N-terminal signal sequence, the TM domain and the C-terminal domain of gH, and which corresponds to amino acid residues 24 to 715 of SEQ ID NO: 1. example of a fragment of the gH comprises the entire N-terminal signal sequence and the ectodomain, but it lacks the C-terminal domain. The ectodomain of gH corresponds to the extracellular domain of gH. The location and length of the ectodomain of a gH (or one of its homologs or variants) can be predicted based on a pairwise alignment of its sequence at SEQ ID NO: 1, 2, 3 , 4, or 5, for example by aligning the amino acid sequence of a gH with SEQ ID NO: 1, and identifying the sequence that aligns with residues 24 to 717 of SEQ ID NO: 1. similarly, the locations of the signal sequence, TM domain, and C-terminal domain can be predicted by aligning the amino acid sequence of a gH with SEQ ID NO: 1, 2, 3, 4, or 5 , and identifying sequences that align with corresponding regions (e.g., residues 1 to 23 (signal sequence), 718 to 736 (TM) and 737 to 742 (C-terminal domain) of SEQ ID NO: 1 , respectively). Alternatively, the location and length of the ectodomain, signal sequence, TM domain, and C-terminal domain can be predicted based on the computerized analysis of hydrophobicity along the entire length of the a given sequence of gH. The signal sequence and the TM domain have the highest levels of hydrophobicity and these two regions flank the less hydrophobic ectodomain.
An appropriate complex-forming fragment of the gH can also be obtained or determined by standard techniques known in the art, such as a co-immunoprecipitation technique, crosslinking, or fluorescent staining colocalization, etc. SDS-PAGE or Western blot analysis may also be used (for example, showing that both subunits are present in gel electrophoresis). In some embodiments, the complex-forming fragment of gH (i) forms part of the gH / UL116 dimer complex; (ii) comprises at least one epitope derived from SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, or SEQ ID NO: 5; and / or (iii) can elicit antibodies in vivo that exhibit cross-immunological reactivity with a CMV virion. Other suitable gH proteins may be gH variants that have varying degrees of identity with SEQ ID NO: 1, 2, 3, 4, or 5, such as at least 60%, 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% with the sequence cited in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, OR SEQ ID NO: 5. In
In some embodiments, variants of gH (i) form part of the gH / ULH6 dimer complex; (ii) comprise at least one epitope derived from SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, or SEQ ID NO: 5; and / or (iii) can elicit in vivo antibodies that exhibit cross-reactivity with a CMV virion.
UL116 CMV Proteins
In all HCMV sequenced genomes, the UL116 ORF is located in the long unique region (ÜL) between the UL115 and UL117 genes on the antisense coding strand (Figure IA). It has been shown that üL116 mRNA appears in the really late stage of infection with strain AD168. Multiple alignments of the UL116 primary translational sequences derived from the HCMV laboratory-adapted clinical strains are shown in Figure 1B (SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, and 17). UL116 from all these examples of CMV strains, as well as other strains, are suitable for use in the invention. UL116 from Merlin strain (SEQ ID NO: 6) has a length of 313 amino acids.
It is also encompassed in the invention, fragments forming a complex of 1MJL116. A complex forming fragment of UL116 can be any portion or portion of the UL116 protein that retains the ability to form a complex with another CMV protein. In some embodiments, a complex forming moiety of ULH6 forms part of the gH / UL116 dimer complex. an appropriate fragment of UL116 may have a length of at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least minus 125, at least 150, at least 175, at least 200, at least 225, at least 250, at least 275, or at least 300 amino acids.
An appropriate fragment of UL110 may also comprise, for example, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of the full length ULllo sequence. Alternatively, to the fragment of ULH6 described herein, there may be between 1 and 20 amino acid residues (for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid residues, or missing residues 1 to 20, residues 1 to 15, residues 1 to 10, residues 2 to 20, residues 1 to residues 2 to 15, residues 2 to 10, residues 5 to 20, residues 5 to 15, or residues 5 to 10) at the N-terminus and / or the C-terminus of UL116 full length.
A suitable ULH6 complex-forming moiety may also be obtained or determined by standard techniques known in the art, such as a co-immunoprecipitation technique, cross-linking, or fluorescent staining colocalization, etc. SDS-PAGE or Western blot analysis may also be used (for example, showing that both subunits are present in gel electrophoresis). In some embodiments, the complex forming fragment of UL116 (i) forms part of the gH / ULH6 dimer complex; (ii) comprises at least one epitope from SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; and / or (iii) can elicit antibodies in vivo that exhibit cross-immunological reactivity with a CMV virion. Other suitable UL116 proteins may be variants of ÜL116 which have varying degrees of identity with SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or As at least 60%, 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% with sequence cited in SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17. In some embodiments, variants of UL116 (i) form a dimer complex gH / UL116; (ii) comprise at least one epitope from SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17; and / or (iii) can elicit in vivo antibodies that exhibit cross-reactivity with a CMV virion.
GH / UL116 Protein Complex
Dimer complexes are disclosed herein including gH (or one of its complex forming fragments, or one of its variants) and 1OL116 (or one of its complex-forming fragments, or one of its variants). For simplicity, the complex is called gH / UL116.
A protein complex may also comprise, in addition to gH and ULH6 (or a complex-forming fragment thereof, or a variant thereof), one or more other CMV proteins as a part thereof. of the complex.
In some embodiments, to facilitate assembly of the complex, it may be desirable to express gH and UL116 as a fusion protein. That is, gH (or one of its fragments forming a complex, or one of its variants) and 1MJL116 (or one of its fragments forming a complex, or one of its variants) are fused into a single polypeptide chain. If one or more additional proteins are also present in the complex, the fusion protein may also comprise this or these additional proteins. The subunit can be directly fused, or it can be linked by a linker sequence. The linker sequence may be from 1 to 50 amino acids in length. The linker sequence can be cleavable.
GH (or one of its fragments forming a complex or one of its variants) and UL116 (or one of its fragments forming a complex, or one of its variants) of the invention can include the addition of an amino acid sequence which constitutes a marker, which may facilitate detection (eg, an epitope tag for detection by monoclonal antibodies) and / or purification (e.g., a polyhistidine marker for allow purification on a nickel chelating resin) proteins. Examples of markers for affinity purification include, for example, the His tag (hexahistidine (SEQ ID NO: 18), binds to the metal ion), the Malosekin Binding Protein (MBP) (binds to amylose), glutathione-S-transferase (GST) (binds to glutathione), the FLAG marker (Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys (SEQ ID NO: 19), binds anti-flag antibody), Strep markers (Ala-Trp-Arg-His-Pro-Gln-Phe-Gly-Gly (SEQ ID NO: 20), or Trp-Ser-His-Pro-Gln-Phe- Glu-Lys (SEQ ID NO: 21), bind to streptavidin or one of its derivatives), the HA tag, the MYC tag, or one of their combinations. The marker can be attached to either gH or UL116 or both. One or more markers may be used (for example, one marker for the gH and another for the UL116). 2. Combination of antigens
Immunogenic compositions comprising gH / UL116 complexes described herein are also provided herein. The immunogenic composition may comprise additional CMV protein or additional CMV protein complex.
For example, the additional CMV protein or the CMV supplemental protein complex may be selected from the group consisting of gB, gH, gL, gO, gM, gN; UL128, UL130, UL131, RL10, RL11, RL12, RL13, UL4, UL5, UL10, UL80.5, UL119, UL122, UL133, UL138, UL148A, UL1, UL7, UL9, UL16, UL18, UL20, UL40, UL41A, UL42, UL47, UL111A, UL124, UL132, UL136, UL141, one of their immunogenic fragments, one of their complex-forming fragments, and one of their combinations.
In some embodiments, the additional CMV protein or the CMV supplemental protein complex may be selected from the group consisting of gB, gH, gL; gO; gM, gN; UL128, UL130, UL131, one of their immunogenic fragments, one of their complex fragments, and one of their combinations. For example, the additional CMV protein may be gB or one of its immunogenic fragments.
In some embodiments, the CMV supplemental protein complex is selected from the group consisting of. of: the pentamer gH / gL / UL128 / UL130 / UL131 complex, the gH / gL complex, the gH / gL / gO trimer complex, or one of their combinations. Other suitable complexes include, for example, the gM / gN complex. Each subunit of these complexes may be full length, or one of its fragments forming a complex. Such complex-forming moieties can be determined by methods known in the art (e.g., by coimmunoprecipitation, crosslinking, colocalization, etc.). 3. Recombinant expression of qH / UL116
Nucleic acids encoding gH (or one of its complex-forming fragments, or one of its variants) and UL116 (or one of its fragments forming a complex, or one of its variants) as described herein. The nucleic acid can be used directly as a nucleic acid vaccine, or it can be used for the recombinant production of the gH / ULH6 protein complex. The nucleic acid (s) may be a single construct (e.g., a single vector encoding both gH and UL116), or they may be a combination of two or more constructs (e.g., a first vector coding for gH, and a second vector coding for UL116). The invention also provides a host cell comprising the nucleic acids described herein. When the host cell is cultured under appropriate conditions, the nucleic acid (s) can express the gH / UL116 protein complex. Preferably, the gH / UL116 protein complex is soluble. Preferably, the gH / λL116 protein complex can be secreted from the host cell.
