The Sequence Listing associated with this application is provided in text format in lieu of a paper copy, and is hereby incorporated by reference into the specification. The name of the XML file containing the Sequence Listing is 930485_436WO_SequenceListing.xml. The text file is 99,639 bytes, was created on Aug. 26, 2022, and is being submitted electronically via EFS-Web.
Vaccine vectors based on Cytomegalovirus (CMV) exploit the natural ability of this virus to elicit and maintain circulating and tissue resident effector-differentiated T cells, including the potential sites of early HIV infection. For example, rhesus CMV (RhCMV) vectors encoding simian immunodeficiency virus (SIV) antigen inserts can (1) superinfect RhCMV-immune primates and elicit high frequency effector-differentiated, SIV-specific CD4+ and CD8+ T cells in both lymphoid and organ tissues, (2) maintain these responses indefinitely, and (3) manifest early stringent control and ultimate clearance of infection with the highly pathogenic SIVmac239 strain. Current CMV manufacturing processes are limited in production and not directly scalable. Deletion of essential viral genes from vaccine vectors is a customary practice to ensure clinical safety. However, to produce a vector with an essential gene deletion, some method of gene complementation must be employed. Standard approaches involve creating stable cell lines that express the essential viral gene or its functional equivalent, however production of HCMV is complicated by the fact that it requires primary normal diploid cells for virus production. Accordingly, there remains a need for manufacturing methods capable of generating CMV vector-based vaccines that can produce vaccine in amounts necessary for clinical and commercial use. Described herein is an approach that utilizes mRNA transfection to deliver an essential viral gene to the host cell for scalable production of CMV vectors.
In certain aspects, the present disclosure provides a method of producing a CMV viral vector, comprising: (a) introducing a mRNA molecule encoding a pp71 protein to a cell (e.g., a MRC-5 cell); (b) infecting the cell with a CMV; (c) incubating the cell; and (d) collecting the CMV viral vector.
In certain aspects, the present disclosure also provides a CMV viral vector produced by any of the aforementioned methods.
The instant disclosure provides methods for producing a CMV vector, by providing complementation of an essential viral gene that has been deleted in the CMV vector, through transfection of mRNA encoding the missing protein into the host cell.
The following sections provide a detailed description of methods for producing CMV vectors. Prior to setting forth this disclosure in more detail, it may be helpful to an understanding thereof to provide definitions of certain terms to be used herein. Additional definitions are set forth throughout this disclosure.
In the present description, the term “about” or “approximately” means±20% of the indicated range, value, or structure, unless otherwise indicated.
The term “comprise” means the presence of the stated features, integers, steps, or components as referred to in the claims, but that it does not preclude the presence or addition of one or more other features, integers, steps, components, or groups thereof. The term “consisting essentially of” limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristics of the claimed invention.
It should be understood that the terms “a” and “an” as used herein refer to “one or more” of the enumerated components. The use of the alternative (e.g., “or”) should be understood to mean either one, both, or any combination thereof of the alternatives, and may be used synonymously with “and/or”. As used herein, the terms “include” and “have” are used synonymously, which terms and variants thereof are intended to be construed as non-limiting.
The word “substantially” does not exclude “completely”; e.g., a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from definitions provided herein.
The terms “nucleotide sequences” and “nucleic acid sequences” refer to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) sequences, including, without limitation, messenger RNA (mRNA), DNA/RNA hybrids, or synthetic nucleic acids. The nucleic acid may be single-stranded, or partially or completely double stranded (duplex). Duplex nucleic acids may be homoduplex or heteroduplex.
Nucleic acid molecules of particular sequence can be incorporated into a vector that is then introduced into a host cell, thereby producing a transformed host cell. A vector may include nucleic acid sequences that permit it to replicate in a host cell, such as an origin of replication. A vector may also include one or more selectable marker genes and other genetic elements known in the art, including promoter elements that direct nucleic acid expression. Vectors can be viral vectors, such as CMV vectors. Viral vectors may be constructed from wild type or attenuated virus, including replication deficient virus.
As used herein, the term “messenger RNA” (mRNA) refers to any polynucleotide which encodes at least one peptide or polypeptide of interest and which is capable of being translated to produce the encoded peptide polypeptide of interest in vitro, in vivo, in situ, or ex vivo. An mRNA may be transcribed from a DNA sequence by an RNA polymerase enzyme, and interacts with a ribosome to synthesize genetic information encoded by DNA. Generally, mRNA are classified into two sub-classes: pre-mRNA and mature mRNA. Precursor mRNA (pre-mRNA) is mRNA that has been transcribed by RNA polymerase but has not undergone any post-transcriptional processing (e.g., 5′ capping, splicing, editing, and polyadenylation). Mature mRNA has been modified via post-transcriptional processing (e.g., spliced to remove introns and polyadenylated) and is capable of interacting with ribosomes to perform protein synthesis. mRNA can be isolated from tissues or cells by a variety of methods. For example, a total RNA extraction can be performed on cells or a cell lysate and the resulting extracted total RNA can be purified (e.g., on a column comprising oligo-dT beads) to obtain extracted mRNA.
Alternatively, mRNA can be synthesized in a cell-free environment, for example by in vitro transcription (IVT). An “in vitro transcription template” as used herein, refers to deoxyribonucleic acid (DNA) suitable for use in an IVT reaction for the production of messenger RNA (mRNA). In some embodiments, an IVT template encodes a 5′ untranslated region, contains an open reading frame, and encodes a 3′ untranslated region and a polyA tail.
The particular nucleotide sequence composition and length of an IVT template will depend on the mRNA of interest encoded by the template.
A “5′ untranslated region (UTR)” refers to a region of an mRNA that is directly upstream (i.e., 5′) from the start codon (i.e., the first codon of an mRNA transcript translated by a ribosome) that does not encode a protein or peptide.
A “3′ untranslated region (UTR)” refers to a region of an mRNA that is directly downstream (i.e., 3′) from the stop codon (i.e., the codon of an mRNA transcript that signals a termination of translation) that does not encode a protein or peptide.
A “polyA tail” is a region of mRNA that is downstream, e.g., directly downstream (i.e., 3′), from the 3′ UTR that contains multiple, consecutive adenosine monophosphates. A polyA tail may contain 10 to 300 adenosine monophosphates. For example, a polyA tail may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290 or 300 adenosine monophosphates. In some embodiments, a polyA tail contains 50 to 250 adenosine monophosphates. In a relevant biological setting (e.g., in cells, in vivo, etc.) the poly(A) tail functions to protect mRNA from enzymatic degradation, e.g., in the cytoplasm, and aids in transcription termination, export of the mRNA from the nucleus, and translation.
