![]() ANTIGENS OF CYTOMEGALOVIRUS AND USES THEREOF
专利摘要:
This disclosure provides modified cytomegalovirus (CMV) gL proteins and complexes comprising gL proteins. The modified gL proteins remain intact and are able to form complexes with other CMV proteins. 公开号:BE1023087B1 申请号:E2016/5050 申请日:2016-01-22 公开日:2016-11-18 发明作者:Andrea Carfi;Claudio Ciferri;Yi Xing 申请人:Glaxosmithkline Biologicals S.A.; IPC主号:
专利说明:
ANTIGENS OF CYTOMEGALOVIRUS AND USES THEREOF The present application contains a sequence listing which has been electronically submitted in ASCII format and which is incorporated herein by reference in its entirety. The ASCII copy, created on January 14, 2016, is named VN056504WO_SL.txt and has a size of 26,466 bytes. Field of invention This invention is in the field of cytomegalovirus (CMV) antigens that can be used for vaccines. Background of the invention Cytomegalovirus is a kind of virus that belongs to the viral family known as herpesviruses or herpesviruses. The species that infects humans is commonly known as human cytomegalovirus (HCMV) or human herpesvirus type 5 (HHV-5). Of the herpesviridae, HCMV belongs to the subfamily beta-herpesviridae, which also includes cytomegaloviruses from other mammals. Although they can be found throughout the body, HCMV infections are often associated with salivary glands. HCMV infected between 50% and 80% of adults in the United States (40% worldwide), as indicated by the presence of antibodies in most of the general population. HCMV infection is generally unheard of in healthy people, but it can be life-threatening for immunocompromised people such as HIV-infected people, organ transplant recipients, or newborns. HCMV is the virus most commonly transmitted to a developing fetus. After infection, HCMV has the ability to remain latent within the body for life of the host, with occasional reactivations since latency. Given the severity and importance of this disease, obtaining an effective vaccine is considered a top priority for public health (Sung, H., et al., (2010) Expert review of vaccines 9, 1303 Schleiss, Expert Opin Ther Ther Apr. 2010; 20 (4): 597602). Genomes from more than 20 different strains of HCMV were sequenced, including both laboratory strains and clinical isolates. For example, the following strains of HCMV were sequenced: Towne (GL239909366), AD169 (GI: 219879600), Toledo (GL290564358) and Merlin (GI: 155573956). Strains of HCMV ADI69, Towne and Merlin can be obtained from the American Type Culture Collection (ATCC VR538, ATCC VR977 and ATCC VR1590, respectively). Cytomegalovirus contains an unknown number of membrane protein complexes. Of the approximately 30 known glycoproteins in the viral envelope, gH and gL appeared to be particularly interesting because of their presence in several different complexes: dimer gH / gL, trimer gH / gL / gO (also known as the gCIII complex ), and pentamer gH / gL / pUL128 / pUL130 / pUL131 (pUL131 is also referred to as "pUL131A", "pUL131a", or "UL131A"; subunits pUL128, pUL130, and PÜL131 are sometimes also referred to as UL128, UL130, UL131; ). CMV is thought to use pentamer complexes to enter epithelial and endothelial cells by endocytosis and low pH-dependent fusion but is thought to enter the fibroblasts by direct fusion at the plasma membrane in a process involving the gH / gL complex or optionally gH / gL / gO. The gH / gL and / or gH / gL / gO complex (s) is / are sufficient for fibroblast infection, whereas the pentamer complex is required to infect endothelial and epithelial cells. The pentamer complex is considered a major target for vaccination against CMV. The viral genes UL128, UL130 and UL131 are required for entry into endothelial cells (Hahn, Journal of Virology 2004; 78: 10023-33). Non-endothelial tropic strains adapted to fibroblasts contain mutations in at least one of these three genes. The Towne strain, for example, contains a two-base insert causing frame shift in the UL130 gene, while strain AD169 contains a base pair insertion in the UL131 gene. Strains both Towne and ADI69 could be adapted for growth in endothelial cells, and in both cases frame shift mutations in the UL130 or UL131 genes were repaired. US Pat. No. 7,704,510 discloses that pUL131A is necessary for tropism of endothelial cells. US Patent 7,704,510 also discloses that pUL128 and pUL130 form a complex with gH / gL, which is incorporated in virions. This complex is required to infect endothelial and epithelial cells but not fibroblasts. Anti-CD46 antibodies have been found to inhibit HCMV infection of epithelial cells. CMV vaccines tested in clinical trials include Towne vaccine, Towne-Toledo chimeras, an alphavirus replicon with gB as an antigen, the gB / MF59 vaccine, a gB-based vaccine produced by GlaxoSmithKline, and a DNA-based vaccine using gB and pp65. Pp65 is a viral protein that is a potent inducer of CD8 + responses to CMV. These vaccines are all weak antibody inducers that block entry into endothelial / epithelial cells (Adler, S.P. (2013), British Medical Bulletin, 107, 5768. doi: 10.1093 / bmb / ldt023). Preclinical animal studies of CMV vaccines include an inactivated strain AD169 that has been repaired in the UL131 gene, a DNA-based vaccine using a wild-type UL130 gene and peptide-based vaccines using pUL130 peptides and 131 (Sauer, A, et al., Vaccine 2011/29: 2705-1, doi: 10.1016). The CMV gB antigen is considered a weak inducer of antibodies that block entry into endothelial / epithelial cells. In a Phase II clinical trial, the gB./MF59 vaccine was only 50% effective in preventing primary infection in young women with a child at home (Pass, RF, et al. , N Engl J Med 2009; 360: 1191-9). Therefore, there is a need to develop vaccines against CMV comprising other antigenic targets such as gH / gL, gH / gL / gO, or the pentamer complex gH / gL / pUL128 / pUL130 / pUL131. Summary of the invention As disclosed and exemplified herein, the inventors have found that when the cytomegalovirus gL antigen is recombinantly expressed and purified from a mammalian host (such as a CHO cell or a HEK cell), a significant portion of the gL is cleaved. To improve recombinant expression and purification of the intact gL protein, mutations have been introduced to reduce protease cleavage of gL. The mutants show an increase in protease cleavage resistance during recombinant production. Accordingly, in one aspect, the invention provides a recombinant CMV gL protein, or a complex-forming fragment thereof, wherein said gL protein or fragment thereof comprises a mutation at the recognition site of proteases, wherein said mutation reduces protease cleavage at said protease recognition site compared to a control. The protease recognition sign refers to residues 91 to 102 (numbering based on SEQ ID NO: 1). Preferably, the mutation reduces protease cleavage compared to a control, without changing the secondary structure of the C-terminal portion of the protease recognition site (which is thought to have a beta-strand conformation). There is also provided here CMV complexes comprising gL proteins or fragments thereof described herein. Such complexes may be a gH / gL complex, a gH / gL / gO complex, and a pentamer gH / gL / pUL128 / pUL130 / pUL131 complex. Also provided herein are nucleic acids encoding CMV gL proteins and their complex-forming fragments as described herein. The nucleic acid can be used as a nucleic acid vaccine (e.g., a self-replicating RNA molecule encoding gL or a complex fragment thereof). The nucleic acid can also be used for the recombinant production of gL proteins or fragments thereof, or a CMV complex comprising gL proteins or fragments thereof. The invention also provides a host cell comprising the nucleic acids described herein. The nucleic acids may be used by the host cell to express a gL protein or a complex-forming fragment thereof, or a CMV complex comprising gL or a fragment thereof forming a complex. Preferably, the CMV complex can 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. When used to express the CMV pentameric complex gH / gL / pUL128 / pUL130 / pUL131, it is preferred that the pentamer complex yield be at least 0.1 g / l. The invention also provides a method of inducing an immune response against CMV, comprising administering to a subject in need of an immunologically effective amount of the gL protein, or a fragment thereof forming a complex, or a CMV complex comprising the gL protein or a fragment thereof, as described herein. The invention also provides a method of inhibiting the entry of cytomegalovirus (CMV) into a cell, comprising contacting the cell with the gL protein, or a fragment thereof forming a complex, or complex CMV comprising the gL protein or a fragment thereof as described herein. It is also proposed here the use of compositions described herein for the induction of an immune response against CMV, and the use of the compositions described herein in the manufacture of a medicament for the induction of an immune response against CMV. Brief description of the figures Figure 1A shows the alignment of partial sequences of gL proteins from different herpesviruses near the protease recognition site (SEQ ID NOs: 12-15, respectively, in order of appearance). Figure 1B shows the secondary structure of the gH / gL complex from HSV-2 and VZV. The arrow indicates the cleavage site. Figure 2A shows the partial mutant sequences of gL (SEQ ID NO: 15-26, respectively, in order of appearance). Figure 2B shows the result of Western blot analysis using anti-gL antibodies. Figure 2C shows the result of Western blot analysis using anti-His antibodies. Figure 3 depicts the Western blot analysis of penta WT and mutant LSG using either unreduced (NR) protein samples or reduced and boiled (RB) samples. Figure 4 shows the Western blot analysis of penta WT and IDG mutant using either unreduced (NR) protein samples or reduced and boiled (RB) samples. Figure 5A shows penta WT and penta mutants IDG and LSG. Figure 5B shows the titer of serum neutralizing antibodies (NAB) of mice immunized with penta WT, mutant LSG, or mutant IDG adjuvanted with MF59. Detailed description of the invention 1. General As disclosed and exemplified herein, the inventors have found that when the cytomegalovirus gL antigen is recombinantly expressed and purified from a mammalian host (such as a CHO cell or a HEK cell), a significant portion of the gL is cleaved (also called "gL cutoff") by an unknown protease. In fact, it has been observed that gL cleavage occurs during recombinant expression and purification of three different CMV complexes: the gH / gL complex, the gH / gL / gO complex, and the pentamer gH / gL complex. / pUL128 / pUL130 / PÜL131. Cleavage of gL caused non-homogeneity of antigen production, and potential loss of neutralizing sites on gL-based antigens. Using Western blot analysis and N-terminal sequencing, the inventors identified and located the cleavage site at the peptide bond between residues 97 and 98 of the gL from the Merlin strain (SEQ ID NO: 1 ) (figure 1) . To solve the cleavage problem, the inventors have studied the structural features of gL proteins from several related herpesviruses, including HSV1, HSV2, and VZV. GL proteins from HSV1, HSV2, and VZV did not appear to have a cleavage problem. Based on the structural studies, the inventors have found that mutations can be introduced into the protease recognition site, including amino acid residues 91 and 102, to reduce protease cleavage of the produced gL. recombinant. For example, as exemplified herein, the A96L / N97S / S98G triple mutation (the "LSG" mutant) and the A96I / N97D / S98G triple mutation (the "IDG" mutant) substantially eliminated the gL cleavage problem. . Two other mutants, the deletion of the Asn97 residue (Asp97 delta), and A96S / N97S / S98T (the "SST" mutant), also showed a considerable decrease in gL cleavage when gH and gL were coexpressed. Based on the structural analysis of gL proteins from other herpesviruses (Figure 1), it appears that the protease recognition site adopts a short helix from the N-terminus to the C-terminus. a possible (91VTPE94) (SEQ ID NO: 27), a short loop (95AA96), and a conserved β strand structure (97NSVLLD102) (SEQ ID NO: 7). Cleavage occurs at the N-terminus of the β strand (Figure 1). A β strand is a structural unit of β sheets in proteins. It is an extended sequence of polypeptide chain generally of a length of 3 to 10 amino acids which form hydrogen bonds with other β strands in the same β sheet. As shown in FIG. 1, this β (β4 in FIG. 1) strand together with the β5 and β6 strands of the gL, as well as the β strands of the gH, form a β-sheet. Therefore, in preferred embodiments, the mutation will maintain the secondary structure of the C-terminal portion of the protease recognition site (i.e., the β-strand conformation is conserved, such that interactions between β4 and other β-strand (s) are substantially maintained). Maintaining the β-strand structure can potentially reduce any negative impact on the assembly of CMV complexes (such as pentamer complexes), and may also potentially preserve important immunogenic epitopes. For example, one or more residues of the protease recognition site may be substituted with a corresponding residue of another herpesvirus (such as HSV-1, HSV-2, or VZV). As shown in Fig. 1, sequence and structure analysis shows that substitution of a CMV residue by a corresponding residue of HSV-1, HSV-2, or VZV does not change the β-strand conformation, and at the same time protease cleavage can be reduced. Optionally, the short loop structure immediately preceding the β strand (95AA96 in FIG. 1) can also be maintained. Therefore, in one aspect, the invention provides a recombinant cytomegalovirus (CMV) gL protein, or a fragment thereof forming a complex, wherein said gL protein or fragment thereof comprises a mutation at the site of protease recognition, wherein said mutation reduces protease cleavage at said protease recognition site compared to a control. The protease recognition site refers to residues 91 to 102 (numbering based on SEQ ID NO: 1). Preferably, the mutation reduces cleavage by proteases compared to a control, without changing the β-strand structure at the C-terminal portion of the protease recognition site. There is also provided here CMV complexes comprising the gL proteins or fragments thereof described herein. Such complexes may be the gH / gL complex, the gH / gL / gO complex, and the pentamer gH / gL / pUL128 / pUL130 / pUL131 complex. There is also provided here host cells for the recombinant expression of gL proteins or fragments thereof described herein, and CMV complexes comprising the gL proteins or fragments thereof described herein. As noted, cleavage of gL was observed in mammalian host cells during the recombinant production process. Therefore, the mutations disclosed herein are particularly suitable for the recombinant production of CMV vaccines in mammalian hosts (which are preferred hosts for many biological products). For example, HEK-293 and CHO cells have been used for a long time for the commercial production of biological products. Therefore, the incorporation of mutations that reduce gL cleavage can improve the efficiency and yield of production, and reduce the formation of a partially degraded contaminant product. 