The gH protein itself has a secretory signal. This signal can be used to express the gH / UL116 complex that can be secreted from the host cell. Alternatively or additionally, a suitable signal peptide can be used in one or more of the five subunits (for example, by making a fusion protein with a secretory signal). Signal sequences (and expression cassette) for the production of secretory proteins are known in the art. In general, the leader peptides are from 5 to 30 amino acids in length, and they are generally present at the N-terminus of a newly synthesized protein. The signal peptide core typically contains a long hydrophobic amino acid sequence that tends to form a single alpha helix. In addition, many signal peptides start with a short positively charged amino acid sequence, which may help enhance the correct topology of the polypeptide during translocation. At the end of the signal peptide, there is generally an amino acid sequence that is recognized and cleaved by a signal peptidase. A peptidase signal may cleave either during or after the completion of the translocation to produce a free signal peptide and a mature protein.
Suitable host cells include, for example, bacteria (e.g., E. coli, Bacillus subtilis, and Streptococcus spp.), Yeast cells (e.g., Saccharomyces cerevisiae, Candida albicans, Candida maltosa, Hansenula polymorph, Kluyveromyces fragilis , Kluyveromyces lactis, Pichia guillerimondii, Pichia pastoris, Schizosaccharomyces pombe and Yarrowia lipolytica), Tetrahymena cells (e.g., Tetrahymena thermophila), insect cells (e.g., Aedes aegypti, Autographa californica, Bombyx mori, Drosophila melanogaster, Spodoptera frugiperda, and Trichoplusia ni), avian cells (e.g., chicken, duck, and goose), mammalian cells (e.g., human cells, nonhuman primates, horse, cow, sheep, dog, cat and rodents (eg, hamster)), or combinations thereof.
Suitable insect cell expression systems, such as baculovirus systems, are known to those skilled in the art and described, for example, in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987). Materials and methods for baculovirus / insect cell expression systems are commercially available in kit form from, inter alia, Invitrogen, San Diego CA. For example, for expression in insect cells, a suitable baculovirus expression vector, such as pFastBac (Invitrogen), is used to produce recombinant baculovirus particles. The baculovirus particles are amplified and used to infect insect cells to express a recombinant protein. Suitable insect cells include, for example, Sf9 cells, cells. Sf21, Tn5 cells, Schneider S2 cells, and High Five cells (a clonal isolate derived from the parental cell line of Trichoplusia nor BTI-TN-5B1-4 (Invitrogen)).
Avian cell expression systems are also known to those skilled in the art and described, for example, in U.S. Patent Nos. 5,340,740; 5,656,479; 5,830,510; 6,114,168; and 6,500,668; European Patent No. EP 0787180B; European Patent Application No. EP 03291813.8; WO 03/043415; and WO 03/076601. Suitable avian cells include, for example, embryonic chicken stem cells (e.g., EBx® cells), chicken embryo fibroblasts, chicken embryo germ cells, duck cells (e.g. AGE1.CR and AGE1.CR.pIX (ProBioGen) cell lines which are described, for example, in Vaccine 27: 4975-4982 (2009) and WO 2005/042728), EB66 cells, and the like.
Preferably, the host cells are mammalian cells (e.g., human, nonhuman primates, horse, cow, sheep, dog, cat and rodent (eg, hamster)). Suitable mammalian cells include, for example, Chinese hamster ovary (CHO) cells, human embryonic kidney cells (HEK-293 cells, generally transformed with type 5 adenovirus sheared DNA), NIH-3T3 cells, Vero cells, HeLa cells, PERC.6 cells (ECACC deposit number 96022940), Hep G2 cells, MRC-5 (ATCC CCL-171), WI-38 (ATCC CCL-75 ), Rhesus fetal lung cells (ATCC CL-160), Madin-Darby bovine kidney cells ("MDBK"), Madin-Darby canine kidney cells ("MDCK") (eg, MDCK (NBL2) ATCC CCL34, or MDCK 33016, DSM ACC 2219), baby hamster kidney (BHK) cells, such as BHK21-F cells, HKCC, and the like.
In some embodiments, the host cell is a CHO cell: In certain embodiments, the polynucleotide encoding gH (or one of its fragments forming a complex, or one of its variants) and UL116 (or one of its complex-forming fragments, or one of its variants) described herein is stably integrated into the genomic DNA of the CHO cell.
Various CHO cell lines are also available from the European Collection of Cell Cultures (ECACC) or the American Type Culture Collection (ATCC), such as CHO cell lines hCBE11 (ATCC® PTA-3357-), E77. 4 (ATCC® PTA-3765 ™), hLT-B: R-hG1 CHO # 14 (ATCC® CRL-11965 ™), MOR-CHO-MORAb-003-RCB (ATCC® PTA-7552 ™), AQ.C2 clone 11B (ATCC® PTA-3274 ™), AQ.C2 clone 11B (ATCC® PTA-3274 ™), hsAQC2 in CHO-DG44 (ATCC® PTA-3356 ™), xrs5 (ATCC® CRL-2348 ™), CHO -Kl (ATCC® CCL-61 ™), Lecl (originally named Pro-5WgaRI3C) (ATCC® CRL-1735 ™), Pro-5 (ATCC® CRL-1781 ™), ACY1-E (ATCC® 65421 ™), ACY1-E (ATCC® 65420 ™), pgsE-606 (ATCC® CRL-2246 ™), CHO-CD36 (ATCC® CRL-2092 ™), pgsC-605 (ATCC® CRL-2245 ™), MC2 ATCC® CRL-2143 ™, CHO-ICAM-1 (ATCC® CRL-2093 ™), and pgsB-618 (ATCC® CRL-2241 ™) Any of these CHO cell lines can be used. Examples of CHO cell lines available from of the European Collection of Cell Cultures (ECACC) are listed in Table 1.
Table 1
Other commercially available CHO cell lines include, for example, CHO-S Freestyle ™ cells and Life Technologies' Flp-ln ™ -CHO cell line.
Methods for the expression of recombinant proteins in CHO cells have generally been disclosed. See, for example, U.S. Patent Nos. 4,816,567 and 5,981,214.
In some embodiments, the recombinant nucleic acids are optimized for codons for expression in a selected prokaryotic or eukaryotic host cell.
To facilitate replication and expression, the nucleic acids may be incorporated into a vector, such as a prokaryotic or eukaryotic expression vector. Examples of vectors include plasmids that are capable of autonomous replication or replication in a host cell. Typical expression vectors contain suitable promoters, enhancers, and terminators that are useful for regulating the expression of the coding sequence (s) in the expression construct.
The vectors may also include selectable markers to provide a phenotypic trait for the selection of transformed host cells (such as conferring resistance to antibiotics such as ampicillin or neomycin).
Examples of procedures sufficient to guide a person of average skill in the field through the production of one or more recombinant nucleic acids for the expression of the gH / UL116 complex can be found in Sambrook et al., Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, 1989; Sambrook et al., Molecular Cloning: A Laboratory Manual, 3d ed., Cold Spring Harbor Press, 2001; Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates, 1992 (and Supplements up to 2003); and Ausubel et al., Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, 4th ed., Wiley & Sounds, 1999.
There is also proposed here a cell culture comprising the host cell described herein. The cell culture can be large scale, for example, at least about 10 1, at least about 20 1, at least about 30 1, at least about 40 1, at least about 50 1, at least about 60 1, at least about 70 1, at least about 80 1, at least about 90 1, at least about 100 1, at least about 150 1, at least about 200 1, at least about 250 1, at least about 300 1, at least about 400 1, at least about 500 1, at least about 600 1, at least about 700 1, at least about 800 1, at least about 900 1, at least about 1000 1, at least about 2000 1, at least about 3000 1, at least about 4000 1, at least about 5000 1, at least about 6000 1, at least about 10,000 1, at least about 15 000 1, at least about 20 000 1, at least about 25 000 1, at least about 30 000 1, at least about 35,000 1, at least about 40,000 1 / at least about 45,000 1, at least about 50,000 1 / at least about 55,000 1, at least about 60,000 1, at least about 65,000 1, at least about 70,000 1, at least about 75,000 1, at least about 80,000 1, at least about 85,000 1, at least about 90,000 1, at least about 95,000 1, at least about 100 000 1, etc.
In some embodiments, the yield of the gH / UL116 complex from the cell culture is at least about 0.01 g / l, at least about 0.02 g / l, at least about 0 , 03 g / l, at least about 0.05 g / l, at least about 0.06 g / l, at least about 0.07 g / l, at least about 0.08 g / l g / 1, at least about 0.09 g / l, at least about 0.1 g / l, at least about 0.15 g / l, at least about 0.20 g / l 1, at least about 0.25 g / l, at least about 0.3 g / l, at least about 0.35 g / l, at least about 0.4 g / l, at least about 0.45 g / l, at least about 0.5 g / l, at least about 0.55 g / l, at least about 0.6 g / l, at least about about 0.65 g / l, at least about 0.7 g / l, at least about 0.75 g / l, at least about 0.8 g / l, at least about about 0.85 g / l, at least about 0.9 g / l, at least about 0.95 g / l, or at least about 1.0 g / l.