As used herein, the term “antigen” refers to a substance, typically a protein, which is capable of inducing an immune response in a subject. The term also refers to proteins that are immunologically active (also referred to as “immunogenic”) in the sense that once administered to a subject (either directly or by administering to the subject a nucleotide sequence or vector that encodes the protein) the protein is able to evoke an immune response of the humoral and/or cellular type directed against that protein.
As used herein, the term “microRNA” refers to a major class of biomolecules involved in control of gene expression. For example, in human heart, liver, or brain, miRNAs play a role in tissue specification or cell lineage decisions. In addition, miRNAs influence a variety of processes, including early development, cell proliferation and cell death, and apoptosis and fat metabolism. The large number of miRNA genes, the diverse expression patterns, and the abundance of potential miRNA targets suggest that miRNAs may be a significant source of genetic diversity. A mature miRNA is typically an 8-25 nucleotide non-coding RNA that regulates expression of an mRNA including sequences complementary to the miRNA. These small RNA molecules are known to control gene expression by regulating the stability and/or translation of mRNAs. For example, miRNAs bind to the 3′ UTR of target mRNAs and suppress translation. MiRNAs may also bind to target mRNAs and mediate gene silencing through the RNAi pathway. MiRNAs may also regulate gene expression by causing chromatin condensation.
A miRNA silences translation of one or more specific mRNA molecules by binding to a miRNA recognition element (MRE) which is defined as any sequence that directly base pairs with and interacts with the miRNA somewhere on the mRNA transcript. Often, the MRE is present in the 3′ untranslated region (UTR) of the mRNA, but it may also be present in the coding sequence or in the 5′ UTR. MREs are not necessarily perfect complements to miRNAs, usually having only a few bases of complementarity to the miRNA and often containing one or more mismatches within those bases of complementarity. The MRE may be any sequence capable of being bound by a miRNA sufficiently that the translation of a gene to which the MRE is operably linked (such as a CMV gene that is essential or augmenting for growth in vivo) is repressed by a miRNA silencing mechanism such as the RISC.
As used herein, the term “heterologous antigen” refers to any protein or fragment thereof that is not derived from CMV. Heterologous antigens may be pathogen-specific antigens, tumor virus antigens, tumor antigens, host self-antigens, or any other antigen.
Orthologs of proteins are typically characterized by possession of greater than 75% sequence identity counted over the full-length alignment with the amino acid sequence of specific protein using ALIGN set to default parameters. Proteins with even greater similarity to a reference sequence will show increasing percentage identities when assessed by this method, such as at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, or at least 98% sequence identity. In addition, sequence identity can be compared over the full length of particular domains of the disclosed peptides.
The term “homologous” or “homolog” refers to a molecule or activity found in or derived from a host cell, species, or strain. For example, a heterologous or exogenous molecule or gene encoding the molecule may be homologous to a native host or host cell molecule or gene that encodes the molecule, respectively, but may have an altered structure, sequence, expression level or combinations thereof.
As used herein, the identity/similarity between two or more nucleic acid sequences, or two or more amino acid sequences, is expressed in terms of the identity or similarity between the sequences. Sequence identity may be measured in terms of percentage identity; the higher the percentage, the more identical the sequences are. Sequence similarity may be measured in terms of percentage identity or similarity (which takes into account conservative amino acid substitutions); the higher the percentage, the more similar the sequences are. Polypeptides or protein domains thereof that have a significant amount of sequence identity and also function the same or similarly to one another (for example, proteins that serve the same functions in different species or mutant forms of a protein that do not change the function of the protein or the magnitude thereof) may be called “homologs.”
Methods of alignment of sequences for comparison are well known in the art. Various programs and alignment algorithms are described in: Smith & Waterman, Adv Appl Math 2, 482 (1981); Needleman & Wunsch, J Mol Biol 48, 443 (1970); Pearson & Lipman, Proc Natl Acad Sci USA 85, 2444 (1988); Higgins & Sharp, Gene 73, 237-244 (1988); Higgins & Sharp, CABIOS 5, 151-153 (1989); Corpet et al., Nuc Acids Res 16, 10881-10890 (1988); Huang et al., Computer App Biosci 8, 155-165 (1992); and Pearson et al., Meth Mol Bio 24, 307-331 (1994). In addition, Altschul et al., J Mol Biol 215, 403-410 (1990), presents a detailed consideration of sequence alignment methods and homology calculations.
The NCBI Basic Local Alignment Search Tool (BLAST) (Altschul et al., (1990) supra) is available from several sources, including the National Center for Biological Information (NCBI, National Library of Medicine, Building 38A, Room 8N805, Bethesda, MD 20894) and on the Internet, for use in connection with the sequence analysis programs blastp, blastn, blastx, tblastn and tblastx. Additional information may be found at the NCBI web site.
BLASTN is used to compare nucleic acid sequences, while BLASTP is used to compare amino acid sequences. If the two compared sequences share homology, then the designated output file will present those regions of homology as aligned sequences. If the two compared sequences do not share homology, then the designated output file will not present aligned sequences.
Once aligned, the number of matches is determined by counting the number of positions where an identical nucleotide or amino acid residue is presented in both sequences. The percent sequence identity is determined by dividing the number of matches either by the length of the sequence set forth in the identified sequence, or by an articulated length (such as 100 consecutive nucleotides or amino acid residues from a sequence set forth in an identified sequence), followed by multiplying the resulting value by 100. For example, a nucleic acid sequence that has 1166 matches when aligned with a test sequence having 1154 nucleotides is 75.0 percent identical to the test sequence (1166÷1554*100=75.0). The percent sequence identity value is rounded to the nearest tenth. For example, 75.11, 75.12, 75.13, and 75.14 are rounded down to 75.1, while 75.15, 75.16, 75.17, 75.18, and 75.19 are rounded up to 75.2. The length value will always be an integer. In another example, a target sequence containing a 20-nucleotide region that aligns with 20 consecutive nucleotides from an identified sequence as follows contains a region that shares 75 percent sequence identity to that identified sequence (that is, 15÷20*100=75).