2. Definitions- The term "complex-forming fragments" of a cytomegalovirus (CMV) protein (such as gL) refers to any portion or portion of the protein that retains the ability to form a complex with another CMV protein. Such complexes include, for example, the dimer complex gH / gL, the trimer complex gH / gL / gO, or the pentamer complex gH / gL / pUL128 / pUL130 / pUL131. A "pentamer-forming moiety" of a CMV protein (such as gL) refers to any portion or portion of the protein that retains the ability to form the pentamer complex gH / gL / pUL128 / pUL130 / pUL131. . As used herein, "pentamer complex" or "pentamer" refers to a CMV complex that comprises five different subunits: gH, gL, pUL128, pUL130, and pUL131. Although generally referred to as pentamer gH / gL / pUL128 / pUL130 / pUL131 (or pentamer complex including gH, gL, pUL128, pUL130, and pUL131) in the memory, each of the five subunits does not need to be full length; the term also encompasses pentamers formed by complex-forming fragments of gH, gL, pUL128, pUL130, and pUL131. The term "mutation" refers to the addition, deletion, or substitution of an amino acid residue. The term also includes modifications that introduce a non-naturally occurring amino acid or amino acid analog into a polypeptide chain. The charged amino acid residues include: D, E, K, R, and H. Uncharged polar residues include: S, T, C, Y, N, and Q. Nonpolar or hydrophobic residues include: A, V, L, I, M, W, F, and P. Amino acids comprising a large side chain include: W, F, M, Y, Q, R, E, H, and K. Amino acid residues lacking a side chain or comprising a small side chain include: , A, V, S, T, C, D, and N. An amino acid residue includes a "bulky side chain" when the side chain comprises a branched or cyclic substituent. Examples of amino acid residues with a bulky side chain include tryptophan, tyrosine, phenylalanine, homophenylalanine, leucine, isoleucine, histidine, 1-methyltryptophan, α-methyltyrosine Α-methyl-phenylalanine, α-methyl-leucine, α-methyl-isoleucine, α-methyl-histidine, cyclopentyl-alanine, cyclohexylalanine, naphthyl-alanine, and the like. Although the present invention is applicable to gL proteins from any CMV strain, for ease of understanding, when reference is made to amino acid positions herein, the numbering is given relative to the sequence. of amino acids of the gL protein of SEQ ID NO: 1 from the Merlin strain, unless otherwise indicated. However, the present invention is not limited to the Merlin strain. Using the teachings of the present invention, comparable amino acid positions in a gL protein of any other CMV strain can be determined by a person of average skill in the art by aligning the amino acid sequences using algorithms easily available and well-known (such as BLAST, using the default settings; ClustalW2, using the default settings; or the algorithm disclosed by Corpet, Nucleic Acids Research, 1998, 16 (22): 10881- 10890, using the default settings). Therefore, when referring to a "CMV gL protein", one must understand a CMV gL protein of any strain (in addition to the Merlin strain). The true number may have to be adjusted for gL proteins from other strains according to the true alignment of the sequence. For example, the "protease recognition site" is defined as consisting of amino acid residues 91 to 102 consisting especially of residues 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101 and 102. These numbers are relative to the amino acid sequence of the gL protein of SEQ ID NO: 1. The protein protease recognition site of the gL proteins of other CMV strains, or other mutants or variants of The gL, or fragments of the gL can be determined using standard sequence alignment programs that align a query sequence with SEQ ID NO: 1, and identify residues that correspond to residues 91 to 102 of SEQ ID NO. : 1. The positions of specific amino acid residues are also numbered according to SEQ ID NO: 1. For example, "S98" refers to position 98 of SEQ ID NO: 1 (which is an S), as well as residues. corresponding to other sequences of gL (or variants or fragments) that correspond to S98 of SEQ ID NO: 1, when the sequence is aligned with SEQ ID NO: 1. For simplicity, any residue of a sequence of gL (or a variant or fragment) that corresponds to S98 of SEQ ID NO: 1 is called S98, although the true position of this residue may or may not be 98, and that the true residue may or may not be S For example, a conservative substitution (e.g., T) may be aligned with S98 of SEQ ID NO: 1. A conservative substitution is generally identified as "positive" or "+" by BLAST 2. Similarly, the mutations are also identified according to the numbering of SEQ ID NO: 1. For example, S98G means that any residue of a gL (or variant or fragment) sequence that corresponds to S98 of SEQ ID NO: 1 is mutated to G. An amino acid residue of a query sequence "corresponds to" a designated position of a reference sequence (eg, S98 of SEQ ID NO: 1) when, by alignment of the query amino acid sequence with the reference sequence, the position of the residue corresponds to the designated position. Such alignments can be done by hand or using well-known sequence alignment programs such as ClutalW2, or "BLAST 2 Sequences" using the default settings. A "" refers to a sequence that is at least 10 amino acid residues long and is at least 50% identical to SEQ ID NO: 5. As shown in FIG. In the wild type of the Merlin strain, a 17 residue fragment unique to CMV gL compared to HSV1, HSV2 and VZV was identified (represented by "ηΐ"). Preferably, the region of the insert comprises at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, or at least 20. residues, and / or is at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least less than 95% or 100% to SEQ ID NO: 5. In some embodiments, the region of the insert comprises a sequence in which one to eight amino acid residues of SEQ ID NO: 5 are substituted so conservative. "Conservatively substituted" means that a residue is replaced by another biologically similar residue. Examples include the substitution of amino acid residues with similar characteristics, for example, small amino acids, acidic amino acids, polar amino acids, basic amino acids, hydrophobic amino acids and aromatic amino acids. Examples of conservative amino acid substitutions include those of the following Table 1, and analogous substitutions of the original residue by unnatural alpha-amino acids that have similar characteristics. Table 1 Unless otherwise specified, the percent identity of two sequences is determined over the entire length of the shorter of the two sequences. 3. Modified gL proteins and CMV complexes A. Modified gL proteins In one aspect, the invention provides a modified CMV gL protein, or complex fragment thereof, that reduces cleavage (cleavage) at the peptide bond between N97 and S98 residues. The glycoprotein L (gL) of human CMV is encoded by the UL115 gene. G1 is thought to be essential for viral replication and all known functional properties of gL are directly associated with its dimerization with gH. The gH / gL complex is required for the fusion of the viral and plasma membranes leading to the entry of the virus into the host cell. The HCLV Merlin strain (GI: 39842115, SEQ ID NO: 1) and the strain were reported to be present. Towne of HCMV (GI: 239909463, SEQ ID NO: 2) is 278 amino acids in length. It has been reported that the HCMV strain AD169 (GI: 2506510, SEQ ID NO: 3) has a length of 278 amino acids, includes a signal sequence at its N-terminus (amino acid residues 1 to 35), has two N-glycosylation sites (at residues 74 and 114) and lacks a TM domain (Rigoutsos, I, et al., Journal of Virology 77 (2003): 4326-44). The N-terminal signal sequence in SEQ ID NO: 1 is predicted to include amino acid residues 1 to 30. SEQ ID NO: 2 shares 98% amino acid identity with SEQ ID NO: 1. Sequencing of full length gL gene from 22 to 39 clinical isolates, as well as AD169 laboratory strains, Towne and Toledo revealed less than 2% variation in amino acid sequences among isolates (Rasmussen, L, et al., Journal of Virology 76 (2002): 10841-10888). Generally, the N-terminal signal sequence of the gL proteins is cleaved by a peptidase signal from the host cell to produce the mature GL proteins. To the gL proteins in the HCMV membrane complexes of the invention, an N-terminal signal sequence may be missing. An example of a gL protein lacking the N-terminal signal sequence is SEQ ID NO: 4, in which an N-terminal signal sequence is missing and which consists of amino acid residues 31 to 278 of SEQ ID NO: 1. While gL is thought to be essential for viral replication, all the functional properties of gL are directly associated with its dimerization with gH. The gL proteins of the invention may be variants of gL which have varying degrees of identity with SEQ ID NO: 1 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, or SEQ ID NO: 3. of the invention may have various degrees of identity with SEQ ID NO: 4 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: 4. In some embodiments, variant gL proteins: (i) form part of the gH / gL dimer complex; (ii) form part of the trimer complex gH / gL / gO; (iii) form part of the pentamer complex gH / gL / pUL128 / pUL130 / pUL131; (iv) comprise at least one epitope derived from SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4; and / or (v) can elicit in vivo antibodies that exhibit cross-reactivity with a CMV virion. Also encompassed herein are complex-forming fragments of the proteins described herein. A complex-forming fragment of the gL may be any portion or portion of the gL protein that retains the ability to form a complex with another CMV protein. In some embodiments, a complex forming fragment of gL forms part of the dimer gH / gL complex. In some embodiments, a complex forming fragment of gL forms part of the trimer complex gH / gL / gO. In some embodiments, a complex fragment of the GL forms part of the pentamer complex. gH / gL / pUL128 / pUL130 / pUL131. A complex-forming fragment of Ig can be obtained or determined by standard techniques known in the art, such as co-immunoprecipitation technique, crosslinking, or colocalization by fluorescent staining, etc. In some embodiments, the complex fragment of the gL further (i) comprises at least one epitope derived from SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4 ; and / or (ii) can elicit antibodies in vivo that exhibit cross-reactivity with a CMV virion. In some embodiments, the gL protein described herein, or one of its complex-forming fragments, includes a mutation at the protease recognition site (residues 91 to 102), wherein said mutation reduces protease cleavage. at said protease recognition site, compared to a control. Various cookies can be used. The level of protease cleavage (at the level of the peptide bond between residues 97 and 98) of a corresponding wild-type gL in substantially the same conditions can be used as a control. Alternatively, a control may be a predetermined rate or a threshold level (e.g., 20%, 25%, or 30% of the total gL protein). The percentage refers to the molar percentage. For example, the mutation may result in reduced protease cleavage at the peptide bond level between residues 97 and 98 of at least 10%, at least 20%, at least 30%, less than 40%, at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%, etc., compared to that of wild type, when recombinant expression in a mammalian host cell under standard culture conditions for this host cell . Alternatively or additionally, protease cleavage is reduced at least 3-fold, at least 5-fold, at least 20-fold, at least 30-fold, at least 40-fold, at least 50 times, at least 60 times, at least 70 times, at least 75 times, at least 80 times, at least 90 times or at least 100 times, wild type, during an expression. recombinant in a mammalian host cell under standard culture conditions for this host cell. Alternatively or additionally, the mutation may be a mutation