There is also provided a method for producing a recombinant human cytomegalovirus (CMV) protein dimer complex, comprising the CMV gH protein or a complex fragment thereof, and CMV üL116 or one of its complex-forming fragments, comprising: (i) culturing the host cell described herein under suitable conditions, thereby expressing said gH / UL116 complex; and (ii) harvesting said gH / UL116 complex from the culture.
In some embodiments, the gH / UL116 complex described herein is purified. The gH / UL116 complex can be purified using any suitable methods, such as HPLC, various types of chromatography (such as hydrophobic interactions, ion exchange, affinity, chelation, and exclusion according to size), electrophoresis, density gradient centrifugation, solvent extraction, or the like. As appropriate, the gH / UL116 complex can be further purified, as required, to substantially eliminate all proteins that are also secreted into the medium or that result from lysis of the host cells, thereby providing a product that is at least substantially free of host debris, for example, proteins, lipids and polysaccharides. See, for example, those presented in Sandana (1997) Bioseparation of Proteins, Academic Press, Inc.; and Bollag et al. (1996) Protein Methods, 2nd Edition Wiley-Liss, NY; Walker (1996) The Protein Protocols Handbook Humana Press, NJ, Harris and Angal (1990) Protein Purification Applications: A Practical Approach IRL Press at Oxford, Oxford, United Kingdom; Scopes (1993) Protein Purification: Principles and Practice 3rd Edition Springer Verlag, NY; Janson and Ryden (1998) Protein Purification:
Principles, High Resolution Methods and Applications, Second Edition Wiley-VCH, NY; and Walker (1998) Protein Protocol on CD-ROM Humana Press, NJ. If desired, the gH / UL116 complex may comprise a "marker" that facilitates purification, as described above. 4. Pharmaceutical Compositions The invention provides pharmaceutical compositions and methods of treatment using the cytomegalovirus (CMV) gH / UL116 complex described herein, or a nucleic acid encoding such a gH / UL116 complex described herein. For example, the proteins may be administered directly as components of an immunogenic composition, or nucleic acids that encode the gH / UL116 complex may be administered to produce the CMV immunogenic protein or fragment in vivo. Some preferred embodiments, such as protein formulations, recombinant nucleic acids (e.g., self-replicating RNA), and alphavirus VRPs that contain sequences encoding gH (or one of its fragments forming a complex, or a variant thereof) and UL116 (or a complex-forming fragment thereof, or a variant thereof) are further described herein.
Protein compositions
In one aspect, the invention provides an immunogenic composition comprising the gH / UL116 complex described herein. The composition may comprise one or more additional CMV antigens as described herein.
The immunogenic composition may comprise an adjuvant. Examples of adjuvants for enhancing the effectiveness of the composition include: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc .; ; (2) oil-in-water emulsion formulations (with or without other specific adjuvants such as muramyl-peptides (see below) or bacterial cell wall components), such as (a) MF59 ( PCT Publication No. WO 90/14837), containing 5% squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of N-acetyl-muramyl-L-alanyl-D- isoglutaminyl-L-alanine-2- (1'-2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy) ethylamine (MTP-PE), although not required) formulated in submicron particles using a microfluidizer such as microfluidizer Model 110Y (Microfluidics, Newton, Mass.), (B) SAF, containing 10% squalane, 0.4% Tween 80, 5% pluronic L121 block polymer, and thr-MDP (see below) either microfluidized in a submicron emulsion or vortexed to produce a larger particle size emulsion, and (c) the Ribi ™ Adjuvant System (RAS), (R) ibi Immunochem, Hamilton, Mont.) containing 2% squalene, 0.2% Tween 80, and one or more bacterial cell wall components of the group consisting of monophosphoryl lipid A (MPL), trehalose dimycolate (CT) ), and the cell wall skeleton (CWS), preferably MPL + CWS (Detox ™); (3) saponin adjuvants, such as Stimulon ™ (Cambridge Bioscience, Worcester, Mass.) May be used or particles produced therefrom such as ISCOMs (immunostimulatory complexes); (4) Freund's Complete Adjuvant (CFA) and Incomplete Freund's Adjuvant (IFA); (5) cytokines, such as interleukins (IL-1, IL-2, etc.), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), etc. ; and (6) other substances that act as adjuvants to enhance the effectiveness of the composition.
Muramyl peptides include, but are not limited to, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor -MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2- (1'-2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy) ethylamine (MTP-PE), etc.
In some embodiments, the adjuvant is an oil-in-water emulsion, such as MF59. In some embodiments, the adjuvant is a TLR7 agonist, such as imidazoquinoline or Imiquimod. In some embodiments, the adjuvant is an aluminum salt, such as aluminum hydroxide, aluminum phosphate, aluminum sulfate.
The adjuvants described herein can be used individually or in any combination, such as the alum / agonist combination of TLR7.
Nucleic acid vaccines and delivery systems
The recombinant nucleic acid molecules that encode the CMV gH / ULH6 complex described herein can be administered to induce the production of the encoded gH and UL116 proteins, and an immune response therefor. The recombinant nucleic acid may be DNA (e.g., plasmid or viral DNA) or RNA, preferably self-replicating RNA, and may be monocistronic or polycistronic. Any suitable DNA or RNA can be used as a nucleic acid vector that carries the open reading frames that encode the gH / UL116 complexes described herein. Suitable nucleic acid vectors have the ability to carry and direct the expression of the individual subunits of the gH / UL116 complexes, as well as any other CMV antigens, if present. DNA obtained from DNA viruses such as vaccinia virus vectors (e.g., NYVAC, see US Patent 5,494,807), and poxvirus vectors (e.g., ALVAC canarypox vector, Sanofi Pasteur ), and RNA obtained from appropriate RNA viruses such as alphaviruses. If desired, the recombinant nucleic acid molecule may be modified, for example, to contain nucleobases and / or modified linkages as further described herein.
The self-replicating RNA molecules of the invention are based on genomic RNA virus RNA, but lack the genes encoding one or more structural proteins. Self-replicating RNA molecules can be translated to produce nonstructural proteins of RNA virus and CMV gH and UL116 proteins (or fragments or variants) encoded by self-replicating RNA. The self-replicating RNA generally contains at least one or more genes selected from the group consisting of viral replicases, viral proteases, viral helicases, and other non-structural viral proteins, and it also includes active replication sequences. in cis at the 5 'and 3' ends, and heterologous sequences which encode one or more CMV antigens (for example, gH (or one of its fragments or variants), UL116 (or one of its fragments or variants), and / or any desired additional CMV protein antigens). A subgenomic promoter that directs the expression of the heterologous sequence (s) may be incorporated into the self-replicating RNA. If desired, a heterologous sequence may be fused in frame to other coding regions in the self-replicating RNA and / or may be under the control of an internal ribosome entry site (IRES ) and / or a sequence of 2A, or one of their combinations. The nucleotide sequences encoding gH (or its fragments or variants), UL116 (or its fragments or variants), and any additional desired CMV protein antigens, if
present, can be administered by a single RNA vector (e.g., bicistronic RNA or RNA
polycistronic), or they can be administered by separate RNA vectors. If administered as bicistronic RNA or polycistronic RNA, IRES and / or 2A sequences can be used to produce individual proteins.
The self-replicating RNA molecules of the invention may be designed such that the self-replicating RNA molecule can not induce the production of infectious viral particles. This can be achieved, for example, by omitting one or more viral genes encoding structural proteins that are necessary for the production of virus particles in self-replicating RNA. For example, when the self-replicating RNA molecule is based on an alphavirus, such as Sindbis virus (SIN), Semliki forest virus and Venezuelan equine encephalitis virus (VEE), one or more genes encoding viral structural proteins, such as capsid glycoproteins and / or envelope, may be omitted. As used herein, the term "alphavirus" has its traditional meaning in the art and includes various species such as Venezuelan equine encephalitis virus (VEE, for example, the Trinidad sage, TC83CR, etc.), Semliki Forest Virus (SFV), Sindbis virus, Ross River virus, Western equine encephalitis virus, Eastern equine encephalitis virus, Chikungunya virus, SA AR86 virus, Everglades virus, Mucambo virus, Barmah forest virus, Middelburg virus, Pixuna virus, 0'nyong-nyong virus, Getah virus, Sagiyama virus, Bebaru virus, Mayaro virus, virus Üna, Aura virus, Whataroa virus, Banbanki virus, Kyzylagach virus, Highlands J virus, Fort Morgan virus, Ndumu virus, and Buggy Creek virus.
A self-replicating RNA molecule can, when administered to a vertebrate cell even without any protein, lead to the production of multiple daughter RNA molecules by transcription from itself (or from an antisense copy of itself). The self-replicating RNA can be translated directly after administration to a cell, and this translation provides an RNA-dependent RNA polymerase which then produces transcripts from the administered RNA. Thus, the administered RNA leads to the production of multiple daughter RNA molecules. These transcripts are antisense to the administered RNA and can be translated themselves to provide in situ expression of the CMV encoded protein, or they can be transcribed to provide other transcripts with the same sense as the Administered RNA, which are translated to provide in situ expression of the CMV encoded protein (s).