For comparisons of amino acid sequences of greater than about 30 amino acids, the Blast 2 sequences function is employed using the default BLOSUM62 matrix set to default parameters, (gap existence cost of 11, and a per residue gap cost of 1). Homologs are typically characterized by possession of at least 70% sequence identity counted over the full-length alignment with an amino acid sequence using the NCBI Basic Blast 2.0, gapped blastp with databases such as the nr database, swissprot database, and patented sequences database. Queries searched with the blastn program are filtered with DUST (Hancock & Armstrong, Comput Appl Biosci 10, 67-70 (1994.) Other programs use SEG. In addition, a manual alignment may be performed. Proteins with even greater similarity will show increasing percentage identities when assessed by this method, such as at least about 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity to a protein.
When aligning short peptides (fewer than around 30 amino acids), the alignment is performed using the Blast 2 sequences function, employing the PAM30 matrix set to default parameters (open gap 9, extension gap 1 penalties). Proteins with even greater similarity to the reference sequence will show increasing percentage identities when assessed by this method, such as at least about 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity to a protein. When less than the entire sequence is being compared for sequence identity, homologs will typically possess at least 75% sequence identity over short windows of 10-20 amino acids, and may possess sequence identities of at least 85%, 90%, 95% or 98% depending on their identity to the reference sequence. Methods for determining sequence identity over such short windows are described at the NCBI web site.
One indication that two nucleic acid molecules are closely related is that the two molecules hybridize to each other under stringent conditions, as described above. Nucleic acid sequences that do not show a high degree of identity may nevertheless encode identical or similar (conserved) amino acid sequences, due to the degeneracy of the genetic code. Changes in a nucleic acid sequence may be made using this degeneracy to produce multiple nucleic acid molecules that all encode substantially the same protein. Such homologous nucleic acid sequences can, for example, possess at least about 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99% sequence identity to a nucleic acid that encodes a protein.
The human cytomegalovirus UL82 gene encodes pp71, a protein that is localized in the tegument domain of the virus particle. The UL82 gene of the CMV TR strain is SEQ ID NO:1, 118811 to 120490 for GenBank Accession No. KF021605.1. As used herein, UL82 refers to SEQ ID NO:1 and orthologs or homologs thereof.
As used herein, “pp71”, refers to a protein that is localized in the tegument domain of the CMV virus particle. Pp71 may perform one or more functions, including inhibition of Daxx repression of viral gene transcription, negative regulation of STING, and evasion of cell antiviral responses (Kalejta R F, et al. Expanding the Known Functional Repertoire of the Human Cytomegalovirus pp71 Protein. Front Cell Infect Microbiol. 2020 Mar. 12; 10:95). Deletion of UL82 or disruption of UL82 by insertion of a foreign gene at the UL82 locus results in the absence of pp71 protein and consequently reduces replication in fibroblasts, endothelial cells, epithelial cells, and astrocytes (Caposio P et al., Characterization of a live-attenuated HCMV-based vaccine platform. Sci Rep. 2019 Dec. 17; 9 (1): 19236). The effects of UL82 deletion or disruption are reversible by cell kinase inhibitors. The rhesus cytomegalovirus (RhCMV) gene RhCMV 110 is homologous to human CMV UL82 (Hansen S G, et al. Complete sequence and genomic analysis of rhesus cytomegalovirus. J Virol. 2003 June; 77 (12): 6620-36).
A challenge for manufacturing HCMV vectors having desirable properties for vaccines is that the vectors are often designed to have reduced viral replication or growth. For example, some live attenuated HCMV-HIV vaccine vectors are engineered to be growth deficient by deletion of the HCMV gene UL82 (SEQ ID NO:1, GenBank Accession No.: KF021605.1 (118815 to 120494)), which encodes the tegument protein pp71 (SEQ ID NO:2, GenBank Accession No.: AGL96671.1; SEQ ID NO:3, UniProtKB-R4SH92), resulting in lower viral yield. Commercial production of live HCMV-based HIV vaccines attenuated by pp71-deletion ultimately requires a complementing cell line permissive for vector growth under good manufacturing practices (GMP).
pp71 is important for wild type HCMV infection because this tegument protein is translocated to the nucleus where it suppresses cellular Daxx function, thus allowing CMV immediate-early (IE) gene expression that triggers the replication cycle. Some manufacturing processes rely on functional complementation using transient transfection of MRC-5 cells with an siRNA targeting DAXX, which mimics one of the functions of HCMV pp71. This method allows vector growth but involves complex manipulations not readily scalable for commercial production and complements only one of the many pp71 functions. In addition, RhCMV vectors grown in pp71 complementing cells have demonstrated an increase in potency as measured by the reduction in focus-forming units (FFUs) per dose required to induce an immune response. These observations suggest that cell-derived pp71 protein can be packaged in the virion of vectors in which this gene is deleted, and the viral vaccine containing pp71 protein can be administered at lower doses due to its increased potency, thereby providing clinical benefit and reducing manufacturing needs.
In some embodiments, the recombinant RhCMV or HCMV vector comprises a deletion in a RhCMV or HCMV gene that is essential for or augments replication (e.g. UL82). CMV essential genes and augmenting have been well described in the art (see, for example, Dunn et al., Proc. Natl. Acad. Sci. USA 100 (24): 14223-14228, 2003; and Dong et al., Proc. Natl. Acad. Sci. USA 100 (21): 12396-12401, 2003). Essential CMV genes include, but are not limited to, UL32, UL34, UL37, UL44, UL46, UL48, UL48.5, UL49, UL50, UL51, UL52, UL53, UL54, UL55, UL56, UL57, UL60, UL61, UL70, UL71, UL73, UL75, UL76, UL77, UL79, UL80, UL82, UL84, UL85, UL86, UL87, UL89, UL90, UL91, UL92, UL93, UL94, UL95, UL96, UL98, UL99, UL100, UL102, UL104, UL105, UL115 and UL122. In some embodiments, the CMV essential or augmenting gene is UL82, UL94, UL32, UL99, UL115 or UL44, or a homolog thereof (i.e., the homologous gene in RhCMV). Other essential or augmenting genes are known in the art and are described herein. In particular examples, the essential gene is UL82, or a homolog thereof.
mRNA transfection can be used to enable the host cell to express the essential viral gene. Transfection of a mRNA for expressing the essential viral gene may be able to provide all of the functions of the gene that are likely to enhance the infection process, such as cell cycle stimulation, efficient virion packaging, and virus stability. In addition, protein present late in infection has the potential to be packaged in the progeny virus, which could lower the required dose of the vaccine by more efficient first round infection and establishment of persistent infection.