where not more than about 35% of the gL molecules, or their complex-forming fragments, are cleaved at a peptide bond between residues 97 and 98, when recombinant expression in a mammalian host cell under standard culture conditions for that host cell. For example, the mutation may result in not more than about 35%, not more than about 30%, not more than about 25%, not more than about 20%, not more than about 15% , not more than about 10%, not more than about 9%, not more than about 8%, not more than about 7%, not more than about 6%, not more than about 5%, not more than more than about 4%, not more than about 3%, not more than about 2%, or not more than about 1% of the gL molecules, or one of their complex fragments, are cleaved at the level of a peptide bond between residues 97 and 98, during recombinant expression in a mammalian host cell under conventional culture conditions for this host cell. The percentage refers to the molar percentage. The conventional culture conditions for commonly used mammalian host cells are known. For example, for a CHO cell, a typical culture condition may be a temperature of 36.5 ° C in a pH 7.0 medium with <10% CO2. In a specific example, Expi293 cells were transfected to express the pentamer complex (gH / gL / pUL128 / pUL130 / pUL131) at 37 ° C and in pH 7.0 medium at 8% CO 2 for three days, and Cell culture supernatants were affinity purified and analyzed by Western blots, as shown in the examples below. The mutation comprises an addition, deletion, substitution, or combination thereof of an amino acid residue. Preferably, the mutation substantially preserves the secondary structure of the C-terminal portion of the protease recognition site. In particular, as shown in FIG. 1, the residues of the C-terminal portion of said protease recognition site form a β-strand, which is thought to interact with other β strands to form a β-sheet. Preferably, said mutation maintains the conformation of this β strand. Potential advantages of maintaining the secondary structure include, for example, facilitating the assembly of complexes containing Ig (eg, gH / gL, gH / gL / gO, or gH / gL / pIL128 / pUL130 / pUL131). , and the maintenance of key immunogenic epitopes. Optionally, the short loop structure immediately preceding the β strand is also preserved. Many computer programs and algorithms are available to predict secondary structure, including I-TASSER, HHpred, RaptorX, MODELLER, SWISS-MODEL, Robetta Beta, SPARKSx, PEP-FOLD, Phyre and Phyre2, RAPTOR, QUARK, Abalone, Foldit etc. Whether a mutation changes the secondary structure of the protease recognition site can be analyzed using these tools. In some embodiments, the mutation comprises the addition of one or more amino acid residues. For example, the mutation may include the addition of two to five amino acid residues. In some embodiments, the two to five amino acid residues comprise both one or more polar residues and one or more nonpolar residues. In some embodiments, the mutation comprises the addition of one or more residues between residues N97 and S98. As shown in the examples, the peptide bond between N97 and S98 is cleaved by a protease; therefore, the introduction of one or more additional residues between N97 and S98 can produce a mutant gL (or fragment) that is more resistant to cleavage. In one exemplary embodiment, the mutation comprises the addition of F, Q, or one of their combinations, between residues 97 and 98. In an exemplary embodiment, the mutation comprises the addition of FQ or QF between residues 97 and 98. In some embodiments, the mutation comprises the deletion of one or more amino acid residues, such as the deletion of one to three amino acid residues. In some embodiments, the mutation comprises deleting at least one residue selected from the group consisting of: V91, T92, P93, E94, A95, A96, N97, S98, V99, L100, L101, D102, and one of their combinations. In some embodiments, the mutation comprises deleting at least one residue selected from the group consisting of: E94, A95, A96, N97, S98, V99, L100, L101, D102, and one of their combinations . In one exemplary embodiment, the mutation comprises the deletion of at least one residue selected from the group consisting of: A96, N97, S98, and one of their combinations. In an exemplary embodiment, the mutation comprises N97 deletion. In some embodiments, the mutation comprises substitution of a residue with a corresponding residue of the gL protein of another herpesvirus. The herpesvirus family (Herpesviridae) includes, for example, Herpes simplex virus 1 and 2 (HSV-1 or HHV-1, HSV-2 or HHV-2), varicella zoster virus (VZV or HHV-3). ), Epstein Barr virus (EBV or HHV-4), human herpesvirus type 6 (HHV-6), human herpesvirus type 7 (HHV-7), and herpesvirus associated with Kaposi (HHV-8). In some embodiments, the gL protein of another herpesvirus is the gL protein of HSV1, HSV2, VZV, EBV, PrV, or bovine herpesvirus type 5. A potential benefit of substituting a CMV residue for a corresponding residue from another herpesvirus is that the secondary structure of the protease recognition site will probably be preserved. As shown in FIG. 1, HSV-1, HSV-2 and VZV all share substantially the same secondary structure, especially the C-terminal portion of the protease recognition sites adopts for all a β stranded structure. . If multiple substitutions are made, it is not necessary that they come from the same herpesvirus. For example, a first residue of CMV may be substituted by the corresponding residue of HSV-1, a second residue by the corresponding residue of HSV-2, and / or a third residue of CMV by the corresponding residue of VZV, etc. Therefore, the mutation may comprise a first amino acid residue substituted by a corresponding residue from a gL protein of a first other herpes virus, and a second amino acid residue substituted by a corresponding residue of a gL protein. a second herpesvirus, and / or a third amino acid residue substituted with a corresponding residue of a third herpesvirus protein, and the like. In some embodiments, the mutation comprises the substitution of E94. In certain embodiments, the mutation comprises substitution of A95 with R, L, or N. In some embodiments, the mutation comprises the substitution of A96 with a nonpolar residue or a residue that includes a large side chain, such as W, F, or M. In some embodiments, the mutation comprises the substitution of A96. by I, L, or S. In some embodiments, the mutation comprises substituting N97 for a polar or non-polar residue. The polar residue may comprise a small side chain or a large side chain. In some embodiments, the mutation comprises substituting N97 for S, D, E, A, or Y. In some embodiments, the mutation comprises substitution of S98 with an amino acid residue with a small side chain, such as G, A, V, S, T, C, D, or N. In some embodiments, the mutation includes the substitution of S98 by G, T, V, or I. In some embodiments, the mutation comprises the substitution of V99 by an amino acid residue I. In some embodiments, the mutation comprises substitution of "L100 with an amino acid residue F or V. In some embodiments, the mutation comprises the substitution of L101 with a V amino acid residue. The addition, deletion, and substitutions described herein may be used individually, or in any combination. For example, the mutant gL may include addition at one position, deletion at a second position, and substitution at a third position. In some embodiments, the gL protein or fragment comprises an insert region at the N-terminus of the protease recognition sign. As shown in Figure 1, compared to gL proteins of HSV-1, HSV-2, and VZV, the CMV gL protein comprises an additional 17-residue insert. As shown in the examples, when this 17-residue insert was partially or completely deleted, the gL protein became more prone to protease cleavage. Therefore, the 17-residue insert appears to at least partially block access of the proteases to the protease recognition site. Therefore, maintaining an insert region at the N-terminus of the protease recognition site may be desirable. An "insert region" should have a length of at least 10 amino acid residues, and is at least 50% identical to SEQ ID NO: 5 (which is the 17-residue fragment of single origin to CMV GL compared to HSV1, HSV2, and VZV). In some embodiments, the mutation comprises introducing a non-naturally occurring amino acid residue that is believed to reduce protease cleavage. In some embodiments, the mutation comprises introducing an amino acid residue comprising a bulky sidechain, which is believed to at least partially block access of the proteases to the protease recognition site, and reduce the cleavage by proteases. B. CMV-protein complexes In another aspect, the invention provides a complex comprising the modified CMV gL protein, or complex fragment thereof, described herein. Such complexes include, for example, (i) isolated dimeric complexes comprising: the modified gL protein, or a complex-forming fragment thereof, described herein, and CMV gH proteins or a fragment thereof a complex ; (ii) an isolated trimer complex comprising the modified CMV gL protein, or one of its complex-forming fragments, described herein, and CMV gH proteins or a complex-forming fragment thereof, and gO or one of their fragments forming a complex; and (iii) isolated pentameric complexes comprising the modified CMV gL protein, or complex-forming fragment thereof, described herein, and CMV pUL128 proteins or a complex-forming fragment thereof, pUL130 or one of their complex-forming fragments, pUL131 or one of their complex-forming fragments, and gH or one of their fragments forming a complex. It is also understood all other complexes comprising the gL (or one of its fragments forming a complex) as a component. Although gH, gL, gO, pUL128, pUL130, pUL131 may be referred to as glycoproteins, this nomenclature should not be taken to mean that these proteins must be glycosylated when used with the invention. In contrast, in some embodiments of the invention, one or more of the peptides are not glycosylated. However, usually one or more (or all) of the polypeptides in a complex of the invention are glycosylated. In some embodiments, one or more (or all) of the polypeptides in a complex of the invention are glycosylated by cultured cell glycosylation mutants, such as mutated mammalian cells. Such glycosylation mutants produce a polypeptide glycosylation profile that differs from a wild-type glycosylation pattern, i.e., the resulting glycoforms of the polypeptides differ from the wild-type glycoforms. In some embodiments, the glycosylation profile of gL (or a complex-forming fragment thereof), or a complex comprising gL (or a fragment thereof forming a complex) has a profile. mammalian glycosylation; and / or does not have an insect cell glycosylation profile. In some embodiments, one or more of the complex proteins contain complex N-linked side chains with penultimate-side galactose and terminal sialic acid. For recombinant production of protein complexes (such as a pentamer complex), it may be desirable for the complex to be soluble. Based on sequence analysis, the CMV gH protein comprises a transmembrane (TM) domain, but gL, gO, pUL128, pUL130, and pUL131 do not have a transmembrane domain. Also, to produce a soluble complex (eg, a pentamer complex), at the gH subunit of the pentamer complex, it may miss the TM domain. For example, a fragment of the gH comprising the N-terminal signal sequence and the ectodomain, but not the TM domain, of the gH can be used. The gH of the CMV Towne strain is represented by SEQ ID NO: 6 (GI: 138314, 742 amino acid residues). 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-terminal end (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 hydrophobic transmembrane (TM) domain (residues 718 to 736); and (v) a C-terminal cytoplasmic domain (residues 737 to 742). TM domains of gH proteins from other strains, or other variants and fragments of gH, can be identified according to sequence alignment. To facilitate production, the recombinantly produced CMV complex (such as a pentamer complex) can be secreted from the host cell into the culture medium. In some embodiments, said pentamer complex is secreted from the host cell. The presence of the five subunits, gH, gL, pUL128, pUL131, and pUL131, has been reported to be sufficient for assembly of the pentamer complex in ER before being exported to the Golgi apparatus. See, Ryckman et al., J Virol. Jan 2008; 82 (1): 60-70. 