A preferred self-replicating RNA molecule thus encodes (i) an RNA-dependent RNA polymerase that can transcribe RNA from the self-replicating RNA molecule and (ii) gH (or its fragments or variants), UL11 (or fragments or variants thereof) and any additional desired CMV protein antigens if present. The polymerase may be an alphavirus replicase, for example, comprising nsP4 nsP1 alphavirus nonstructural proteins.
The self-replicating RNA molecules of the invention may contain one or more modified nucleotides and therefore have improved stability and are resistant to in vivo degradation and clearance, and other advantages. There are more than 96 naturally occurring nucleoside modifications found on mammalian RNA. See, for example, Limbach et al., Nucleic Acids Research, 22 (12): 2183-2196 (1994). The preparation of nucleotides and nucleotides and modified nucleosides is well known in the art, for example, according to US Pat. Nos. 4,373,071, 4,458,066, 4,500,707, 4,668,777, 4,973,679, 5,047,524, 5,132,418, 5,153,319, 5,262,530, 5,700,642, all of which are hereby incorporated by reference in their entirety, and many modified nucleosides and modified nucleotides are commercially available. If desired, the self-replicating RNA molecule may contain phosphoramidate, phosphorothioate, and / or methylphosphonate linkages. The self-replicating RNA described herein is suitable for administration in various modalities, such as administration of naked RNA or in combination with lipids, polymers or other compounds that facilitate cell entry. The self-replicating RNA molecules can be introduced into target cells or into subjects using any suitable technique, for example by direct injection, microinjection, electroporation, lipofection,
biolistics, and the like. The self-replicating RNA molecule can also be introduced into cells by means of receptor-mediated endocytosis. See, for example, US Patent
No. 6,090,619; Wu and Wu, J. Biol. Chem., 263: 14621 (1988); and Curiel et al., Proc. Natl. Acad. Sci. USA, 88: 8850 (1991). For example, US Patent No. 6,083,741 discloses the introduction of an exogenous nucleic acid into mammalian cells by combining the nucleic acid with a polycationic radical (for example, poly-L-lysine having 3 to 100 amino acids lysine), which itself is coupled to an integrin receptor binding moiety (e.g., a cyclic peptide having the Arg-Gly-Asp sequence).
The self-replicating RNA molecules can be administered into cells via amphiphiles. See, for example, US Patent No. 6,071,890. Generally, a nucleic acid molecule can form a complex with the cationic amphiphile. Mammalian cells brought into contact with the complex can easily absorb it. The self-replicating RNA can be administered in the form of naked RNA (for example, simply in the form of an aqueous solution of RNA) but, to enhance entry into cells and also intercellular effects Subsequently, the self-replicating RNA is preferably administered in combination with a delivery system, such as a particle-based or emulsion-based delivery system. A large number of delivery systems are well known in the art. Three particularly useful delivery systems are (i) liposomes, (ii) non-toxic and biodegradable polymeric microparticles, and (iii) submicron cationic oil emulsions in water. Non-virion-based administration, in which the RNA is not packaged in a virion particle, generally has better safety profiles.
If desired, the self-replicating RNA molecules of the invention may be designed to induce the production of infectious viral particles that are attenuated or virulent, or to produce virus particles that are capable of a single subsequent infection cycle. The invention also provides an immunogenic composition comprising the nucleic acid (e.g., self-replicating RNA) described herein. The immunogenic composition may comprise an adjuvant as described above. Preferred adjuvants include, for example, an aluminum salt or an oil-in-water emulsion (such as MF59). VRP of alphavirus
In some embodiments, the CMV gH / UL116 complexes described herein are administered using alphavirus replicon (VRP) particles. An "alphavirus replicon particle" (VRP) or "replicon particle" is an alphavirus replicon packaged with alphavirus structural proteins. An "alphavirus replicon" (or "replicon") is an RNA molecule that can direct its own amplification in vivo in a target cell. The replicon encodes the polymerase (s) that catalyze the amplification of RNA (nsP1, nsP2, nsP3, nsP4) and contains cis RNA sequences necessary for replication that are recognized and used by the polymerase (s). encoded. An alphavirus replicon generally contains the following elements in the order: 5 'viral sequences required in cis for replication, sequences that encode biologically active alphavirus non-structural proteins (nsP1, nsP2, nsP3, nsP4 ), 3 'viral sequences required in cis for replication, and a polyadenylation sequence. An alphavirus replicon may also contain one or more subgenomic promoters of the "junction region" directing the expression of the heterologous nucleotide sequences, which may, in some embodiments, be modified to increase or decrease viral transcription subgenomic fragment and the heterologous sequence (s) to be expressed. Other control elements may be used, such as IRES or 2A sequences.
Pharmaceutical formulations
Each of the immunogenic compositions described herein may be used alone or in combination with one or more other antigens, the latter from either the same viral pathogen or from another source or other pathogenic source. These pharmaceutical formulations can be either prophylactic (to prevent an infection) or therapeutic (to treat the disease after an infection).
Such pharmaceutical formulations include an immunogenic composition, usually in combination with "pharmaceutically acceptable carriers," which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Suitable carriers are generally large, slowly metabolized macromolecules such as proteins, polysaccharides, lactic acid polyacids, polyhydric acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive viral particles. Such media are well known to a person of average skill in the field. In addition, these supports can function as adjuvants. In addition, the antigen may be conjugated with a bacterial toxoid, such as toxoid of diphtheria, tetanus, cholera, H. pylori, etc.
The pharmaceutical formulations may include an adjuvant as described above.
The pharmaceutical formulations (e.g., the immunogenic composition, the pharmaceutically acceptable carrier, and the adjuvant) will generally contain diluents, such as water, saline, glycerol, ethanol, etc. In addition, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles. Generally, the pharmaceutical formulations are prepared in the form of injectables, in the form of either solutions or liquid suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. The preparation may also be emulsified or encapsulated in liposomes to enhance the adjuvant effect, as described above in the section of pharmaceutically acceptable carriers.
The pharmaceutical formulations comprise an immunologically effective amount of the antigenic polypeptides, as well as all the other components mentioned above, as needed. By "immunologically effective amount" is meant that administration of that amount to an individual, either in a single dose or as part of a series, is effective for the treatment or prevention of a disease, a infection or pathology. This quantity varies according to the state of health and the physical condition of the individual to be treated, the taxonomic group of the individual to be treated (for example, non-human primate, primate, etc.), the capacity of the immune system of the individual to be treated. individual to synthesize antibodies, the degree of protection desired, the vaccine formulation, the physician's estimate of the medical situation, and other relevant factors. The quantity is expected to be within a relatively wide range that can be determined through routine testing.
The pharmaceutical formulations are conveniently administered parenterally, for example by injection, either subcutaneously or intramuscularly. Additional appropriate formulations for other modes of administration include oral and pulmonary formulations, suppositories, and transdermal applications. Oral formulations may be preferred for certain viral proteins.
The dosage treatment may be a single dose schedule or a multiple dose schedule. The immunogenic composition may be administered together with other immunoregulatory agents. 5. Treatment processes
In another aspect, the invention provides a method of inducing an immune response against cytomegalovirus (CMV), comprising administering to a subject in need of an immunologically effective amount of the immunogenic composition described herein, which comprises proteins, DNA molecules, RNA molecules (e.g., self-replicating RNA molecules), or VRPs as described herein.
In some embodiments, the immune response comprises the production of neutralizing antibodies against CMV. In some embodiments, the neutralizing antibodies are complement-independent.
The immune response can include a humoral immune response, a cell-mediated immune response, or both. In some embodiments, an immune response is induced against each administered CMV protein. A cell-mediated immune response may include a helper T cell response (Th), a CD8 + cytotoxic T lymphocyte (CTL) response, or both. In some embodiments, the immune response comprises a humoral immune response, and the antibodies are neutralizing antibodies. Neutralizing antibodies block viral cell infection. CMV infects endothelial cells, epithelial cells and also fibroblast cells. In some embodiments, the immune response reduces or prevents infection of one or more of these cell types. Neutralizing antibody responses may be complement-dependent or complement-independent. In some embodiments, the neutralizing antibody response is complement-independent. In some embodiments, the neutralizing antibody response produces cross-neutralization; i.e., an antibody produced against an administered composition neutralizes a CMV virus of a strain other than the strain used in the composition.
A useful measure of the potency of an antibody in the art is the "50% neutralization titer". To determine the 50% neutralization titer, serum from immunized animals is diluted to estimate how a diluted serum may still retain the ability to block entry of 50% of the viruses into the cells. For example, a titre of 700 means that the serum has retained the ability to neutralize 50% of the viruses after being diluted 700 times. Thus, higher titers indicate stronger neutralizing antibody responses. In some embodiments, this title is in a range having a lower limit of about 200, about 400, about 600, about 800, about 1000, about 1500, about 2000, about 2500, about 3000, about 3500, about 4000, about 4500, about 5000, about 5500, about 6000, about 6500, or about The range of 50% neutralization titers may have an upper limit of about 400, about 600, about 800, about 1000, about 1500, about 2000, about 2500, about 3000, about 3500, about 4000, about 4500, about 5000, about 5500, about 6000, about 6500, about 7000, about 8000, about 9000, about 10,000, about 11000, about 12000, about 13000, about 14000, about 15000, about 16000, about 17000, about 18,000, about 19,000, about 20,000, about 21,000, about 22,000, about 23,000, about 24,000, about No. 25,000, about 26,000, about 27,000, about 28,000, about 29,000, or about 30,000. For example, the 50% neutralization titer can be from about 3,000 to about 25,000. Approximately »means plus or minus 10% of the quoted value. The neutralization title can be measured, for example, as described in the specific examples below.