In some embodiments, mRNA transfection of the essential viral gene into a host cell provides functional complementation resulting in successful propagation of gene-deleted HCMV virus vector. In some embodiments, the functional complementation results in accelerated HCMV spread, increased maximal titers, earlier maximal virus titers, and/or enhanced reconstitution of virus from BAC DNA.
In some embodiments, transient transfection of mRNA is used to identify functions that permit HCMV to grow to higher titers by supplementing infection with combinatorial libraries of mRNAs from laboratory strains known to grow to high titer.
In a natural infection, infectious parental CMV particles enter cells through interaction with cellular receptors, and capsid and tegument proteins are delivered into the cytosol. The capsid enters the nucleus and delivers the CMV genome, while tegument proteins are involved in initiating viral gene expression and regulating host cell responses. The viral genome is replicated and encapsulated into capsids that have been assembled in the nucleus, and the genome-containing capsids are transported to the cytosol where they associate with tegument proteins and acquire a viral envelope in the viral assembly complex. The enveloped infectious progeny CMV particles are then released from the cell (Jean Beltran P M and Cristea I M. The life cycle and pathogenesis of human cytomegalovirus infection: lessons from proteomics. Expert Rev Proteomics. 2014 December; 11 (6): 697-711). The parental viral particles and progeny particles may be structurally and genetically identical, or may be different, for example in the case of viral recombination when the cell is co-infected by multiple parental strains. The process of natural infection is harnessed in the laboratory manufacture of CMV viral particles, where a cell line is infected with a parental CMV, the cells produce progeny CMV, and the progeny CMV are then collected. In these processes, “parental” refers to the viral particles that infect the cells, and “progeny” refers to the produced viral particles.
In some embodiments, transfected mRNA is applied to cell lines for use in determining the infectious titer of viral stocks.
MRC-5 cells are well characterized primary normal diploid fibroblasts suitable for HCMV production. In some embodiments, MRC-5 cells are used in the methods disclosed herein for generating cells that express pp71.
In some embodiments, transfection of naturally permissive MRC-5 cells with UL82 mRNA leads to higher apparent titers as compared to either transfection of BJ-5ta cells or the pp71 BJ-5ta cell line. This likely provides a more accurate titer of the material and allows better quantification of diluted material used in dose range studies. Transfection of pp71 mRNA may allow for lower titer viruses to be tested (e.g., less than 5e4 FFU/mL) with greater assay reproducibility and confidence.
In some embodiments, a method of producing a CMV viral vector is provided, comprising: (a) introducing a mRNA molecule encoding a pp71 protein to a cell; (b) infecting the cell with a CMV; (c) incubating the cell; and (d) collecting the CMV viral vector. In some embodiments, the mRNA molecule encoding a pp71 protein comprises a sequence according to SEQ ID NOs: 4-10. In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with pseudouridine (pseudoU) and 5-methylcytidine (5meC), e.g. SEQ ID NOs: 14-20. In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with 5-methoxyuridine (5moU), e.g. SEQ ID NOs: 21-27. In some embodiments, the mRNA molecule encoding a pp71 protein is delivered to the cell using transfection. In some embodiments, the cell is a MRC-5 cell.
The mRNA molecule can be manufactured by in vitro transcription which typically uses a double stranded DNA template in buffer with an RNA polymerase and a mix of NTPs. The polymerase can synthesize the mRNA molecule. The DNA can then be enzymatically degraded. The mRNA molecule can be purified away from polymerase, free NTPs, and degraded DNA.
Transfection of cells with unmodified mRNA molecules can lead to cell death due to activation of innate immune pathways. Modifications to the mRNA molecule can act as marks of self-RNA to reduce innate immune responses to endogenous cellular RNA and enhance stability. Such modifications include, but are not limited to, modified nucleosides, elongation of the poly-adenosine (poly(A)) tail, and modified 5′ cap structures. Furthermore, removal of dsRNA contaminants through high-performance liquid chromatography (HPLC) purification can also enhance stability and reduce immune recognition.
The term “RNA” or “mRNA” or “mRNA molecule” encompasses not only RNA molecules containing natural ribonucleotides, but also analogs and derivatives of RNA comprising one or more nucleotide/nucleoside or ribonucleotide/ribonucleoside analogs or derivatives as described herein or as known in the art. Strictly speaking, a “nucleoside” includes a nucleoside base and a deoxyribose sugar, and a “nucleotide” is a nucleoside with one, two or three phosphate moieties. Strictly speaking, a “ribonucleoside” includes a nucleoside base and a ribose sugar, and a “ribonucleotide” is a ribonucleoside with one, two or three phosphate moieties.
The RNA molecule can be modified in the nucleobase structure or in the ribose-phosphate backbone structure, e.g., as described in greater detail below. As non-limiting examples, an RNA molecule can also include at least one modified ribonucleoside including, but not limited to, a 5-methoxyuridine (5moU) modified nucleoside, a 5-methylcytidine (5meC) modified nucleoside, a N6-methyladenosine (m6A) modified nucleoside, a 5-methyluridine (m5U) modified nucleoside, a pseudouridine (pseudoU) modified nucleoside, a2-thioruridine (s2U) modified nucleoside, or any combination thereof. In another example, an RNA molecule can comprise at least two modified ribonucleosides, at least 25, at least 50, at least 100, at least 500, at least 1000, at least 2000, or more, up to the entire length of the mRNA molecule. The modifications need not be the same for each of such a plurality of modified nucleosides/ribonucleosides in an RNA molecule.
In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with pseudouridine (pseudoU), 5-methylcytidine (5meC), 5-methoxyuridine (5moU), or any combination thereof. In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with pseudouridine (pseudoU) and 5-methylcytidine (5meC), e.g. SEQ ID NOs: 14-20. In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with 5-methoxyuridine (5moU), e.g. SEQ ID NOs: 21-27. In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with pseudouridine (pseudoU). In some embodiments, the mRNA molecule encoding a pp71 protein is produced using full substitution with 5-methylcytidine (5meC).
In some embodiments, a poly-adenosine (poly(A)) tail of variable or pre-determined length can be added to the 3′ end of the mRNA molecule encoding the pp71 protein. In some embodiments, the poly(A) tail is approximately 60-100 nucleotides long. A poly(A) tail may be added in a template-dependent fashion during transcription and/or may be added enzymatically post-transcription. In certain embodiments, a poly(A) tail is added enzymatically post-transcription by a poly(A) polymerase, which variably adds a tail of approximately 60-100 nucleotides. In certain other embodiments, a poly(A) tail is synthesized from a double-stranded DNA template, e.g., a linearized plasmid template (“Run-off Transcription” (TriLink)), wherein transcription stops when RNA polymerase falls off the DNA. In further embodiments the plasmid encodes a poly(A) tail of a pre-determined length of approximately 80 nt.