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 producing 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. C. Nucleic acid encoding modified gL proteins and complexes In another aspect, the invention provides a nucleic acid comprising a sequence that encodes the modified gL protein, or a complex fragment thereof, described herein. The nucleic acid may be DNA or RNA. In some embodiments, the nucleic acid is DNA. DNA expression systems for the expression and purification of recombinant proteins are well known in the art. For example, the expression system may be a vector comprising a nucleotide sequence that encodes the modified gL or fragment of the gL described herein, which is operably linked to an expression control sequence that regulates the expression. expressing the modified gL or fragment of the gL in a host cell, such as a mammalian host cell, a bacterial host cell, or an insect host cell. The expression control sequence may be a promoter, an enhancer, a ribosome entry site, or a polyadenylation sequence, for example. Other expression control sequences contemplated for use in the invention include introns and 3 'UTR sequences. The modified gL protein or fragment thereof, or a complex comprising the modified gL protein or a fragment thereof, expressed recombinantly can be purified using methods described herein, such as purification methods disclosed in US Pat. WO 2014/005959, or other methods known in the art. ' In some embodiments, the nucleic acid molecule is a vector derived from an adenovirus, an adeno-associated virus, a lentivirus, or an alphavirus. In some embodiments, the nucleic acid molecule is a deficient viral vector for replication. In some embodiments, the nucleic acid is RNA. In some embodiments, the nucleic acid is a self-replicating RNA molecule, such as an alphavirus-derived RNA replicon. Self-replicating RNA molecules are well known in the art and can be produced using replication elements derived from, for example, alphaviruses, and substituting the viral structural proteins with a nucleotide sequence coding for a protein of interest. One self-replicating RNA molecule is usually a positive strand molecule that can be translated directly after administration to a cell, and this translation provides an RNA-dependent RNA polymerase that then produces both antisense and sense transcripts from administered RNA. Thus, the administered RNA leads to the production of multiple daughter RNA molecules. These daughter RNA molecules, as well as colinear subgenomic transcripts, can be translated itself to provide in situ expression of an encoded antigen, or they can be transcribed to provide other transcripts with the same meaning as the Administered RNA, which are translated to provide in situ expression of the antigen. The overall result of this transcription sequence is an important amplification of the number of RNAs of the introduced replicons and thus, the coded antigen becomes a major polypeptide product of the cells. Cells transfected with self-replicating RNA briefly produce the antigen before undergoing apoptotic death. This death is the likely result of the required double-stranded RNA (db) intermediates, which have also been shown to overactivate dendritic cells. Thus, the enhanced immunogenicity of the self-replicating RNA may be a result of pro-inflammatory dsRNA production, which mimics an infection with an RNA virus of the host cells. An appropriate system for obtaining self-replication in this way is to use an alphavirus-based replicon. The alphaviruses comprise a set of viruses carried by genetically, structurally and serologically related arthropods of the family Togaviridae. Twenty-six known viruses and virus subtypes have been classified within the alphavirus genus, including Sindbis virus, Semliki forest virus, Ross river virus, and equine encephalitis virus. Venezuela. As such, the self-replicating RNA of the invention can incorporate a replicon RNA derived from Semliki Forest Virus (SFV), Sindbis Virus (SIN), Venezuelan Equine Encephalitis Virus (VEE). , Ross River Virus (RRV), Eastern Equine Encephalitis Virus, or other viruses belonging to the alphavirus family. An alphavirus-based replicon expression vector can be used in the invention. The replicon vectors can be used in a variety of formats, including DNA, RNA, and recombinant replicon particles. Such replicon vectors have been derived from alphaviruses which include, for example, Sindbis virus (Xiong et al (1989) Science 243: 1188-1191, Dubensky et al., (1996) J. Virol 70: 508). Hariharan et al., (1998) J. Virol 72: 950-958, Polo et al (1999) PNAS 96: 4598-4603), the Semliki forest virus (Liljestrom (1991) Bio / Technology 9 1356-1361, Berglund et al (1998) Nat Biotech 16: 562-565), and Venezuelan equine encephalitis virus (Pushko et al (1997) Virology 239: 389-401). Alphavirus-derived replicons are generally rather similar in their overall characteristics (eg, structure, replication), individual alphaviruses may have some particular property (eg, receptor binding, interferon sensitivity, and the pathology profile) which is unique. Therefore, chimeric alphavirus replicons made from divergent families of viruses may also be useful. In some embodiments, the CMV gL proteins (or fragments thereof) 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. Uses of alphavirus-based RNA replicon are known in the art, see, for example, WO 2013006837, paragraphs [0155] to [0179]. The RNA replicon can be administered without the need for purification of the protein encoded therein. In some embodiments, the nucleic acid molecule is part of a vector derived from an adenovirus. The adenovirus genome is an approximately 36,000 base pair linear double-stranded DNA molecule with the 55 kDa terminal protein covalently linked to the 5 'end of each strand. Adenoviral DNA ("Ad") contains identical inverted terminal repeats ("ITRs") of about 100 base pairs with the exact length dependent on the. serotype. The origins of viral replication are located within the ITRs exactly at the ends of the genomes. The adenoviral vectors for use with the present invention may be derived from any of the various adenoviral serotypes, including, without limitation, any of more than 40 serotypes of adenovirus strains, such as serotypes 2, 5 , 12, 40 and 41. In some embodiments, the nucleic acid molecule is part of a vector derived from an adeno-associated virus (AAV). The AAV genome is a linear double-stranded DNA molecule containing approximately 4681 nucleotides. The AAV genome generally comprises an internal non-repetitive genome flanked at each end of inverted terminal repeats (ITRs). The ITRs are approximately 145 base pairs (bp) in length. RTIs have multiple functions, including serving as origins of DNA replication and packaging signals for the viral genome. AAV is an auxiliary-dependent virus; that is, it requires co-infection with a helper virus (e.g., adenovirus, herpesvirus, or vaccinia virus) to form AAV virions in nature. In the absence of co-infection with a helper virus, AAV establishes a latent state in which the viral genome inserts into a chromosome of the host cell, but it is not produced from infectious virions. Subsequent infection with a helper virus releases the integrated genome, allowing it to replicate and encapsulate its genome in infectious AAV virions. While AAV can infect cells of different species, the helper virus must be of the same species as the host cell. Thus, for example, human AAV will replicate in canine cells co-infected with canine adenovirus. In some embodiments, the nucleic acid molecule is part of a vector derived from a retrovirus. A selected gene may be inserted into a vector and packaged into retroviral particles using techniques known in the art. The recombinant virus can then be isolated and administered to cells of the subject either in vivo or ex vivo. A number of retroviral systems have been described. See, for example, US Patent No. 5,219,740; Miller and Rosman (1989) BioTechniques 7: 980-90; Miller, A.D. (1990) Human Gene Therapy 1: 5-14; Scarpa et al. (1991) Virology 180: 849-52; Burns et al. (1993) Proc. Natl. Acad. Sci. USA 90: 8033-37; Boris-Lawrie and Temin (1993) Curr. Opin. Broom. Develop. 3: 102-09. The invention also provides host cells comprising the nucleic acid molecules disclosed herein. Host cells suitable for harboring the nucleic acid molecules and / or for expressing recombinant proteins, and methods for introducing a nucleic acid into a suitable host cell, are known in the art. 4. Recombinant Generation of gL Proteins and Complexes The invention also provides a host cell comprising nucleic acids encoding the gL protein and a fragment thereof as described above. Preferably, the host cells are mammalian cells (e.g., human, nonhuman primate, horse, cow, sheep, dog, cat, and rodent (eg, hamsters)), avian cells (e.g., chicken , duck, and goose). 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, 293-T cells, Vero cells, HeLa cells, PERC.6 cells (ECACC deposit number 96022940), Hel G2 cells, MRC-5 cells (ATCC CCL-171), · WI-38 (ATCC CCL-75), fetal Rh lung cells (ATCC CL-160), Madin-Darby bovine kidney cells ("MDBK"), Madin-Darby canine kidney cells ("MDCK") (e.g., 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 HEK-293 cell. In some embodiments, the host cell is a CHO cell. In some embodiments, the polynucleotide encoding the gL protein (or fragment thereof) described herein is integrated into the genomic DNA of the CHO cell. For the recombinant production of a CMV protein complex, the nucleotide sequence coding for other subunits of the complex will also be integrated into the genomic DNA of the CHO cell. ' Accordingly, in some embodiments, the host cell comprises one or more polynucleotide sequences encoding the CMV pentameric complex, said pentamer complex comprising: gH or one of its pentamer-forming fragments, gL or one of its pentamer-forming fragments, pUL128 or one of its pentamer-forming fragments, pUL130 or one of its pentamer-forming fragments, and pUL131 or one of its pentamer-forming fragments. In some embodiments, the one or more polynucleotide sequences encoding the CMV pentamer complex are integrated into the genomic DNA of said host cell. In some embodiments, the host cell, when cultured under appropriate conditions, expresses said CMV pentamer (which is preferably soluble and / or secreted from the host cell). Examples of CHO cell lines available from the European Collection of Cell Cultures (ECACC) are listed in Table 2. All CHO cells listed in Table 2 can be used. Table 2 Various CHO cell lines are also available from 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-K1 (ATCC® CCL-61 ™), Lecl [originally named Pro-5WgaR13C] (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 / 3 (ATCC® CRL-2143"), CHO -ICAM-1 (ATCC® CRL-2093 "), and pgsB-618 (ATCC® CRL-2241 ™). Any of these CHO cell lines can be used. Other commercially available CHO cell lines include, for example, FreeStyle ™ CHO-S cells and Life Technologies' Flp-In ™ cell line. Other suitable host cells include, for example, a CHO cell in which the level of expression or C12orf35 protein activity is reduced compared to a control (see, for example, WO 2015/092735, incorporated herein by reference). by reference, which provides a detailed description of mammalian cells in which the level of expression or activity of the C12orf35 protein is reduced compared to a control), a CHO cell in which the level of expression or the activity of the FAM60A protein is reduced compared to a control (see, for example, WO 2015/092737, incorporated herein by reference, which provides a detailed description of mammalian cells in which the level of expression or activity of the FAM60A protein is reduced); a CHO cell in which the level of expression or activity of the matriptase is reduced, compared to a control (provisional US patent application No. 61 / 985,589, filed April 29, 2014 and incorporated herein by reference), and Provisional US Patent Application No. 61/994 310, filed May 16, 2014 and incorporated herein by reference, provides a detailed description of mammalian cells in which the level of expression or activity of matriptase is reduced). Methods for the expression of recombinant proteins in CHO cells have generally been disclosed. See, for example, US Pat. Nos. 4,816,567 and 5,981,214. Patent Application EP EP14191385.5 filed October 31, 2014 (incorporated herein by reference) discloses mammalian host cells, in particular CHO cells, in which the sequence or sequences coding for the CMV proteins gH, gL, pUL128, pUL130, pUL131 (or one of their complex-forming fragments) are stably integrated into the genome. 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, at least about 20,000, at least about 25,000, at least about 30,000. 