An immune response can be stimulated by administering proteins, DNA molecules, RNA molecules (e.g., self-replicating RNA molecules), or VRPs to an individual, typically a mammal , including a human being. In some embodiments, the induced immune response is a protective immune response, i.e. the response reduces the risk or severity of CMV infection. Stimulation of a protective immune response is particularly desirable in certain populations particularly at risk for infection and CMV disease. For example, at-risk populations include solid organ transplant (TOS) recipients, bone marrow recipients, and hematopoietic stem cell transplant (HCT) recipients. VRPs may be administered to a transplant donor prior to transplantation, or to a transplant recipient before and / or after transplantation. Because vertical transmission from mother to child is a common source of infant infection, administering VRP to a woman who is pregnant or who may become pregnant is particularly helpful.
Any appropriate route of administration may be used. For example, a composition can be administered intramuscularly, intraperitoneally, subcutaneously, or transdermally. Some embodiments will be administered via an intramucosal route, such as the intraoral, intranasal, intravaginal and intrarectal routes. The compositions may be administered according to any appropriate schedule.
There is also provided herein a method of inhibiting the entry of cytomegalovirus (CMV) into a cell, comprising contacting the cell with the immunogenic composition described herein.
This invention is further illustrated by the following examples which should not be construed as limiting.
Examples • Example 1 - Materials and Processes
Cell Lines, Plasmids and TR Virus is a clinical strain of human CMV that has been derived from a vitreous humor sample of the eye of a patient with HIV disease and has been cloned into a chromosome artificial bacterial (BAC) after limited subculture in fibroblasts.
The TR strain of HCMV and the UL116 Flag-TR recombinant virus were propagated in human foreskin fibroblasts HFF-1 (ATCC: SCRC-1041) grown in minimal essential medium (Invitrogen) supplemented with 10% fetal calf serum. , glutamine (100 mg / liter), and gentamycin (350 mg / liter). The virions were isolated by glycerol-tartrate gradient centrifugation as described. HEK293T cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal calf serum, glutamine and gentamycin. Lipofectamine 2000 (Invitrogen) was used to transfect HEK293T cells. The human HCMV genes optimized for the codons of UL116, gH (UL75), gL (UL115) and gB (UL55) based on the sequence of the TR strain were synthesized by Geneart and cloned into the plasmid pcDNA3.1 (-) / myc-His C (Invitrogen) in frame with C-terminal sequences of myc markers and six histidines. Simple point mutations were made with the Quick Change Mutagenesis Kit (Stratagene) producing marker and marker-free versions of these genes.
Construction and production of the UL116-Flaq TR virus
The insertion of the Flag marker at the C-terminus of the UL11δ ORF on TR BAC was achieved using the two-step Red-mediated recombination method. The primer pair used to amplify the kanamycin insertion cassette consists of the direct primer 5'-TTC GGC GCC AAC TGG CTC CTT ACC GTC ACA CTC TCA TCG TGC CGC AGA CTG ATT ACA AGG ATG AC G AC G ATA AG-3 '(SEQ ID NO: 22) and the reverse primer 5'-TAT CAC CGG TCC AGG TGA GAA AGA GAA GCC GCA
ATC CGG GCG GCG GCA CA TCA CTT ATC GTC GTC ATC CTT GTA
ATC AGT CTG CGG CAC GAT GAG CAA CCA TAA TAA ACCA ATT CTG ATT TAG-3 '(SEQ ID NO: 23) where the underlined base pairs encode Flag peptide.
Reconstitution of infectious virus
To reconstitute the virus, 2 μg of BAC-HCMV DNA and 1 μg of the expression plasmid pp71 were transfected into MRC-5 cells by electroporation. The culture medium was changed 24 hours later and the cells were divided and cultured until the virus plaques appeared.
The virus stock was prepared by harvesting the cell-free culture supernatant when the entire monolayer of the cells was lysed or the cells were divided and cultured until the virus plaques appeared.
Purification of HCMV-TR virions
Human CMV particles in cell supernatants were separated into fractions of virions, dense bodies, and non-infectious enveloped particles (NIEP) by positive density gradient / negative viscosity gradient centrifugation as previously described. The concentrations of the particles in the preparations were estimated by counting negatively stained samples by electron microscopy against a standard concentration of latex beads. To separate the virion envelope proteins from the capsid and the integument proteins, 108 particles were mixed in a ratio of 1/1 with envelope stripping buffer (2% Nonidet-P40 in PBS ) and incubated for 15 minutes at 4 ° C. The particles were agglomerated (12,000 g for 5 minutes at 4 ° C), and the fraction of the soluble shell was harvested. The insoluble capsid / integument material was washed twice with the envelope stripping buffer and once in PBS before being solubilized in the SDS-PAGE sample buffer.
Flow cytometry
For the detection of UL116 exposed on the membrane, HEK293T cells transiently transfected with UL116, gH, gB and an empty vector encoding vectors were detached with trypsin 48 h after transfection, incubated for 30 min at RT with Live & Dead Agua reagent (Invitrogen), diluted 1/400 in PBS and then incubated with different dilutions of anti-UL116 mouse polyclonal sera for 60 min on ice. Secondary antibody committee, a goat anti-mouse antibody conjugated to Alexa Fluor 647 reagent was used for 30 min on ice at a dilution of 1/200. A total of 104 cells were analyzed for each curve using a FACSCanto II instrument (Becton Dickinson, Heidelberg, Germany). The experiments were performed in triplicate for statistical consistency; averages and standard deviations were analyzed and plotted using the Graphpad Prism software.
Glycosidase treatment
Samples were treated with PNGase F (P0705S, New England BioLabs) or ENDO H (P0703S, New England BioLabs) according to the manufacturer's instructions. Briefly, the samples were denatured in glycoprotein denaturant buffer at 100 ° C for 10 minutes and cooled to 0 ° C for 5 minutes. The samples were then digested overnight at 37 ° C with PNGase F or ENDO H before being analyzed by Western immunoblot.
Electron microscopy with Immunogold labeling
The purified virions were air-dried on the surface of Formvar coated ME grids. The grids were treated with anti-Flag mouse antibody (Sigma-F3165) for four hours at room temperature, washed three times with PBS, and incubated with goat anti-mouse antibody conjugated to gold particles. of 5 nm for one hour at room temperature. After another washing in PBS, the grids were stained negatively with phosphotungstic acid and subjected to ME.
Purification of the gH / UL116 and gH / gLC144A-3G16 Complexes The gH / UL116 complex was purified from the supernatants of HEK-293EBNA cells transfected in duplicate with plasmids encoding C-terminally labeled ULll6 by a Strep double marker and unmarked gH. Following purification by affinity chromatography for the Strep marker, the complex was incubated with the 3G16-Fab antibody. The gH / UL116 / 3G16-Fab trimer complex was further purified by exclusion chromatography (SEC). The gH / gL complex harboring the gL-C144A mutation was purified from the supernatants of the HEK-293EBNA cells transfected in duplicate with plasmids encoding a His-tagged gH at the C-terminus and unlabeled gL as he has been previously described (Ciferri et al., submitted). The purified gH / gL-C144A complex was mixed in a ratio of 1: 1.2 with the 3G16 antibody and the ternary complex was isolated by SEC.
Negative staining electron microscopy and simple particle analysis
Five microliters of the purified gH / UL116-3G16 and gH / gL (C144A) -3G16 samples (approximately 100 ng) were placed on a freshly glow discharge Holey Carbon grid covered with a continuous carbon thin film. The grid was stained sequentially with drops of 75 μl of a freshly prepared solution of 2% (w / v) uranyl formate. The samples were imaged using a Tecnai Spirit T12 transmission electron microscope operating at 120 keV at a nominal magnification of 49,000x (1.57 A / pixel at the detector) using a defocus range of -0, 8 to -1.2 μιη. The images were recorded under low dose conditions on a Gatan CCD camera 4096 x 4096 pixels.