The mRNA molecule can be transcribed to contain a 5′ cap structure. In some embodiments the mRNA molecule is transcribed with a 7-methylguanylate cap, creating a cap0 structure. In some embodiments the mRNA molecule is transcribed with a modified 5′-methoxyuridine (5moU) nucleoside, creating a cap1 structure.
In some embodiments, the mRNAs can be transcribed to contain the full-length viral pp71 5′ UTR and 3′ UTR (untranslated region) to enable nuclear localization.
RNA can be introduced into target cells using any of a number of different methods, for instance, commercially available methods which include, but are not limited to, lipid-mediated transfection, electroporation, multiporation, polymer encapsulation, peptide mediated transfection, or biolistic particle delivery systems.
In some embodiments, the mRNA molecule is delivered to cells using lipid-based transfection. Commercially available lipid-mediated transfection reagents include Lipofectamine 2000 (ThermoFisher), MessengerMax (ThermoFisher), Jet-mRNA (PolyPlus) and Trans-IT (Mirus Bio). In certain embodiments the mRNA is introduced to a cell with MessengerMax, which provides high transfection efficiency and low toxicity. In some embodiments, the mRNA molecule is delivered at a dose of 5 ng/cm2 to 500 ng/cm2. In some embodiments, the mRNA molecule is delivered at a dose of 50 ng/cm2 to 100 ng/cm2. In some embodiments, the mRNA molecule is delivered at a dose of 50 ng/cm2. In some embodiments, the mRNA molecule is delivered at a dose of 100 ng/cm2. In some embodiments, the mRNA molecule is delivered at a dose of at least 5 ng/cm2, at least 50 ng/cm2, at least 100 ng/cm2, at least 150 ng/cm2, at least 200 ng/cm2, at least 250 ng/cm2, at least 300 ng/cm2, at least 350 ng/cm2, at least 400 ng/cm2, at least 450 ng/cm2, or at least 500 ng/cm2. In some embodiments, the mRNA molecule is delivered at a dose of 100 ng/cm2. In some embodiments, the mRNA molecule is delivered at a dose of no more than 50 ng/cm2, no more than 100 ng/cm2, no more than 150 ng/cm2, no more than 200 ng/cm2, no more than 250 ng/cm2, no more than 300 ng/cm2, no more than 350 ng/cm2, no more than 400 ng/cm2, no more than 450 ng/cm2, or no more than 500 ng/cm2. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 0.01 μl to 1000 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 0.1 μl to 100 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 0.1 μl to 50 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 0.1 μl to 10 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 0.5 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 0.75 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 1 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 1.25 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 1.5 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 2 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 2.5 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 3 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is 3.6 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is at least 0.01 μl, at least 0.1 μl, at least 0.5 μl, at least 1 μl, at least 2 μl, at least 3 μl, at least 4 μl, at least 5 μl, at least 10 μl, at least 50 μl, at least 100 μl, or at least 1000 μl. In some embodiments, the lipid-mediated transfection reagent amount used in the transfection is no more than 0.01 μl, no more than 0.1 μl, no more than 0.5 μl, no more than 1 μl, no more than 2 μl, no more than 3 μl, no more than 4 μl, no more than 5 μl, no more than 10 μl, no more than 50 μl, no more than 100 μl, or no more than 1000 μl.
In any of the aforementioned methods and compositions, the CMV may be a HCMV or a RhCMV. In some embodiments, the CMV is a HCMV. In some embodiments, the CMV is a genetically modified TR strain of HCMV. In some embodiments, the CMV comprises a TR3 backbone.
In some embodiments, the recombinant CMV vector is or is derived from HCMV TR3. As referred to herein, “HCMV TR3” or “TR3” refers to a HCMV-TR3 vector backbone derived from the clinical isolate HCMV TR, as described in Caposio, P et al. (Characterization of a live attenuated HCMV-based vaccine platform. Scientific Reports 9, 19236 (2019)).
In some embodiments, the recombinant CMV vector (e.g., a recombinant HCMV vector comprising a TR3 backbone) comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE). In some embodiments, the HCMV vector comprises a nucleic acid sequence encoding an MRE that contains target sites for microRNAs expressed in endothelial cells. Examples of miRNAs expressed in endothelial cells are miR126, miR-126-3p, miR-130a, miR-210, miR-221/222, miR-378, miR-296, and miR-328. In some embodiments, the HCMV vector lacks UL18, UL128, UL130, UL146, and UL147 (and optionally UL82) and expresses UL40 and US28 and the MRE contains target sites for microRNAs expressed in endothelial cells.
In some embodiments, the recombinant CMV vector (e.g., a recombinant HCMV vector comprising a TR3 backbone) comprises a nucleic acid sequence encoding an MRE that contains target sites for microRNAs expressed in myeloid cells. Examples of miRNAs expressed in myeloid cells are miR-142-3p, miR-223, miR-27a, miR-652, miR-155, miR-146a, miR-132, miR-21, miR-124, and miR-125.
MREs that may be included in the recombinant CMV vector disclosed herein may be any miRNA recognition element that silences expression in the presence of a miRNA expressed by endothelial cells, or any miRNA recognition element that silences expression in the presence of a miRNA expressed by myeloid cells. Such an MRE may be the exact complement of a miRNA. Alternatively, other sequences may be used as MREs for a given miRNA. For example, MREs may be predicted from sequences using publicly available data bases. In one example, the miRNA may be searched on the website microRNA.org (www.microrna.org). In turn, a list of mRNA targets of the miRNA will be listed. For each listed target on the page, ‘alignment details’ may be accessed and putative MREs accessed. One of ordinary skill in the art may select a validated, putative, or mutated MRE sequence from the literature that would be predicted to induce silencing in the presence of a miRNA expressed in a myeloid cell such as a macrophage. One example involves the above referenced website. The person of ordinary skill in the art may then obtain an expression construct whereby a reporter gene (such as a fluorescent protein, enzyme, or other reporter gene) has expression driven by a promoter such as a constitutively active promoter or cell specific promoter. The MRE sequence may then be introduced into the expression construct. The expression construct may be transfected into an appropriate cell, and the cell transfected with the miRNA of interest. A lack of expression of the reporter gene indicates that the MRE silences gene expression in the presence of the miRNA.