000 1, at least about 35,000, at least about 40,000, at least about 45,000, at least about 50,000, at least about 55,000, at least about 60,000, 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 CMV complex (such as the pentamer complex) is at least about 0.01 g / l, at least about 0.02 g / l, at least about 0, 0.3 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 / 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 / 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 less than 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 the cytomegalovirus (CMV) gL protein, or a fragment thereof, or a complex comprising said gL protein or fragment, comprising: (i) culturing the host cell described herein under suitable conditions, thereby expressing said gL protein, or a fragment thereof; and (ii) harvesting said gL protein, or a fragment thereof, or complex comprising said gL protein or fragment thereof from the culture. In some embodiments, the gL protein (or a fragment thereof), or the complex comprising said gL protein or a fragment thereof described herein is purified. The gL protein (or a fragment thereof) can be purified using any suitable method, such as HPLC, various types of chromatography (such as hydrophobic interactions, ion exchange, affinity, chelation, and size exclusion), electrophoresis, density gradient centrifugation, solvent extraction, or the like. For example, ion exchange may be used to purify the gL protein (or a fragment thereof), or a complex comprising said gL protein or fragment. Examples of materials useful in ion exchange chromatography include DEAE-cellulose, QAE-cellulose, DEAE-cephalose, QAE-cephalose, DEAE-Toyopearl, QAE-Toyopearl, Mono Q, Mono S, Q Sepharose, SP Sepharose etc. in one exemplary embodiment, the method uses a Mono S column. In another exemplary embodiment, the method uses a Mono Q column. Alternatively or additionally, affinity purification may be used. Examples of affinity purification markers include, for example, the His tag (binds to the metal ion), an antibody (binds to protein A or protein G),. maltose binding protein (MBP) (binds to amyloidosis), glutathione-S-transferase (GST) (binds to glutathione, FLAG marker (Asp-Tyr-Lys-Asp-Asp-Asp-Asp -Lys) (SEQ ID NO: 8) (binds to an anti-flag antibody), the Strep marker (binds to streptavidin or to one of its derivatives). An exemplary embodiment is the Strep marker (or streptavidin affinity tag), a marker that binds streptavidin or one of its derivatives, such as Strep-Tactin. The Strep marker comprises a peptide of nine amino acids: Ala-Trp-Arg-His-Pro-Gln-Phe-Gly-Gly (SEQ ID NO: 9), or eight amino acids (also called strep marker II): Trp -Ser- His-Pro-Gln-Phe-Glu-Lys (SEQ ID NO: 10). Elution of a protein attached to a strep marker from the column may be effected using biotin or a derivative or homologue thereof, such as desthiobiotin. The affinity purification marker may be set by any suitable means, and may be attached directly or indirectly. For example, the label may be covalently attached to the N-terminus of the polypeptide sequence, or to the C-terminus of the polypeptide sequence. This can be achieved by recombinant expression of a fusion protein comprising the polypeptide and the marker, or by conventional methods of conjugation that binds the polypeptide to the marker. The label may be attached to the side chain functional group of an amino acid residue of the polypeptide using standard conjugation techniques. Alternatively, the label may be noncovalently attached. Fixation of the marker can be direct or indirect (via a linker). Suitable linkers are known to those skilled in the art and include, for example, straight or branched chain carbon linkers, heterocyclic carbon linkers, carbohydrate linkers and polypeptide linkers. In one embodiment, cleavable linkers can be used to bind the molecule of interest to the label. This allows the label to be separated from the purified complex, for example, by the addition of an agent capable of cleaving the linker. A number of different cleavable linkers are known to those skilled in the art. Such linkers can be cleaved, for example, by irradiation of a photolabile bond or acid catalyzed hydrolysis. There are also polypeptide linkers that incorporate a protease recognition site and that can be cleaved by the addition of a suitable protease enzyme. When a complex comprising the gL protein (or a fragment thereof) is purified, the label may be attached to one or more other components of the complex. For example, during the purification of the CMV pentamer complex, a marker may be attached to pUL128, pUL130, or pUL131. 5. Pharmaceutical Compositions and Administration The invention also provides pharmaceutical compositions comprising the CMV proteins, complexes, and nucleic acids described herein. The invention also provides pharmaceutical compositions comprising the nucleic acid encoding the CMV proteins, complexes and nucleic acids described herein. The CMV proteins, complexes, and nucleic acids described herein may be incorporated into an immunogenic composition, or a vaccine composition. Such compositions can be used to raise antibodies in a mammal (eg, a human). The invention provides pharmaceutical compositions comprising the CMV proteins, complexes, and nucleic acids described herein, and methods of making a pharmaceutical composition involving the combination of the CMV proteins, complexes, and nucleic acids described herein. with a pharmaceutically acceptable carrier. The pharmaceutical compositions of the invention generally comprise a pharmaceutically acceptable carrier, and a full description of such carriers is available in Remington: The Science and Practice of Pharmacy. The pH of the composition is usually from about 4.5 to about 11, such as from about 5 to about 11, from about 5.5 to about 11, from about 6 to about 11, from about 5 to about 10.5, and about 5.5 and about 10.5, between about 6 and about 10.5, between about 5 and about 10, between about 5.5 and about 10, between about 6 and about 10, between about 5 and about 9.5 from about 5.5 to about 9.5, from about 6 to about 9.5, from about 5 to about 9, from about 5.5 to about 9, from about 6 to about 9, from about 5 to about 8.5 from about 5.5 to about 8.5, from about 6 to about 8.5, from about 5 to about 8, from about 5.5 to about 8, from about 6 to about 8, about 4.5, about 5, about 6.5, about 6, about 6.5, about 7, about 7.5, about 8, about 8.5, about 9, about 9.5, about 10, about 10.5, about 11, etc. A stable pH can be maintained by the use of a buffer, for example, a Tris buffer, a citrate buffer, a phosphate buffer, or a histidine buffer. Thus, a composition will generally include a buffer. A composition may be sterile and / or pyrogen-free. The compositions can be isotonic with respect to humans. A composition comprises an immunologically effective amount of its antigen (s). An "immunologically effective amount" is an amount that, when administered to a subject, is effective in eliciting an antibody response against the antigen. This amount may vary according to the health status and physical condition of the individual to be treated, his or her age, the ability of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the doctor's estimate of the medical situation, and other relevant factors. The quantity is expected to be in a relatively wide range which can be determined by routine testing. The antigen content of the compositions of the invention will generally be expressed in terms of protein mass per dose. A dose of 10 to 500 μg (e.g., 50 μg) per antigen may be useful. The immunogenic compositions may comprise an immunological adjuvant. Examples of adjuvants include compositions containing minerals; oily emulsions; saponin formulations; virosomes and pseudoviral particles; bacterial or microbial derivatives; bioadhesives and mucoadhesives; liposomes; polyoxyethylene ether and polyoxyethylene ester formulations; polyphosphazenes (pcpp); muramyl peptides; imidazoquinolone compounds; thiosemicarbazone compounds; tryptanthrin compounds; human immunomodulators; lipopeptides; benzonaphthyridines; microparticles; an immunostimulatory polynucleotide (such as RNA or DNA, eg oligonucleotides containing CpG). For example, the composition may comprise an aluminum salt adjuvant, an oil-in-water emulsion (for example, an oil-in-water emulsion comprising squalene, such as MF59 or ASO3), a TLR7 agonist (such as imidazoquinoline or Imiquimod), or one of their combinations. Suitable aluminum salts include hydroxides (eg, oxyhydroxides), phosphates (eg, hydroxyphosphates, orthophosphates), (e.g., see Chapters 8 and 9 of Vaccine Design (1995) eds Powell & Newman, ISBN: 030644867X, Plenum), or their mixtures. The salts may take any suitable form (e.g., gel, crystalline, amorphous, etc.), with the adsorption of the antigen on the salt being an example. The concentration of Al3 + in a composition for administration to a patient may be less than 5 mg / ml, for example, <4 mg / l, <3 mg / l, <2 mg / l, <1 mg / l, etc. . A preferred range is between 0.3 and 1 mg / ml. A maximum of 0.85 mg / dose is preferred. Adjuvants of aluminum hydroxide and aluminum phosphate are suitable for use with the invention. A suitable immunological adjuvant comprises a compound of formula (I) as defined in WO 2011/027222, or a pharmaceutically acceptable salt thereof, adsorbed on an aluminum salt. Many other adjuvants can be used, including any of those disclosed in Powell & Newman (1995). The compositions may include an antimicrobial, particularly when packaged in a multiple dose format. Antimicrobials such as thimerosal and 2-phenoxyethanol are commonly found in vaccines, but sometimes it may be desirable to use either a mercury-free preservative or no preservative at all. The compositions may comprise a detergent, for example, a polysorbate, such as polysorbate 80. Detergents are generally present at low levels, for example <0.01%. · The compositions may include sodium salts (e.g., sodium chloride) to provide tonicity. A concentration of 10 ± 2 mg / ml NaCl is typical, for example, about 9 mg / ml. 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 above. 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 (LTC) 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 epithelial cells and also fibroblast cells. In some embodiments, the immune response reduces or prevents infection of both 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 the antibody in the art is the "50% neutralization titer". To determine the 50% neutralization titer, serum from immunized animals is diluted to estimate how the 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 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. "Means more or less 10% of the quoted value. The compositions of the invention will generally be administered directly to a subject. Direct administration may be accomplished by parenteral injection (eg, subcutaneously, intraperitoneally, intravenously, intramuscularly, or into the interstitial space of tissue), or by any other appropriate route. For example, intramuscular administration may be used, for example, in the thigh or upper arm. Injection may be by needle (e.g., hypodermic needle), but needleless injection may be used alternatively. A typical intramuscular dosage volume is about 0.5 ml. The dosage may be by a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primoimmunization schedule and / or in a booster immunization schedule. In a multiple dose schedule, the various doses may be given by the same or different routes, for example parenteral sensitization and mucosal booster, mucosal sensitization and parenteral booster, etc. The multiple doses will generally be administered at an interval of at least 1 week (eg, about 2 weeks, about 3 weeks, about 4 weeks, about 6 weeks, about 8 weeks, about 10 weeks, about 12 weeks, about 16 weeks). weeks, etc.). The subject may be an animal, preferably a vertebrate, more preferably a mammal. An example of a subject includes, for example, a human being, a cow, a pig, a chicken, a cat or a dog, because the pathogens covered here can be problematic over a wide range of species. When the vaccine is for prophylactic use, the human being is preferably a child (eg, a young child or an infant), a teenager, or an adult; when the vaccine is for therapeutic use, the human being is preferably a teenager or an adult. A vaccine intended for children may also be administered to adults, for example, to estimate safety, dosage, immunogenicity, etc. The vaccines of the invention may be prophylactic (i.e., to prevent disease) or therapeutic (i.e., to reduce or eliminate the symptoms of a disease). The term prophylactic may be considered to reduce the severity or prevent the establishment of a particular condition. For the avoidance of doubt, the term prophylactic vaccine may also refer to vaccines that enhance the effects of a future infection, for example, by reducing the severity or duration of such an infection. . The isolated and / or purified CMV proteins, complexes, and nucleic acids described herein may be administered alone or in the form of sensitization or boosting in mixed-mode regimens, such as RNA sensitization followed by recall by the protein. The benefits of a protein-boosting RNA sensitization strategy, compared to a protein-boosting protein sensitization strategy, include, for example, an increase in antibody titers, a more balanced profile of IgG1 / IgG2a types, the induction of a CD4 + T-cell mediated Th1 immune response that was similar to that of viral particles, and the reduction of non-neutralizing antibody production. RNA sensitization can increase the immunogenicity of the compositions regardless of whether or not they contain an adjuvant. In the protein RNA sensitization strategy, the RNA and protein are directed against the same target antigen. Examples of suitable modes of RNA delivery include pseudoviral replicon (VRP) particles, alphavirus RNA, lipid nanoparticle-encapsulated replicons (LNP) or formulated RNAs, such as replicons formulated with nanoemulsions cationic (CNE). Suitable cationic water-in-water nanoemulsions are disclosed in WO 2012/006380, for example, comprising an oily core (e.g., comprising squalene) and a cationic lipid (e.g., DOTAP, DMTAP, DSTAP, DC- cholesterol, etc.). WO 2012/051211 discloses that antibodies directed against the pentamer complex are produced in mice that have been immunized with VRPs and formulated RNAs (CNE and LNP) that encode the protein constituents of the pentamer complex. These antibodies have been found to be capable of neutralizing CMV infection in epithelial cells. The protein-boosting RNA sensitization regime may involve performing first (eg, weeks 0-8) one or more RNA sensitization immunizations (which may be administered in the form of VRP, LNP, CNE, etc.) which encodes one or more of the protein components of a CMV protein complex of the invention and then performs one or more booster immunizations subsequently (e.g., at weeks 24 to 58) with: an isolated CMV protein complex of the invention, optionally formulated with a purified CMV adjuvant or protein complex of the invention, optionally formulated with an adjuvant. In certain embodiments, the RNA molecule is encapsulated in, bound to or adsorbed on a cationic lipid, a liposome, a cochleate, a virosome, an immunostimulatory complex, a microparticle, a microsphere, a nanosphere, a unilamellar vesicle, a multilamellar vesicle, an oil-in-water emulsion, a water-in-oil emulsion, an emulsome, a polycationic peptide, a