The particles were picked manually using an e2boxer instrument (EMAN2,) and extracted using a box size of 224 pixels. Both sets of data were bandwidth-filtered with a high-pass threshold of 200-Â and a low-pass threshold of 20-Â. For the 2D ranking, it was used an iterative multiple-variable statistical analysis (MSA) and a multiple reference alignment (MRA) in Imagic. The 2D classes from gH / gL and gH / U1116 were aligned and compared by cross-correlation using the SPIDER ΆΡ SH 'command. SDS-PADE and immunoblot
Proteins were separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) on 10 or 15% polyacrylamide gels under standard conditions. The proteins were transferred to nitrocellulose membranes, and the membranes were blocked with PBS containing 0.1% Tween 20 and 5% milk powder. Antibodies and sera were diluted in PBS containing 0.1%
Tween 20. For the detection of the binding of the primary antibody, a horseradish peroxidase-conjugated anti-rabbit immunoglobulin G antibody (Perkin Eimer) and the amplified chemiluminescence detection system (Pierce) were used according to the manufacturer's instructions. Expression of late-phase proteins was inhibited by the use of phosphonacetic acid (PAA, Sigma-Aldrich). A total of 250 μg / ml PAA was added to the medium at the time of infection and maintained throughout the infection.
immunofluorescence
The cells were plated on glass slides and infected with HCMV. At 7 days pi, the cells were fixed in 4% paraformaldehyde, permeabilized with 0.5% NP-40, preblocked with HCMV seronegative human IgG and incubated with the primary antibody for one hour. at room temperature. After washing, the secondary antibodies were incubated for one hour at 37 ° C, the cells were washed again, and mounted with the Pro-long Safe Stain DAPI mounting medium (Invitrogen). The primary antibodies were anti-Flag rabbit antibodies (F7425; Sigma), anti-Flag mouse antibodies (F1804; Sigma), anti-human sheep TGN46 antibodies (AHP500; Serotec), mouse anti-mouse mAbs. pp28 (6502; Abcam), anti-gH mouse antibodies (2470-5437; Adb Serotech) and anti-gL rabbit antibodies (1260A-OHSU). Secondary antibodies were goat anti-mouse and anti-rabbit antibodies conjugated to Alexa Fluor 488, 568, and 647 (Invitrogen). Intracellular locations of the antibody-labeled proteins were examined under laser illumination in a Zeiss LMS 700 confocal microscope, and the images were captured using the ZEN 2009 software.
immunoprecipitation
HFF-1 cells were infected with stocks of wtTR and UL116-Flag-TR. Protein expression was allowed to proceed for 72 hours before the cells were washed in IX PBS (0.137 M NaCl, 0.0027 M KCl, 0.1 M Na 2 HPO 4, 0.002 M KH 2 PO 4 pH 7.4) and lysed (0.025 M Tris, 0.15 M NaCl, 0.001 M EDTA, 1% NP-40, 5% glycerol, pH 7.4) in the presence of inhibitors of proteases. 500 μg of the proteins were incubated overnight at 4 ° C with 5 μg of the MSL-109 monoclonal antibody (courtesy of A. Feire). The complexes were immunoprecipitated using Protein G Dynalbeads (Invitrogen) according to the manufacturer's protocol. The complexes were washed four times in lysis buffer. The samples were boiled for 5 minutes before the immunoblot. The same procedure as that described above was adopted for the immunoprecipitation experiments carried out on transient expression HEK293T cells. The complexes were captured in parallel experiments with magnetic beads of anti-His Ab (Genescript) and magnetic beads of covalently bonded anti-c-myc (Pierce) Ac to avoid background signals in the materials. eluted resultants.
Example 2 - Results
Primary structure of the UL116 gene product
In all sequenced human CMV genomes, the u1116 gene is located in the long unique region (UL) between the u1115 (gL) and u117 genes on the antisense coding strand (FIG. 1A). It has been previously shown that u1116 mRNA appears in the really late stage of AD169 infection as part of the UL119-UL115 transcription unit but the gene product has never been analyzed before. Multiple alignments of the primary translation sequences of u1116 derived from clinical strand sequences and adapted to the HCMV laboratory are shown in Figure 1B. The degree of conservation among the most representative strains of human CMV is 98% of identity resulting in a very strong conservation of this protein among the cytomegalovirus population. ORF u1116 is predicted to encode a 313 amino acid (UL116) glycoprotein comprising a signal peptide (position of amino acids 1 to 24, SP in FIG. 1B), a threonine-rich domain (amino acids 27 to 157) and 14 predicted N-glycosidic glycosylation consensus sites. The resulting polypeptide backbone is predicted to have a molecular weight of 34.2 kDa (supplemented with 3 kDa on the FLAG-tagged viral protein and approximately 6 kDa for myc-His-UL116). In addition, UL116 lacks the membrane anchoring sequences and is expected to be a secreted protein.
Kinetics of ULllô expression during the
HCMC replication
To study the kinetics of UL116 expression during human CMV-producing infection, we produced a recombinant human CMV (TR-derived BAC) with a Flag marker fused to the C-terminus of the ULllô (see materials and processes). The reconstituted TR UL116-Flag virus was used to infect HFF-1 cells and the kinetics of ULllo expression was followed by immunoblotting using an anti-Flag antibody. Infected cells were harvested at different time points, ranging from 4 to 120 hours p.i., and the extracts were prepared. As controls, the expressions of the major immediate protein IE1 pp72 (UL123) and the late phosphoprotein pp28 (UL99) were revealed in the same samples. The results are shown in Figure 2A. At time points after infection, when UL110 signals were not observed, the immediate early protein IE1 / pp72 was present. As described in the literature, the IE1 protein began to be revealed at 4 hours p.i. and was present during the entire HCMV replication cycle. Detection of ULllo expression occurred in parallel with pp28, a late phase marker. Consistent with the observed kinetic profile, metabolic blockade with PAA resulted in the disappearance of the ULllδ bands (Figure 2B). In the extracts of infected human fibroblasts, ULll6 migrated as two species, a faster migrating protein of 76 kDa and a slower migrating protein of approximately 125 kDa (Fig. 2A). The latter appeared to be the mature product of the 76 kDa species, as indicated by the increased accumulation of the 125 kDa species from 72 hours pi up to 120 hours pi which did not not observed for the migrating band more rapidly (Figure 2A). The routing of herpesvirus glycoproteins to the Golgi apparatus is associated with the treatment of N-linked oligosaccharides as complex oligosaccharides. The considerable changes in sugar content produce carbohydrate chains which are resistant to the action of endoglycosidase H (endoH). EndoH completely cleaves only ER-type N-linked carbohydrates, whereas hybrid and complex mannose-rich N-linked carbohydrates, cleavage and addition products of Golgi sugars, are cleaved by the action of the peptide. N-glycosidase F (PNGase F). The high number of putative N-linked glycine sites (n = 14) predicted on UL116, the difference between the apparent molecular weight observed (125 kDa) by immunoblot and the calculated molecular mass (34.2 kDa) of the polypeptide of 313 amino acids, suggested that UL116 undergoes an extensive process of glycosylation after translation. To verify the glycosylation status of UL116 during the viral life cycle, HFF cells were infected with TR UL116-Flag for three days before harvesting, and then glycosidase digestion was performed on the extract. As shown in Figure 2C, endoh digestion has different results for both UL116 species: a) the 125 kDa band is weakly affected and glycosidase digestion has reduced its apparent molecular weight (PM) about 10 kDa; b) the 75 kDa band dropped to ~ 38 kDa, a value very close to the predicted PM based on the amino acid sequence (Figure 2C, conquering lanes 1 and 2). PNGase digestion resulted in the disappearance of the faster migrating 76 kDa band that migrated to approximately 35 kDa, while the apparent MW of the 125 kDa band was reduced to ~ 78 kDa (Figure 2C, compare lanes 1 and 3). This result is consistent with the presence of carbohydrate modification after Golgi.
Confocal analysis of pUL116 in infected human fibroblasts.
Confocal microscopy was used to study the localization of ULllo in HCMV infected cells. Anti-Flag antibody was used to trace the subcellular distribution of UL116 together with antibodies specific for cellular compartments or different HCMV proteins. Human fibroblasts were infected with TR UL116-Flag and at 96 hr, fixed and stained for confocal analysis.
Among the cell markers used, ULll6 showed strong colocalization with trans-Golgi TGN 46 marker. Based on the cytoplasmic compartmentalization of UL116, we investigated whether other HCMV structural proteins clustered at the of the site containing UL116. For this purpose, in UL166-Flag TR-infected HFF-1 cells after attachment at 72 hr, antibodies to the pp28 seed coat phosphoprotein and the gL envelope glycoprotein were used. It has been reported previously that both HCMV proteins localize with other coat (pp28) or envelope (gL) proteins at the site of the complex assembly site (AC) of the virus during the late phase of the cycle. infectious and that they are acquired by the virion performing the sorting. Based on confocal microscopy, we found a strong collocation of 1MJL116 with pp28 and gL. These data are consistent with the routing of UL116 to the viral CA site and led us to assume that it could be inserted into the viral particle. UL116 is a component of the envelope of the virion of
HCMC
To verify whether UL116 was incorporated into the virion and to establish its location, we purified recombinant TR UL116-Flag TR viral particles from the supernatant of infected HFF cells and performed both Western blot and electron microscopy.
The UL116-Flag TR particles purified by glycerol density gradient centrifugation / positive tartrate-negative viscosity were subjected to detergent extraction to separate the shell and integument fractions. Both fractions were then immunoblotted with anti-Flag antibody. The glycoprotein B (gB or UL55) and the glycoprotein L (gL or UL115) were also detected by specific antibodies as markers of the envelope fraction. Finally, pp65 (UL83) was chosen as a marker for the seed coat fraction and the non-structural IE1 protein of HCMV (pp72) to rule out the possibility of samples being contaminated with cell extracts. The results of this analysis are shown in Fig. 3. UL116 was present in the same fraction as gB and completely missing in the fraction of the integument in which pp65 was observed (Fig. 3).