In some embodiments, the CMV vector comprises a nucleic acid sequence that does not encode any MREs.
In any of the aforementioned methods and compositions, the CMV may be genetically modified. In some embodiments, the CMV comprises a deletion of a gene, or does not express an active gene, wherein the gene is UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof. In some embodiments, the CMV comprises a deletion of a gene, or does not express an active gene, wherein the gene is UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof. In some embodiments, the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, and does not express an active UL147 or homolog thereof. In some embodiments, the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, and does not express an active UL82 or homolog thereof. In some embodiments, the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, does not express an active UL82 or homolog thereof, and does not express an active UL18 or homolog thereof. In some embodiments, the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, and does not express an active UL82 or homolog thereof, wherein the CMV additionally expresses a target sequence for mir124. In some embodiments, the CMV does not express an active UL82 or homolog thereof. In some embodiments, the CMV comprises a deletion of UL128, UL130, UL146, and UL147, or homologs thereof. In some embodiments, the CMV comprises a deletion of UL128, UL130, UL146, UL147, and UL82, or homologs thereof. In some embodiments, the CMV comprises a deletion of UL128, UL130, UL146, UL147, UL82, and UL18, or homologs thereof. In some embodiments, the CMV comprises a deletion of UL128, UL130, UL146, UL147, and UL82, or homologs thereof, wherein the CMV additionally expresses a target sequence for mir124. In some embodiments, the CMV comprises a deletion of UL82 or homolog thereof. In some embodiments, a nucleic acid encoding a heterologous antigen replaces UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof. In some embodiments, a nucleic acid encoding a heterologous antigen replaces UL82.
In any of the aforementioned methods and compositions, the CMV may comprise a nucleic acid encoding a heterologous antigen. In some embodiments, the heterologous antigen comprises a pathogen-specific antigen or a tumor antigen. In some embodiments, the heterologous antigen comprises a pathogen-specific antigen comprising a human immunodeficiency virus (HIV) antigen, a simian immunodeficiency virus (SIV) antigen, a human cytomegalovirus (HCMV) antigen, a hepatitis B virus (HBV) antigen, a hepatitis C virus (HCV) antigen, a papilloma virus antigen (e.g., a human papilloma virus (HPV) antigen), a Plasmodium antigen, a Kaposi's sarcoma-associated herpesvirus antigen, a Varicella zoster virus (VZV) antigen, an Ebola virus, a Mycobacterium tuberculosis antigen, a Chikungunya virus antigen, a dengue virus antigen, a monkeypox virus antigen, a herpes simplex virus (HSV) 1 antigen, a herpes simplex virus (HSV) 2 antigen, an Epstein-Barr virus (EBV) antigen, a poliovirus antigen, an influenza virus antigen, or a Clostridium tetani antigen. In some embodiments, the heterologous antigen comprises a HIV antigen. In some embodiments, the heterologous antigen comprises a HIV antigen, wherein the HIV antigen is Gag, Pol, Nef, Env, Tat, Rev, Tat, Vpr, Vif, or Vpu, or an epitope or antigenic fragment thereof. In some embodiments, the heterologous antigen comprises a HIV antigen, wherein the HIV antigen comprises more than one of Gag, Pol, Nef, Env, Tat, Rev, Tat, Vpr, Vif, and Vpu, or an epitope or antigenic fragment thereof. In some embodiments, the heterologous antigen comprises a HIV antigen, wherein the HIV antigen comprises more than one of Gag, Pol, Nef, Env, Tat, Rev, Tat, Vpr, Vif, and Vpu, or an epitope or antigenic fragment thereof, comprised in a fusion molecule. In some embodiments, the heterologous antigen comprises a Mycobacterium tuberculosis antigen. In some embodiments, the heterologous antigen comprises a Mycobacterium tuberculosis antigen, wherein the Mycobacterium tuberculosis antigen is Ag85A, ESAT-6, Rv3407, Rv2626c, Rv2626c, RpfA, or RpfD or an epitope or antigenic fragment thereof. In some embodiments, the Mycobacterium tuberculosis antigen comprises more than one of Ag85A, ESAT-6, Rv3407, Rv2626c, Rv2626c, RpfA, and RpfD, or an epitope or antigenic fragment thereof. In some embodiments, the Mycobacterium tuberculosis antigen comprises more than one of Ag85A, ESAT-6, Rv3407, Rv2626c, Rv2626c, RpfA, and RpfD, or an epitope or antigenic fragment thereof, comprised in a fusion molecule.
In some embodiments, the heterologous antigen comprises a prostate cancer antigen.
In some aspects, the present disclosure provides a CMV viral vector produced by any of the aforementioned methods.
In some embodiments, the present disclosure provides:
1. A method of producing a progeny cytomegalovirus (CMV), comprising:
2. A method of producing a progeny CMV, comprising:
3. A method of producing a progeny CMV, comprising:
4. A method of producing a CMV viral vector, comprising:
5. The method of embodiment 1, wherein the gene that is essential for or augments CMV replication is UL82, UL32, UL34, UL37, UL44, UL46, UL48, UL48.5, UL49, UL50, UL51, UL52, UL53, UL54, UL55, UL56, UL57, UL60, UL61, UL70, UL71, UL73, UL75, UL76, UL77, UL79, UL80, UL84, UL85, UL86, UL87, UL89, UL90, UL91, UL92, UL93, UL94, UL95, UL96, UL98, UL99, UL100, UL102, UL104, UL105, UL115, or UL122, or a homolog thereof.
6. The method of any one of embodiments 1-3 and 5, wherein the progeny CMV comprises pp71 protein.
7. The method of embodiment 4, wherein the CMV viral vector comprises pp71 protein.
8. The method of any one of embodiments 1-7, wherein the cell is a MRC-5 cell.
9 The method of any one of embodiments 1-8, wherein the mRNA molecule comprises the sequence according to SEQ ID NO:14.
10. The method of any one of embodiments 1-8, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOs: 14-20.
11. The method of any one of embodiments 1-8, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOs: 4-10.
12. The method of any one of embodiments 1-8, wherein the mRNA molecule comprises the sequence according to SEQ ID NO:4.
13. The method of any one of embodiments 1-8, 11, and 12, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOS: 4-10, wherein each uridine is substituted with pseudouridine and each cytidine is substituted with 5-methylcytidine.
14. The method of any one of embodiments 1-8, 11, and 12, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOs: 4-10, wherein each uridine is substituted with 5-methoxyuridine.