cationic nanoemulsion, or combinations thereof. Also provided herein are kits for administering a nucleic acid (eg, RNA), purified proteins, and purified complexes described herein, and instructions for use. The invention also provides a pre-filled delivery device with a composition or vaccine described herein. The pharmaceutical compositions described herein may be administered in combination with one or more additional therapeutic agents. The additional therapeutic agents may include, but are not limited to, antibiotics or antibacterial agents, antiemetic agents, antifungal agents, anti-inflammatory agents, antiviral agents, immunomodulatory agents, cytokines, antidepressants, hormones, alkylating agents, antimetabolites, antitumor antibiotics, antimitotic agents, topoisomerase inhibitors, cytostatic agents, anti-invasion agents, antiangiogenic agents, growth factor function inhibitors, inhibitors of viral replication, viral enzyme inhibitors, anti-cancer agents, α interferons, β-interferons, ribavirin, hormones, and other Toll-like receptor modulators, immunoglobulins (Ig), and modulating antibodies the function of Ig (such as anti-IgE (omalizumab)). In some embodiments, the compositions disclosed herein may be used as a drug, for example, for use in inducing or enhancing an immune response in a subject in need, such as a mammal. In some embodiments, the compositions disclosed herein may be used in the manufacture of a medicament for the induction or enhancement of an immune response in a subject in need thereof, such as a mammal. One way of verifying the effectiveness of a therapeutic treatment involves monitoring of pathogen infection following administration of the compositions or vaccines described herein. Another way of verifying the effectiveness of prophylactic treatment involves monitoring immune responses, at the systemic level (such as monitoring the IgG1 and IgG2a production rate) and / or at the mucosal level (such as rate monitoring). IgA production) against the antigen. Generally, antigen specific serum antibody responses are determined after immunization but prior to challenge while antigen specific mucosal antibody responses are determined after immunization and after challenge. This invention is further illustrated by the following examples which should not be construed as limiting. Examples Example 1 - Materials and Methods Analysis of the sequence and the structure. The CMV, VZV, HSV1 and HSV2 gL sequences were aligned using CLUSTALW (https://npsa-prabi.ibcp.fr/cgi-bin/npsa_automat.pl page= npsa_clustalw.html) and manually adjusted to align residues that contribute to the conserved β strands in VZV and HSV2. Expression of the penta complex and gH / gL. Pentameric wild type (penta) complex (WT) or penta with mutations in gL (mutants "LSG" and "IDG") were expressed using a two vector system with gH and gL in one vector, and three ULs in the other. The sequence of the IRES (internal ribosome entry site) separates different genes in each vector. GH has a C-terminal 6xHis tag (SEQ ID NO: 11), and UL130 has a C-terminal cleavable strep tag. DNA from both vectors with 1 mg of total DNA for each liter of culture was transfected into Expi293 cells using the Expifectamine transfection kit (Life Technologies) following the manufacturer's protocol. Cells were grown to ~ 2.5 x 10 6 cells / ml on the day of transfection with viability> 97% in roller bottles. The transfected cells were cultured for three days up to ~ 8 x 10 6 cells / ml with viability of ~ 60% in an incubator with agitation operating at 37 ° C, 150 rpm and with 8% CO2. Supernatants from the expression media were harvested by centrifugation at 4200 rpm for 30 minutes. The gH / gL WT or gH / gL complexes with mutations in gL were expressed using the vector containing both gH and gL in the same manner as described above. N-terminal sequencing. N-terminal sequencing was used to identify unknown bands visible on SDS-PAGE and Western blot (WB) of affinity purified penta WT. Penta on SDS-PAGE was transferred to a depressed PVDF membrane. ethanol, which was stained with 0.02% Coomassie brilliant blue in 40% methanol and then washed in distilled water several times before complete air drying. The bands of interest were cut and shipped to Tufts University Protein Core Facility for sequencing. . Purification and Western blot analysis. The harvested supernatant was concentrated and the buffer was exchanged for affinity column binding buffer (50 mM Hepes pH 7.0, 150 mM NaCl, 1 mM EDTA) using a KrosFlo Research II system. TFF and a hollow fiber cartridge (Spectrumlabs). The concentrated supernatant was loaded onto an HP StrepTrap cartridge (GE Life Sciences), and eluted with elution buffer (50 mM Hepes pH 7.0, 150 mM NaCl, 2.5 mM desthiobiotin, and 1 mM EDTA). Peak fractions from the eluate were analyzed by SDS-PAGE and Western blot using antibodies directed against either the gL or the His marker placed at the C-terminus of the gH. Immunization studies in mice. Ten mice per group were immunized with pentamer gH / gL / pUL128 / pUL130 / pUL131 WT or mutant adjuvanted with MF59 at three different doses of 0.03 μg, 0.1 μg and 1 μg with three injections at intervals. three weeks. The serum samples were heat-inactivated at 56 ° C for 30 min, serially diluted in steps of 1/2 (two replicates per dilution), mixed with an equal volume of diluted HCMV virus to a target concentration of 200 250 infected cells / count field in medium ± 10% guinea pig complement (Cedarlane Labs, Burlington, NC, USA), and incubated for 2 h at 37 ° C / 5% CO2. These serum / virus samples were added to ARPE-19 cells or MRC-5 cells prepared in 96-well cell culture plates (Corning Inc., Corning, NY, USA). The infected monolayers were incubated for 48 hours (± 8 h) at 37 eC / 5% CO 2, fixed with 10% buffered formalin (EMD Chemicals Inc., Gibbstown, NJ, USA) for one hour and washed. three times with washing buffer (PBS / 0.05% Tween-20), blocked with PBS / 2.5% fetal bovine serum, 0.5% saponin, 0.1% sodium azide for one hour at room temperature. The plates were washed three times, tapped and incubated in a humid incubator at 25 ° C for one hour. The plates were then incubated for one hour at room temperature with anti-HCMV IE1 antibody derived from L14 hybridoma (diluted in saponin buffer). The plates were washed three times and incubated for one hour with anti-mouse IgG conjugated with AlexaFluor 488 reagent (diluted in saponin buffer), and then washed three times with PBS / 0.05% Tween-20. . Fluorescent cells were counted using an Immunospot S5 UV Analyzer instrument (Cellular Technology Limited, Shaker Heights, OH, USA), and the 50% neutralization titer, defined as the reciprocal of serum dilution producing 50% reduction in the number of infected cells (relative to the number of infected cells in the diluent plus virus control wells), was calculated by interpolation of the linear regression between the two dilutions with producing wells. average numbers of infected cells above and below the 50% value. Example 2 - Results 1. The cut of the gL occurs near a strand β conserved N-terminal sequencing determined that a band identified by Western blotting using an anti-gL antibody starts with residue 97 of gL. Thus, the pentameric complex gH / gL / pUL128 / pUL130 / pUL131 expressed in mammalian cells contains a population of gL proteins cut between the Asn97 and Ser98 residues of gL. The sequence alignment based on the structure further disclosed that the cleavage site is in a loop region near a β strand conserved in the gH / gL structures of both VZV and HSV-2 (FIG. 1). Various mutations introduced into the gL sequence near this cleavage site resulted in a reduction in the cleavage amount of the G1. Addition mutations inserted between two and five residues with a mixture of polar and non-polar residues in the cleavage site. Deletion mutations deleted one to three residues around the cleavage site. Substitution mutations have changed Ala96 to hydrophobic residues or residues with large side chains; changed Asn97 to polar residues with either smaller or larger side chains, or non-polar residues; or have changed Ser98 into residues with small side chains having either polar or non-polar character. 2. Comparison of various mutant gH / gLs with wild-type gH / gL Figure 2A shows various mutant gL proteins that were tested. The gH / gL complexes containing various mutations of the gL were expressed in Expi293 cells and compared to the gH / gL WT. Mutations at the protease recognition site reduced the cleavage of the gL in the expressed gH / gL complexes. For example, anti-gL Western blot analysis of crude WT supernatant showed a clearly visible band of a fragment of gL with residue 98 at its N-terminus, determined by N-terminal sequencing. In contrast, a similar band was not detected in the "LSG" mutant and either was not detected or was significantly reduced in the "delta Asn97" and "SST" mutants (Figure 2). The three tailings variants introduced near the cleavage site reduced the cleavage to a greater extent than a single residue variant near the cleavage site. The "LSG" mutant reduced the intensity of the gL cutoff band most significantly in an anti-His Western blot. In addition, removal of the 17-residue insert amplified the intensity of the cutoff band of the gl observed by Western blot analysis, gH / gL (N), suggesting that this insertion may protect the cleavage site (Figure 2C). 3. Comparison of the "LSG" and "IDG" penta mutants with the wild-type pentamer To analyze whether the "LSG" and "IDG" mutants also eliminated or reduced the cleavage of gL in the pentamer gH / gL / pUL128 / pUL130 / pUL131, WT pentamers and affinity purified mutants were analyzed by Western blots. anti-His and anti-gL. In the anti-His western blot of pentamere WT, there is a pronounced band with a smaller molecular weight than full-length gH / gL, consistent with a complex of gH and the N-terminal region of the gL after cleavage. . Note that this N-terminal fragment of the gL is not recognized by the anti-gL antibody. In the anti-gL Western blot, the C-terminal region of gL, beginning with residue 98 determined by N-terminal sequencing, forms a complex with UL128 in an unreduced sample. The same C-terminal fragment of gL by itself was observed in a reduced sample. In comparison, these bands resulting from gL cleavage were not detected in either the "LSG" or "IDG" mutant pentamers (Figures 3 and 4). Both "LSG" and "IDG" mutants produced a pentamer that behaved similarly to the pentameric WT complex. Therefore, these mutations do not affect the assembly of the pentamer complex gH / gL / pUL128 / pUL130 / pUL131, but they eliminated the proteolytic cleavage of the gL protein (FIG. 5A). Immunogenicity analysis showed that the LSG and IDG mutants did not compromise the immunogenicity of the pentameric gH / gL / pUL128 / pUL130 / pUL131 complex (Figure 5B). The fragment of Ig resulting from the cleavage was detected during the expression of gH / gLf gH / gL / gO complexes (data not shown) and pentamer gH / gL / pUL128 / pUL130 / p1L131. The cleavage sites in these three complexes are identical between the residues of gL 97 and 98. Therefore, mutations that prevent cleavage of gL during expression of gH / gL also prevent cleavage of gL during treatment. expression of the gH / gL / gO and pentamer gH / gL / pUL128 / pUL130 / pUL131 complexes. With as few as three substitutions of residues or a single deletion, the cleavage of the gL can, respectively, be eliminated or significantly reduced. The location of these mutations is not expected to affect the secondary structure conserved in their vicinity. This allows the production of a homogeneous pentamer gH / gL / pUL128 / pUL130 / pUL131 with its three-dimensional structure, and its largely unaffected antigenicity / immunogenicity. We conclude that the mutation strategy of the sequence near the cleavage site with a homologous sequence has been shown to be effective. 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 publications and patents 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. The practice of the present invention will employ, unless otherwise indicated, conventional methods of chemistry, biochemistry, molecular biology, immunology and pharmacology within the skill of the art. Such techniques are fully explained in the literature. The term "comprising" includes "including" as well as "consisting of", for example, a composition "comprising" X may consist exclusively of X or it may include something additional, for example, X + Y. The term "essentially consisting of" means that the composition, process or structure may include additional components, steps and / or parts, but only if the additional components, steps and / or parts do not materially alter the basic and novel features of the claimed composition, process or structure. The term "constituted of" is generally taken to mean that the invention as claimed is limited to those elements specifically mentioned in the claim (and that it may include their equivalents, as long as the doctrine of equivalents is applicable). 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 cytomegalovirus (CMV) gL protein, or a fragment thereof forming a complex, where. said gL protein or said fragment comprises a mutation within the protease recognition site, wherein said mutation reduces protease cleavage at said protease recognition site compared to a control. 2. The gL protein or fragment of Embodiment 1, wherein said mutation comprises adding, deleting, substituting, or one of their combinations, an amino acid residue. 