To confirm the presence of UL110 on the viral envelope, we performed immunoelectron microscopy on the purified particles of TR UL116-Flag using a viral preparation of wt TR as a control. The anti-Flag secondary antibody labeled with 5 nm gold particles showed a different labeling of the envelope (data not shown). Taken together, these results are consistent with the location of ÜL116 on the surface of the HCMV envelope.
Cotransfection and co-immunoprecipitation of recombinant ÜL116 and glycoprotein H
To further characterize the product of the UL116 gene, we produced a codon-optimized myc-His labeled recombinant version for codons of the protein and cloned it into a eukaryotic expression vector. Surprisingly, transfection into MRC-5 or HEK293T cells did not result in secretion of the recombinant product or its transport to the cell surface. Confocal analysis of the transfected MRC-5 cells showed that UL116 was colocalized almost completely with PDI, a marker of the endoplasmic reticulum. We have assumed that the correct location of UL116 can be obtained after association with one or more of the major glycoproteins in the HCMV envelope.
To investigate this possibility, first, we performed binary cotransfection experiments in HE-293T cells of TR-UL116 with the well-characterized glycoproteins of the HCMV envelope (gH, gL and gB and gO). The detection of the membrane localization of ÜL116 was obtained by a cytofluorometric technique on non-permeabilized cells with anti-UL116 mouse antiserum and a FITC-conjugated secondary anti-mouse antibody. To rule out possible nonspecific detection, we included transfected populations in singles as negative controls. In Figure 4A, we present the results of a representative experiment performed with the UL116 / gH and UL116 / gB complexes. Figure 4A shows a strong signal from UL116 in the plasma membrane only in cells coexpressing UL116-gH (for clarity, cotransfection data of gL and gO are not shown). These data suggest a physical interaction between the gH and UL116 proteins.
To test the formation of a gH-UL116 complex, we performed immunoprecipitation experiments on extracts of HEK293T cells expressing UL116-His plus gH-myc and UL116-
His plus gB. As a control, simple transfections with gH and gB were performed. On a single extract, co-immunoprecipitations were performed by anti-His antibodies, to detect the associéesL116-associated proteins, and by anti-myc antibodies, to reveal the species associated with the gH. Each immunoprecipitated sample was then divided into three aliquots and subjected to Western blot analysis using anti-His (for UL116), anti-myc (for gH) and anti-gB antibodies as probes. The results are shown in Figure 4B. Primary immunoprecipitation of UL116 resulted in coprecipitation with gH but not gB. As a counter-check, primary immunoprecipitation of gH by the anti-myc antibody resulted in co-immunoprecipitation of UL116 (Figure 4B, middle panel). These results demonstrate the formation of a gH / L116 complex using recombinant proteins. UL116 localized in the assembly complex and co-immunoprecipitated with gH in infected HFF-1 cells.
Having demonstrated the interaction between recombinant gH and UL116, we sought to confirm these data in an infection system. We first performed co-immunoprecipitation using anti-gH MSL-109 human monoclonal mAb to extract gH from lysates of HFF-1 cells infected with TR UL116-Flag at 96 hr. gH were separated on SDS-PAGE and Western blotted with an anti-gL antibody as a positive control. As a negative control, a cell lysate from HFF-1 cells infected with wild type TR was used. The results are shown in Figure 5. As eluted proteins, we found the gL in cells infected with both TR-UL116-Flag and wild-type TR, but we also observed that a clear signal for UL-Flag was not present when the wild-type virus was used for infection (Figure 5).
These data suggest that gH and UL116 are cotransported by the secretory pathway, so they must also colocalize intracellularly. To further prove this assumption, confocal microscopy was performed on HFF-1 cells infected with TR UL116-Flag. HFF-1 cells infected with TR UL116-Flag were fixed at 96 hr and stained with both anti-gH mAb and anti-Flag mAb. The results revealed an almost complete overlap of the two proteins, which was restricted to the site of the viral assembly complex. These data demonstrate that 1MJL116 and gH associate during the replication cycle of TR-HCMV and proceed to the cell site of virus assembly and budding.
Negative staining electron microscopy analysis of the purified UL116 / gH complex
We previously used negative-particle electron microscopy to characterize the gH / gL, gH / gL / gO and pentamer complexes bound to neutralizing antibodies. Our data revealed that the HCMV gH / gL complex structurally resembles the HSV-2 gH / gL complex and that the gO and UL128 / UL130 / UL131 subunits bind to the N-terminus of the gH / gH complexes. gL, gH / gL / gO and pentamers respectively. We have also characterized a gL mutant, C144A, which prevents the homodimerization of gH / gL. Here we extend the ME assay to the 3G16-linked gH / UL116 complex, a neutralizing Fab binding to the C-terminal domain of gH (Ciferri et al., Submitted). The trimer complex gH / UL116 / 3G16-Fab was analyzed by electron microscopy of single particles. A 2D class average of the UL116 / gH / 3G16 complex compared to the gH / gLci44A-3G16 complex revealed that the gH / UL116 complex has the same bent structure observed for the gH / gL complex and that the UL116 binds to the region. N-terminal of the gH on the opposite side to 3G16. Further, the ME analysis suggests that UL116 occupies the same site as that occupied by gL in the gH / gL complexes and that the presence of UL116 does not affect the 3G16 binding site on the gH.
Antigenic properties in vivo
An RNA vector expressing both gH and UL116 was produced. Separately, plasmids expressing gH and UL116 were also produced. The recombinantly expressed gH / UL116 complex was purified from a HEK293EBNA cell line. Then, using a mouse model, infection neutralization tests were performed on sera derived from immunization with gH / UL116 (both in the form of RNA and protein complex). The CMV gH / gL complex was used as a reference for immunogenicity tests. In vivo experiments were performed according to Tables 2 and 3. CNE refers to an oil-in-cation emulsion (comprising squalene oil, Span 85 and Tween 80 surfactants, and DOTAP cationic lipid). RV01 refers to a liposome composition (including cationic lipid, zwitterionic lipid, cholesterol, and PEGylated lipid). Both CNE and RV01 were used for the administration of the RNA replicons used in this example. Balb-C mice were injected intramuscularly.
Table 2
Design of the study
Table 3
Immunization and sampling schedules
The results of these tests (Table 4) show that the gH / UL116 complex has antigenic properties.
Table 4
50% Neutralization Securities on Day 63
The various features and embodiments of the present invention, to which reference has been made in individual sections above apply, as appropriate, to the other sections by analogy. Therefore, the characteristics specified in one section may be combined with features specified in other sections, as appropriate.