15. The method of any one of embodiments 1-8, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOs: 21-27.
16. The method of any one of embodiments 1-15, wherein the mRNA molecule further comprises a poly(A) tail.
17. The method of any one of embodiments 1-16, wherein a poly(A) tail has been added to the 3′ end of the mRNA molecule encoding the pp71 protein.
18. The method of embodiment 16 or embodiment 17, wherein the mRNA molecule was produced using a double-stranded DNA template encoding the poly(A) tail.
19. The method of embodiment 18, wherein the double-stranded DNA template is a plasmid.
20. The method of any one of embodiments 16-19, wherein the poly(A) tail is approximately 60-100 nucleotides long.
21. The method of any one of embodiments 16-19, wherein the poly(A) tail is 80 nucleotides long. 22. The method of any one of embodiments 1-10 and 16-21, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOs: 14-20, wherein each uridine is substituted with pseudouridine and each cytidine is substituted with 5-methylcytidine, and has a poly(A) tail 80 nucleotides in length.
23. The method of any one of embodiments 1-8, 11-13, and 16-21, wherein the mRNA molecule comprises the sequence according to one of SEQ ID NOs: 4-10, wherein each uridine is substituted with pseudouridine and each cytidine is substituted with 5-methylcytidine, and has a poly(A) tail 80 nucleotides in length; and wherein the poly(A) tail was produced using a plasmid template.
24. The method of any one of embodiments 1-8, 11, 14, and 16-21, wherein the mRNA molecule comprises the sequence according to SEQ ID NOs: 4-10, wherein each uridine is substituted with 5-methoxyuridine, and has a poly(A) tail 80 nucleotides in length; wherein the poly(A) tail was produced using a plasmid template.
25. The method of any one of embodiments 1-8 and 15-21, wherein the mRNA molecule comprises a sequence according to SEQ ID NOs: 21-27, wherein each uridine is substituted with 5-methoxyuridine, and has a poly(A) tail 80 nucleotides in length.
26. The method of embodiment 16 or embodiment 17, wherein the poly(A) tail is or has been added by an enzyme after transcription. 27. The method of embodiment 26, wherein the poly(A) tail is approximately 50-100 nucleotides long.
28. The method of any one of embodiments 1-27, wherein the mRNA molecule is introduced using transfection.
29. The method of embodiment 28, wherein the transfection is accomplished using a lipid transfection reagent.
30. The method of embodiment 29, wherein the lipid transfection reagent comprises MessengerMax, Lipofectamine 2000, Jet-mRNA, or Trans-IT.
31. The method of any one of embodiments 1-30, wherein the mRNA molecule is delivered at a dose of 5 ng/cm2 to 500 ng/cm2.
32. The method of any one of embodiments 1-31, wherein the mRNA molecule is delivered at a dose of 50 ng/cm2 to 100 ng/cm2.
33. The method of any one of embodiments 1-32, wherein the mRNA molecule is delivered at a dose of 50 ng/cm2.
34. The method of any one of embodiments 1-33, wherein the mRNA molecule is delivered at a dose of 100 ng/cm2.
35. The method of any one of embodiments 4, 7, and 8-34, wherein the CMV is a HCMV.
36. The method of any one of embodiments 1-3, 5, 6, and 8-34, wherein the parent CMV is a HCMV.
37. The method of any one of embodiments 1-3, 5, 6, 8-34, and 36, wherein the progeny CMV is a HCMV.
38. The method of embodiment 35, wherein the CMV is a genetically modified TR strain of HCMV.
39. The method of embodiment 36 or embodiment 37, wherein the parent CMV is a genetically modified TR strain of HCMV.
40. The method of any one of embodiments 36, 37, and 39, wherein the progeny CMV is a genetically modified TR strain of HCMV.
41. The method of any one of embodiments 4, 7, 8-35, and 37, wherein the CMV comprises a TR3 backbone.
42. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, and 40, wherein the parent CMV comprises a TR3 backbone.
43. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, and 42, wherein the progeny CMV comprises a TR3 backbone. 44. The method of any one of embodiments 4, 7, 8-34, 35, 38, and 41, wherein the CMV comprises a nucleic acid encoding a heterologous antigen.
45. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, and 43, wherein the parent CMV comprises a nucleic acid encoding a heterologous antigen.
46. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, and 45, wherein the progeny CMV comprises a nucleic acid encoding a heterologous antigen.
47. The method of any one of embodiments 44-46, where in the heterologous antigen comprises a pathogen-specific antigen or a tumor antigen.
48. The method of any one of embodiments 44-46, wherein the heterologous antigen comprises a pathogen-specific antigen comprising a human immunodeficiency virus (HIV) antigen, a simian immunodeficiency virus (SIV) antigen, a human cytomegalovirus (HCMV) antigen, a hepatitis B virus (HBV) antigen, a hepatitis C virus (HCV) antigen, a papilloma virus antigen (e.g., a human papilloma virus (HPV) antigen), a Plasmodium antigen, a Kaposi's sarcoma-associated herpesvirus antigen, a Varicella zoster virus (VZV) antigen, an Ebola virus, a Mycobacterium tuberculosis antigen, a Chikungunya virus antigen, a dengue virus antigen, a monkeypox virus antigen, a herpes simplex virus (HSV) 1 antigen, a herpes simplex virus (HSV) 2 antigen, an Epstein-Barr virus (EBV) antigen, a poliovirus antigen, an influenza virus antigen, or a Clostridium tetani antigen.
49. The method of any one of embodiments 44-46, wherein the heterologous antigen comprises a HIV antigen.
50. The method of embodiment 49, wherein the HIV antigen is Gag, Pol, Nef, Env, Tat, Rev, Tat, Vpr, Vif, or Vpu, or an epitope or antigenic fragment thereof.
51. The method of embodiment 49, wherein the HIV antigen comprises more than one of Gag, Pol, Nef, Env, Tat, Rev, Tat, Vpr, Vif, and Vpu, or an epitope or antigenic fragment thereof.
52. The method of embodiment 49, wherein the HIV antigen is a fusion protein comprising more than one of Gag, Pol, Nef, Env, Tat, Rev, Tat, Vpr, Vif, and Vpu, or an epitope or antigenic fragment thereof.
53. The method of embodiment 49, wherein the HIV antigen comprises SEQ ID NO: 11 or 12.
54. The method of any one of embodiments 44-46, wherein the heterologous antigen comprises a Mycobacterium tuberculosis antigen.