3. The gL protein or the fragment of Embodiment 1 or 2, where three or more residues of said protease recognition site form a β-strand, and said mutation maintains β-strand conformation. 4. The gL protein or fragment of any one of embodiments 1 to 3, wherein said mutation results in not more than 20% (molar percentage) of the gL being cleaved at said protease recognition site. during recombinant expression in a mammalian host cell. 5. The gL protein or fragment of any one of embodiments 1 to 4, wherein said mutation comprises the addition of one or more amino acid residues. 6. The gL protein or fragment of any one of embodiments 1 to 5, wherein said mutation comprises the addition of two to five amino acid residues. 7. The gL protein or fragment of Embodiment 6, wherein said two to five amino acid residues comprise both one or more polar residues and one or more non-polar residues. 8. The gL protein or fragment of any one of embodiments 1 to 7, wherein said mutation comprises adding one or more residues between residues N97 and S98. 9. The gL protein or fragment of any one of embodiments 1 to 8, wherein said mutation comprises adding F, Q, FQ or QF between residues N97 and S98. The gL protein or fragment of any one of embodiments 1 to 9, wherein said mutation comprises deletion of one or more amino acid residues. 11. The gL protein or fragment of any one of embodiments 1 to 10, wherein said mutation comprises deletion of one to three amino acid residues. 12. The gL protein or the fragment of any one of embodiments 1 to 11, wherein said mutation comprises the deletion of a residue selected from the group consisting of: V91, T92, P93, E94, A95, A96, N97, S98, V99, L100, L101, D102 and one of their combinations. 13. The gL protein or fragment of any one of embodiments 1 to 12, wherein said mutation comprises the deletion of a residue selected from the group consisting of: A95, A96, N97, and one of their combinations. 14. The gL protein or fragment of any one of embodiments 1 to 13, wherein said mutation comprises N97 deletion. The gL protein or fragment of any one of embodiments 1 to 14, wherein said mutation comprises substitution of a residue with a corresponding residue from a gL protein of another herpes virus. 16. The gL protein or the fragment of embodiment 15, wherein said gp protein of another herpesvirus is a gL protein of HSV1, HSV2, VZV, EBV, PrV, or bovine herpesvirus. of type 5. 17. The gL protein or fragment of any one of embodiments 1 to 16, wherein said mutation comprises the substitution of A96 with a non-polar residue or a residue that comprises a large side chain. 18. The gL protein or the fragment of any one of embodiments 1 to 17, wherein said mutation comprises the substitution of A96 with I, L, V or S. 19. The gL protein or the fragment of one any of embodiments 1 to 18, wherein said mutation comprises substitution of A95 by R, L, E or N. 20. The gL protein or fragment of any one of embodiments 1 to 19, wherein said mutation comprises the substitution of E94 for A or L. 21. The gL protein or fragment of any one of embodiments 1 to 21, wherein said mutation comprises substituting N97 for a polar or non-polar residue. 22. The gL protein or fragment of Embodiment 21, wherein said polar residue comprises a small side chain. 23. The gL protein or fragment of Embodiment 21, wherein said polar residue comprises a large side chain. 24. The gL protein or fragment of any one of embodiments 1 to 23, wherein said mutation comprises substitution of N97 by S, D, E, A, T or Y. The gL protein or fragment of any one of embodiments 1 to 24, wherein said mutation comprises substitution of N97 by S or D. 26. The gL protein or fragment of any one of embodiments 1 to 25, wherein said mutation comprises the substitution of S98 with an amino acid residue with a small side chain. 27. The gL protein or fragment of any one of Embodiments 1 to 26, wherein said mutation comprises substitution of S98 by G, T, V, or I. 28. The gL protein or fragment thereof any one of embodiments 1 to 27, wherein said mutation comprises substituting S98 with G, or T. 29. The gL protein or fragment of any one of embodiments 1 to 28, wherein said mutation comprises the substitution of V99 with I. The gL protein or fragment of any one of Embodiments 1 at 29, where said mutation comprises the substitution of L100 by F or V31 amino acid residue. The gL protein or fragment of any one of embodiments 1 to 30, wherein said mutation comprises the substitution of L101 by an amino acid residue V or I. 32. The gL protein or fragment of any one of embodiments 1 to 31, wherein said gL protein or fragment comprises an insert region at the N-terminus of the protease recognition site. 33. The gL protein or fragment of any one of embodiments 1 to 32, wherein said mutation comprises introducing a non-naturally occurring amino acid residue. 34. The gL protein or fragment of any one of embodiments 1 to 33, wherein said mutation comprises introducing an amino acid residue comprising a bulky sidechain. 35. A CMV complex comprising the recombinant gL protein or fragment of any one of embodiments 1 to 34. 36. The complex of embodiment 35, comprising a CMV protein selected from the group consisting of gH , gL, pUL128, pUL130, pUL131, gO, one of their complex-forming fragments, and one of their combinations. 37. The complex of embodiment 35 or 36, wherein said complex is a pentamer complex comprising: gH or one of its pentamer-forming fragments, gL or one of its pentamer-forming fragments, pUL128 or one of its pentamer-forming fragments, pUL130 or one of its pentamer-forming fragments, and pUL131 or one of its pentamer-forming fragments. 38. The complex of embodiment 35 or 36, wherein said complex is a gH / gL complex comprising: gH or one of its complex-forming fragments, and gL or one of its fragments forming a complex. 39. The complex of embodiment 35 or 36, wherein said complex is a trimer complex comprising: gH or one of its complex-forming fragments, gL or one of its fragments forming a complex, and gO or one of its fragments forming a complex. 40. An immunogenic composition comprising the CMV recombinant gL protein or a fragment of any one of embodiments 1-34, or the complex of any one of embodiments 35-39. 41. The immunogenic composition of Embodiment 40, further comprising an adjuvant. 42. The immunogenic composition of embodiment 41, wherein said adjuvant comprises an aluminum salt, a TLR7 agonist, an oil-in-water emulsion, or a combination thereof. 43. The immunogenic composition of embodiment 42, wherein said oil-in-water emulsion is MF59. 44. An isolated nucleic acid comprising a polynucleotide sequence encoding the recombinant CMV gL protein or a fragment of any of embodiments 1-34. 45. The isolated nucleic acid of embodiment 44, wherein said acid is isolated nucleic acid is an RNA, preferably a self-replicating RNA. 46. The isolated nucleic acid of embodiment 45, wherein said self-replicating RNA is an alphavirus replicon. 47. An alphavirus replication particle (VRP) comprising the alphavirus replicon of embodiment 46. 48. An immunogenic composition comprising the nucleic acid of any one of embodiments 44 to 46. 49. A An immunogenic composition comprising the VRP of Embodiment 47. 50. The immunogenic composition of Embodiment 48 or 49, further comprising an adjuvant. 51. The immunogenic composition of Embodiment 50, wherein said adjuvant comprises an aluminum salt, a TLR7 agonist, an oil-in-water emulsion (such as MF59), or a combination thereof. 52. A host cell comprising the nucleic acid of any of embodiments 44 to 46. 53. The host cell of embodiment 52, wherein said nucleic acid is a DNA. 54. The host cell of Embodiment 53, wherein said host cell is a mammalian cell. 55. The host cell of embodiment 54, wherein said mammalian cell is a CHO cell or HEK-293 cell. 56. The host cell of any of embodiments 53 to 55, wherein said DNA encoding the CMV gL protein or fragment thereof is integrated into the genomic DNA of said host cell. 57. The host cell of any one of embodiments 52 to 56, wherein said host cell comprises one or more polynucleotide sequences encoding the CMV pentamer complex, said CMV pentameric complex comprising: gH or one of its pentamer-forming fragments, gL or one of its pentamer-forming fragments, pUL128 or one of its pentamer-forming fragments, pUL130 or a pentamer-forming fragment thereof, and pUL131 or one of its fragments forming a pentamer. 58. The host cell of Embodiment 57, wherein said one or more polynucleotide sequences encoding the CMV pentamer complex are integrated into the genomic DNA of said host cell. . 59. The host cell of embodiment 57 or 58, wherein said cell, when cultured under appropriate conditions, expresses said pentameric CMV complex. 60. The host cell of Embodiment 59, wherein said pentamer complex is secreted. 61. A cell culture comprising the host cell of any one of embodiments 52 to 60, wherein said culture has a size of at least 20 liters. 62. A cell culture comprising the host cell of any one of embodiments 52 to 60, wherein said culture has a size of at least 100 liters. 63. A cell culture comprising the host cell of any one of embodiments 57 to 60, wherein the yield of said pentamer complex is at least 0.05 g / l. 64. A cell culture comprising the host cell of Embodiment 63, wherein the yield of said pentamer complex is at least 0.1 g / l. 65. A method of producing a recombinant cytomegalovirus (CMV) gL protein, or complex fragment thereof, comprising: (i) culturing the host cell of any of the modes of embodiment 52-60 under appropriate conditions, thereby expressing said gL protein, or a fragment thereof forming a complex; and (ii) harvesting said gL protein, or a complex fragment thereof, from the culture. 66. 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 one of embodiments 43 and 48-51. 67. The method of embodiment 66, wherein the immune response comprises the production of neutralizing antibodies against CMV. 68. The method of embodiment 67, wherein the neutralizing antibodies are complement-independent. 69. 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 40 to 43 and 48 to 51. The immunogenic composition of any of embodiments 40 to 43 and 48 to 51 for use in inducing an immune response against cytomegalovirus (CMV). 71. Use of the immunogenic composition of any of embodiments 40 to 43 and 48 to 51 for the induction of an immune response against cytomegalovirus (CMV). 72. Use of the immunogenic composition of any of embodiments 40 to 43 and 48 to 51 in the manufacture of a medicament for inducing an immune response against cytomegalovirus (CMV). ·
权利要求:
Claims (15) [1] 1. Cytomegalovirus (CMV) recombinant gL protein, or a fragment thereof forming a complex, wherein said gL protein or said fragment comprises a mutation within the protease recognition site, wherein said mutation reduces protease cleavage at said protease recognition site compared to a control. [2] 2. The G1 protein or fragment of claim 1, wherein said mutation comprises adding, deleting, substituting, or one of their combinations, an amino acid residue. [3] 3. The gL protein or fragment according to claim 1 or 2, wherein the residues of said protease recognition site form a β-strand, and said mutation maintains the β-strand conformation. [4] 4. The gL protein or fragment according to any one of claims 1 to 3, wherein said mutation results in not more than 20% (molar percentage) of the gL being cleaved at said protease recognition site at a time. recombinant expression in a mammalian host cell. [5] 5. The G1 protein or fragment of any one of claims 1 to 4, wherein said mutation comprises the addition of two to five amino acid residues. [6] The gL protein or fragment of any one of claims 1 to 5, wherein said mutation comprises deletion of one to three amino acid residues. [7] 7. The G1 protein or fragment of any one of claims 1 to 6, wherein said mutation comprises substituting a residue with a corresponding residue from a gL protein of another herpes virus. [8] 8. A gL protein or fragment according to claim 7, wherein said gp protein of another herpesvirus is a gL protein of HSV1, HSV2, VZV, EBV, PrV, or bovine herpesvirus type 5. . [9] 9. A CMV complex comprising the recombinant gL protein or fragment according to any one of claims 1 to 8. [10] 10. Complex according to claim 9, wherein said complex is a pentamer complex comprising: gH or one of its pentamer-forming fragments, gL or one of its pentamer-forming fragments, pUL128 or one of its pentamer-forming fragments, pUL130 or one of its pentamer-forming fragments, and pUL131 or one of its pentamer-forming fragments. [11] An isolated nucleic acid comprising a polynucleotide sequence encoding the CMV recombinant gL protein or fragment according to any one of claims 1 to 8. [12] A host cell, preferably a mammalian host cell, comprising the nucleic acid of claim 11. [13] The host cell of claim 12, wherein said host cell comprises one or more polynucleotide sequences encoding the CMV pentameric complex, said CMV pentameric complex comprising: gH or one of its pentamer-forming fragments, gL or one of its pentamer-forming fragments, pUL128 or one of its pentamer-forming fragments, pUL130 or one of its pentamer-forming fragments, and pUL131 or one of its pentamer-forming fragments. [14] 14. An immunogenic composition comprising the recombinant cytomegalovirus gL protein or a fragment according to any one of claims 1 to 8, wherein the complex according to any of claims 9 to 10, and optionally comprising an adjuvant. [15] 15. An immunogenic composition according to claim 14 for use in inducing an immune response against cytomegalovirus.