The dissertation is most fully understood in light of the teachings of the references cited in this memoir. Embodiments within the memory provide an illustration of the embodiments of the invention and should not be construed as limiting the scope of the invention. Those skilled in the art will readily understand that many other embodiments are encompassed by the invention. All of the GenBank publications, patents, and sequences cited in this disclosure are incorporated by reference in their entirety. Insofar as the material incorporated by reference contradicts or is inconsistent with this memory, the memory will supplant any material of this type. The citation of all references herein is not an admission that such references are prior art to the present invention. Those skilled in the art will understand, or will be able to determine using experimentation not exceeding routine, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following embodiments. 1. A recombinant human cytomegalovirus (CMV) protein dimer complex, comprising the CMV gH protein or complex-forming fragment thereof, and CMV λ110 or a complex-forming fragment thereof. 2. The dimer complex of Embodiment 1, wherein said gH complex-forming fragment does not comprise the signal sequence of a full length gH protein. 3. The dimer complex of Embodiment 1 or 2, wherein said gH complex-forming fragment does not comprise the transmembrane domain of a full-length gH protein. 4. The dimer complex of any one of embodiments 1 to 3, wherein said complex-forming fragment of gH comprises the ectodomain of a full-length gH protein. 5. The dimer complex of any one of embodiments 1 to 4, wherein said complex-forming fragment of gH comprises at least one epitope derived from SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO : 3, SEQ ID NO: 4, or SEQ ID NO: 5. 6. The dimer complex of any of Embodiments 1 to 5, wherein said gH or one of its complex-forming fragments comprises a a sequence selected from the group consisting of SEQ ID NOs: 1, 2, 3, 4, and 5. 7. The dimer complex of any one of embodiments 1 to 6, wherein said complex-forming moiety of UL116 comprises at least one epitope derived from SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17. 8. The dimer complex of any of the embodiment 1 to 7, wherein said UL116 or complex-forming fragment thereof comprises a sequence selected from the group consisting of SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, and 17. 9. The dimer complex of any one of embodiments 1 to 8, wherein said gH protein or complex-forming fragment thereof, and CMV UL116 or one of its complex-forming fragments, are fused into one single polypeptide chain. 10. An immunogenic composition comprising the dimer complex of any of embodiments 1-9. The immunogenic composition of embodiment 10, further comprising a CMV protein or additional CMV protein complex. 12. The immunogenic composition of Embodiment 11, wherein said CMV protein or said additional CMV protein complex is selected from the group consisting of gB, gH, gL, gO, gM, gN; UL128, UL130, UL131, RL10, RL11, RL12, RL13, UL4, UL5, UL10, UL80.5, UL119, UL122, UL133, UL138, UL148A, UL1, UL7, UL9, UL16, UL18, UL20, UL40, UL41A, UL42, UL47, UL111A, UL124, UL132, UL136, UL141, one of their immunogenic fragments, one of their complex-forming fragments, and one of their combinations. 13. The immunogenic composition of Embodiment 11 or 12, wherein said additional CMV protein is gB or an immunogenic fragment thereof. 14. The immunogenic composition of any of embodiments 11 to 13, wherein said additional CMV protein complex is selected from the group consisting of the pentamer complex gH / gL / ULl28 / UL130 / UL131, gH complex / gL, trimer complex gH / gL / gO, gM / gN complex, or a combination thereof. 15. The immunogenic composition of any one of embodiments 10 to 14, further comprising an adjuvant. 16. The immunogenic composition of embodiment 14, wherein the adjuvant is selected from the group consisting of: an aluminum salt (such as aluminum phosphate, aluminum hydroxide), an oil emulsion, water (such as MF59), a TLR7 agonist, IC31, Eisai 57, ISCOMs, a CpG oligonucleotide, PET-lipid A, and one of their combinations. 17. An isolated nucleic acid, or a combination of isolated nucleic acids, comprising one or more polynucleotide sequences encoding the dimer complex of any one of embodiments 1 to 9. 18. The isolated nucleic acid (s) of Embodiment 17, wherein said isolated nucleic acid (s) is RNA, preferably self-replicating RNA. 19. The isolated nucleic acid (s) of Embodiment 18, said self-replicating RNA being an alphavirus replicon. 20. An alphavirus replication particle (VRP) comprising the alphavirus replicon of Embodiment 19. An immunogenic composition comprising the nucleic acid (s) of any one of Embodiments 17-19. 22. An immunogenic composition comprising the VRP of Embodiment 20. 23. The immunogenic composition of Embodiment 21 or 22, further comprising an adjuvant. 24. The immunogenic composition of embodiment 23 wherein the adjuvant is selected from the group consisting of: an aluminum salt (such as aluminum phosphate, aluminum hydroxide), an oil-in-water emulsion, water (such as MF59), a TLR7 agonist, IC31, Eisai 57, ISCOMs, a CpG oligonucleotide, PET-lipid A, and one of their combinations. 25. A host cell comprising the nucleic acid (s) of any one of embodiments 17 to 19. 26. The host cell of embodiment 25, wherein said one or more nucleic acids are DNA. 27. The host cell of Embodiment 26, said host cell being a mammalian cell. 28. The host cell of Embodiment 27, said mammalian cell being a CHO cell or HEK-293 cell. 29. The host cell of any one of embodiments 26 to 28, wherein said DNA encoding the CMV dimer complex is stably integrated into the genomic DNA of said host cell. 30. The host cell of any one of embodiments 26 to 29, when grown under suitable conditions, said host cell expressing a soluble gH / ULH6 dimer complex. 31. The host cell of Embodiment 30, wherein said soluble dimer complex is secreted from the host cell. 32. A cell culture comprising the host cell of embodiments 25 to 31, said culture having a size of at least 20 liters. 33. A cell culture comprising the host cell of embodiments 25 to 31, said culture having a size of at least 100 liters. 34. A cell culture comprising the host cell of embodiments 25 to 31, wherein the yield of said dimer complex is at least 0.05 g / l. 35. A cell culture comprising the host cell of embodiments 25 to 31, wherein the yield of said dimer complex is at least 0.1 g / l. 36. A method of producing a recombinant human cytomegalovirus (CMV) protein dimer complex comprising the CMV gH protein or one of its complex-forming fragments, and CMV UL116 or a fragment thereof complex forming, comprising: (i) culturing the host cell of any one of embodiments 25 to 31 under appropriate conditions, thereby expressing said dimer complex; and (ii) harvesting said dimeric complex from the culture. 37. The method of Embodiment 36, further comprising purifying said dimer complex. 38. A recombinant human cytomegalovirus (CMV) protein dimeric complex, including the CMV gH protein or complex-forming fragment thereof, and CMV ULll6 or complex fragment thereof, produced by the method of embodiment 36 or 37. 39. A method of inducing an immune response against cytomegalovirus (CMV), comprising administering to a subject in need of an immunologically effective amount of the immunogenic composition of any of Embodiments 10 to 16 and 21 to 24. 40. The method of Embodiment 39, wherein the immune response comprises the production of neutralizing antibodies against CMV. 41. The method of embodiment 40, wherein the neutralizing antibodies are complement-independent. 42. A method of inhibiting the entry of cytomegalovirus (CMV) into a cell, comprising contacting the cell with the immunogenic composition of any one of Embodiments 10 to 16 and 21 to 24. 43 The immunogenic composition of any of Embodiments 10 to 16 and 21 to 24 for use in inducing an immune response against cytomegalovirus (CMV). 44. Use of the immunogenic composition of any of embodiments 10 to 16 and 21 to 24 for inducing an immune response against cytomegalovirus (CMV). 45. Use of the immunogenic composition of any of embodiments 10 to 16 and 21 to 24 in the manufacture of a medicament for inducing an immune response against cytomegalovirus (CMV).
权利要求:
Claims (15)
[1]
A recombinant human cytomegalovirus (CMV) protein dimer complex, comprising the CMV gH protein or complex-forming fragment thereof, and CMV UL116 or a fragment thereof forming a complex.
[2]
2. A dimer complex according to claim 1, wherein said gH complex-forming fragment does not comprise the transmembrane domain of a full-length gH protein.
[3]
The dimer complex of claim 1 or 2, wherein said gH complex-forming fragment comprises 1'ododomain of a full-length gH protein.
[4]
A dimer complex according to any one of claims 1 to 3, wherein said gH or a complex-forming fragment thereof comprises a sequence selected from the group consisting of SEQ ID NO: 1, 2, 3, 4, and 5.
[5]
A dimer complex according to any one of claims 1 to 4, wherein said UL116 or complex-forming fragment thereof comprises a sequence selected from the group consisting of SEQ ID NO: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, and 17.
[6]
6. A dimer complex according to any one of claims 1 to 5, wherein said gH protein or complex-forming fragment thereof, and CMV λ110 or a complex fragment thereof, are fused together. into a single polypeptide chain.
[7]
An immunogenic composition comprising the dimer complex of any one of claims 1 to 6, optionally comprising an adjuvant.
[8]
The immunogenic composition of claim 7, further comprising a CMV protein or an additional CMV protein complex.
[9]
The immunogenic composition of claim 8, wherein said additional CMV protein is gB or an immunogenic fragment thereof.
[10]
The immunogenic composition according to claim 8 or 9, wherein said additional CMV protein complex is selected from the group consisting of pentamer gH / gL / UL128 / UL130 / UL131, gH / gL complex, trimer gH / gL / gO, complex gM / gN, or one of their combinations.
[11]
An isolated nucleic acid, or combination of isolated nucleic acids, comprising one or more polynucleotide sequences encoding the dimer complex of any one of claims 1 to 6.
[12]
12. Isolated nucleic acid (s) according to claim 11, wherein said isolated nucleic acid (s) is (are) RNA, preferably self-replicating RNA.
[13]
An immunogenic composition comprising the acid or nucleic acids of claim 11 or 12, optionally comprising an adjuvant.
[14]
14. Host cell comprising the acid or nucleic acids according to claim 11 or 12.
[15]
15. An immunogenic composition according to any one of claims 7 to 10 and 13 for use in inducing an immune response against cytomegalovirus (CMV).
类似技术:
公开号 | 公开日 | 专利标题
BE1024516B9|2018-04-23|ANTIGENS OF CYTOMEGALOVIRUS
BE1023077A1|2016-11-17|CMV ANTIGEN AND USES THEREOF
JP6657150B2|2020-03-04|Compositions and methods for the treatment of cytomegalovirus
BE1023087B1|2016-11-18|ANTIGENS OF CYTOMEGALOVIRUS AND USES THEREOF
JP6643239B2|2020-02-12|Immunogenic middle east respiratory syndrome coronavirus | compositions and methods
MX2014003777A|2015-05-15|Recombinant nanoparticle rsv f vaccine for respiratory syncytial virus.
US20150307850A1|2015-10-29|Conditional replicating viral vectors
US7223584B2|2007-05-29|Recombinant rabies vaccine and methods of preparation and use
BE1023077B1|2016-11-17|CMV ANTIGEN AND USES THEREOF
US11253587B2|2022-02-22|Vaccine compositions for the treatment of coronavirus
WO2022040220A2|2022-02-24|Severe acute respiratory syndrome coronavirus [sars-cov-2]-virus-like particle [vlp] vaccine: compositions, delivery strategies, methods and uses
同族专利:
公开号 | 公开日
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

法律状态:
优先权:
申请号 | 申请日 | 专利标题
EP15152415.4|2015-01-23|
[返回顶部]