类似技术:
公开号 | 公开日 | 专利标题 BE1023087B1|2016-11-18|ANTIGENS OF CYTOMEGALOVIRUS AND USES THEREOF CA2718868C|2017-03-14|Polynucleotides and chimaera polypeptides for releasing a polypeptide of interest combined with exosomes and use thereof for the production of immunogenic compositions EP2961846B1|2018-01-10|Crimean-congo haemorrhagic fever virus antigenic composition KR20160055164A|2016-05-17|Immunogenic Middle East Respiratory Syndrome Coronavirus | Compositions and Methods BE1023213B1|2016-12-21|COMPLEXES FROM CYTOMEGALOVIRUS AND USES THEREOF JPH09501933A|1997-02-25|Attenuation of immunodominant epitopes on antibodies useful as vaccines for plants, animals, and humans and for immunotherapy US20100047277A1|2010-02-25|Virosomes, methods of preparation, and immunogenic compositions WO2010063685A1|2010-06-10|Vaccine KR20180127402A|2018-11-28|Methods for preparing viral particles comprising cyclic dinucleotide and uses of said particles for cancer treatment CN113666990A|2021-11-19|T cell vaccine immunogen for inducing broad-spectrum anti-coronavirus and application thereof KR20190129032A|2019-11-19|Influenza vaccine CA2486282C|2012-04-10|Virus-like particles, methods of preparation, and immunogenic compositions TW202206598A|2022-02-16|A vaccine against sars-cov-2 and preparation thereof US20210261647A1|2021-08-26|Lamp constructs comprising allergens KR20150074714A|2015-07-02|Infectious clone comprising full-length nucleotide of porcine epidemic diarrhea virus FR2799974A1|2001-04-27|New complex of human immune deficiency virus envelope glycoprotein with CD4, useful as immunogen for vaccines, is deglycosylated enzymatically and generates neutralizing antibodies CN113151184A|2021-07-23|Method for cell membrane-based display of coronavirus immunogens to induce neutralizing antibodies WO2021214703A1|2021-10-28|A vaccine against sars-cov-2 and preparation thereof Andreasson2009|Vaccination against Her2/neu-expressing cancer using chimeric virus-like particles
同族专利:
公开号 | 公开日 US20190276498A1|2019-09-12| AU2018226521B2|2019-10-31| CA2974041A1|2016-07-28| EP3048114A1|2016-07-27| CN107531761A|2018-01-02| CN107531761B|2022-01-14| AU2016210548B2|2018-11-08| WO2016116904A1|2016-07-28| MX2017009538A|2017-11-02| JP2018504117A|2018-02-15| US20170369532A1|2017-12-28| KR20170100039A|2017-09-01| US10167321B2|2019-01-01| AU2018226521A1|2018-09-27| IL253366D0|2017-09-28| SG11201705740UA|2017-08-30| EP3247722A1|2017-11-29| EA038250B1|2021-07-29| ZA201704912B|2018-12-19| JP6717836B2|2020-07-08| AU2016210548A1|2017-08-10| BE1023087A1|2016-11-18| IL253366A|2022-03-01| EA201791562A1|2018-04-30| BR112017015567A2|2018-03-13|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 WO2014005959A1|2012-07-06|2014-01-09|Novartis Ag|Complexes of cytomegalovirus proteins| US4816567A|1983-04-08|1989-03-28|Genentech, Inc.|Recombinant immunoglobin preparations| US5219740A|1987-02-13|1993-06-15|Fred Hutchinson Cancer Research Center|Retroviral gene transfer into diploid fibroblasts for gene therapy| DK0747485T3|1989-11-06|1999-08-16|Cell Genesys Inc|Preparation of proteins using homologous recombination| EP3031469A1|2006-06-07|2016-06-15|The Trustees Of Princeton University|Cytomegalovirus surface protein complex for use in vaccines and as a drug target| MX343490B|2008-07-16|2016-11-08|Inst For Res In Biomedicine|Human cytomegalovirus neutralizing antibodies and use thereof.| WO2011027222A2|2009-09-02|2011-03-10|Novartis Ag|Immunogenic compositions including tlr activity modulators| NZ606591A|2010-07-06|2015-02-27|Novartis Ag|Cationic oil-in-water emulsions| RU2597974C2|2010-10-11|2016-09-20|Новартис Аг|Antigen delivery platforms| RU2649133C2|2011-07-06|2018-03-29|Новартис Аг|Cationic oil-in-water emulsions| TWI570240B|2011-09-09|2017-02-11|默沙東公司|A conditional replicating cytomegalovirus as a vaccine for cmv| BR112015008930A2|2012-10-30|2017-11-21|Redvax Gmbh|recombinant particle-based vaccines against human cytomegalovirus infection| PL3083676T3|2013-12-20|2020-02-28|Novartis Ag|Novel eukaryotic cells and methods for recombinantly expressing a product of interest| WO2015092737A1|2013-12-20|2015-06-25|Novartis Ag|Novel eukaryotic cells and methods for recombinantly expressing a product of interest| EP3015475A1|2014-10-31|2016-05-04|Novartis AG|Mammalian cells expressing cytomegalovirus antigens| EP3047856A1|2015-01-23|2016-07-27|Novartis AG|Cmv antigens and uses thereof|CA2798136C|2010-05-14|2019-06-04|Oregon Health & Science University|Recombinant hcmv and rhcmv vectors and uses thereof| SI2691530T1|2011-06-10|2018-08-31|Oregon Health & Science University|Cmv glycoproteins and recombinant vectors| WO2015048744A2|2013-09-30|2015-04-02|Moderna Therapeutics, Inc.|Polynucleotides encoding immune modulating polypeptides| UA120938C2|2014-07-16|2020-03-10|Орегон Хелс Енд Сайєнс Юніверсіті|Human cytomegalovirus comprising exogenous antigens| WO2016130693A1|2015-02-10|2016-08-18|Oregon Health & Science University|Methods and compositions useful in generating non canonical cd8+ t cell responses| WO2017015463A2|2015-07-21|2017-01-26|Modernatx, Inc.|Infectious disease vaccines| CA3002922A1|2015-10-22|2017-04-27|Modernatx, Inc.|Human cytomegalovirus vaccine| EP3364950A4|2015-10-22|2019-10-23|ModernaTX, Inc.|Tropical disease vaccines| AU2016355468A1|2015-11-20|2018-05-31|Oregon Health & Science University|CMV vectors comprising microRNA recognition elements| WO2017152146A2|2016-03-03|2017-09-08|Duke University|Compositions and methods for inducing hiv-1 antibodies| CA3039089A1|2016-10-03|2018-04-12|Duke University|Methods to identify immunogens by targeting improbable mutations| CR20190205A|2016-10-18|2020-01-07|Univ Oregon Health & Science|Cytomegalovirus vectors eliciting t cells restricted by major histocompatibility complex e molecules| CA3041307A1|2016-10-21|2018-04-26|Giuseppe Ciaramella|Human cytomegalovirus vaccine| WO2018151816A1|2017-02-16|2018-08-23|Modernatx, Inc.|High potency immunogenic compositions| CA3060019A1|2017-04-19|2018-10-25|Glaxosmithkline Biologicals Sa|Modified cytomegalovirus proteins and stabilized complexes| EP3681514A4|2017-09-14|2021-07-14|ModernaTX, Inc.|Zika virus rna vaccines| CN112088014A|2018-02-28|2020-12-15|华盛顿大学|Self-assembling nanostructured vaccines| BR112021006812A2|2018-10-17|2021-07-20|Glaxosmithkline Biologicals S.A.|stabilized proteins and modified cytomegalovirus complexes| CN109627330A|2018-12-18|2019-04-16|马鞍山史记动物健康管理有限公司|A kind of porcine pseudorabies virus high-titer positive serum preparation method| WO2021014385A1|2019-07-24|2021-01-28|Glaxosmithkline Biologicals Sa|Modified human cytomegalovirus proteins|
法律状态:
2019-10-17| MM| Lapsed because of non-payment of the annual fee|Effective date: 20190131 |
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 EP15152221.6|2015-01-22| EP15152221.6A|EP3048114A1|2015-01-22|2015-01-22|Cytomegalovirus antigens and uses thereof| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|