55. The method of embodiment 54, wherein the Mycobacterium tuberculosis antigen is Ag85A, ESAT-6, Rv3407, Rv2626c, Rv2626c, RpfA, or RpfD or an epitope or antigenic fragment thereof.
56. The method of embodiment 54, wherein the Mycobacterium tuberculosis antigen comprises more than one of Ag85A, ESAT-6, Rv3407, Rv2626c, Rv2626c, RpfA, and RpfD, or an epitope or antigenic fragment thereof.
57. The method of embodiment 54, wherein the Mycobacterium tuberculosis antigen comprises more than one of Ag85A, ESAT-6, Rv3407, Rv2626c, Rv2626c, RpfA, and RpfD, or an epitope or antigenic fragment thereof, comprised in a fusion molecule.
58. The method of embodiment 54, wherein the Mycobacterium tuberculosis antigen comprises SEQ ID NO:13.
59. The method of any one of embodiments 44-46, wherein the heterologous antigen comprises a prostate cancer antigen.
60. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-59, wherein the CMV does not express an active UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
61. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-60, wherein the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, and does not express an active UL147 or homolog thereof.
62. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-61, wherein the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, and does not express an active UL82 or homolog thereof.
63. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-62, wherein the CMV comprises a deletion of UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
64. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-63, wherein the CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, and a deletion of UL147 or homolog thereof.
65. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-64, wherein the CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, and a deletion of UL82 or homolog thereof.
66. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-65, wherein the CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, does not express an active UL82 or homolog thereof, and does not express an active UL18 or homolog thereof.
67. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-66, wherein the CMV further comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE), wherein the MRE contains a target site for a miRNA expressed in endothelial cells or myeloid cells.
68. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-67, wherein the CMV does not express an active UL82 or homolog thereof.
69. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-68, wherein the CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, a deletion of UL82 or homolog thereof, and a deletion of UL18 or homolog thereof.
70. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-68, wherein the CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, and a deletion of UL82 or homolog thereof, wherein the CMV further comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE), wherein the MRE contains a target site for a miRNA expressed in endothelial cells or myeloid cells.
71. The method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, and 47-70, wherein the CMV comprises a deletion of UL82 or homolog thereof.
72. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, and 45-59, wherein the parent CMV does not express an active UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
73. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72, wherein the parent CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, and does not express an active UL147 or homolog thereof.
74. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, 72, and 73, wherein the parent CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, and does not express an active UL82 or homolog thereof.
75. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-74, wherein the parent CMV comprises a deletion of UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
76. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-75, wherein the parent CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, and a deletion of UL147 or homolog thereof.
77. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-76, wherein the parent CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, and a deletion of UL82 or homolog thereof.
78. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-77, wherein the parent CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, does not express an active UL82 or homolog thereof, and does not express an active UL18 or homolog thereof.
79. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-78, wherein the parent CMV further comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE), wherein the MRE contains a target site for a miRNA expressed in endothelial cells or myeloid cells.
80. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-79, wherein the parent CMV does not express an active UL82 or homolog thereof.
81. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-80, wherein the parent CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, a deletion of UL82 or homolog thereof, and a deletion of UL18 or homolog thereof.
82 The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, 72-77, and 79-81, wherein the parent CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, and a deletion of UL82 or homolog thereof, wherein the parent CMV further comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE), wherein the MRE contains a target site for a miRNA expressed in endothelial cells or myeloid cells.
83. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-82, wherein the parent CMV comprises a deletion of UL82 or homolog thereof.
84. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-83, wherein the progeny CMV does not express an active UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
85. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-84, wherein the progeny CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, and does not express an active UL147 or homolog thereof.
86. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-85, wherein the progeny CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, and does not express an active UL82 or homolog thereof.
87. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-86, wherein the progeny CMV comprises a deletion of UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
88. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-87, wherein the progeny CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, and a deletion of UL147 or homolog thereof.
89. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-88, wherein the progeny CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, and a deletion of UL82 or homolog thereof.
90. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-89, wherein the progeny CMV does not express an active UL128 or homolog thereof, does not express an active UL130 or homolog thereof, does not express an active UL146 or homolog thereof, does not express an active UL147 or homolog thereof, does not express an active UL82 or homolog thereof, and does not express an active UL18 or homolog thereof.
91. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-90, wherein the progeny CMV further comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE), wherein the MRE contains a target site for a miRNA expressed in endothelial cells or myeloid cells.
92. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-91, wherein the progeny CMV does not express an active UL82 or homolog thereof.
93. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-92, wherein the progeny CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, a deletion of UL82 or homolog thereof, and a deletion of UL18 or homolog thereof.
94. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, 72-77, 79-89, and 91-94, wherein the progeny CMV comprises a deletion of UL128 or homolog thereof, a deletion of UL130 or homolog thereof, a deletion of UL146 or homolog thereof, a deletion of UL147 or homolog thereof, and a deletion of UL82 or homolog thereof, wherein the progeny CMV further comprises a nucleic acid sequence encoding a microRNA (miRNA) recognition element (MRE), wherein the MRE contains a target site for a miRNA expressed in endothelial cells or myeloid cells.
95. The method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-94, wherein the progeny CMV comprises a deletion of UL82 or homolog thereof.
96. The method of any one of embodiments 44-95, wherein the nucleic acid encoding the heterologous antigen replaces UL128, UL130, UL146, UL147, UL82, or UL18, or homologs thereof.
97. The method of any one of embodiments 44-96, wherein the nucleic acid encoding the heterologous antigen replaces UL82 or a homolog thereof.
98. A progeny CMV produced by the method of any one of embodiments 1-3, 5, 6, 8-34, 36, 37, 39, 40, 42, 43, 45-59, and 72-97.
99. A CMV viral vector produced by the method of any one of embodiments 4, 7, 8-34, 35, 38, 41, 44, 47-71, 96, or 97.
100. A mRNA molecule comprising the nucleotide sequence of SEQ ID NOs: 14-20.
101. A mRNA molecule comprising the nucleotide sequence of SEQ ID NOs: 4-10.
102. The mRNA molecule of embodiment 101, wherein each uridine is substituted with pseudouridine and each cytidine is substituted with 5-methylcytidine.
103. The mRNA molecule of embodiment 101, wherein each uridine is substituted with 5-methoxyuridine.
104. A mRNA molecule comprising the nucleotide sequence of SEQ ID NO: 21-27.
105. The mRNA molecule of any one of embodiments 100-104, wherein the mRNA molecule further comprises a poly(A) tail.
106. The mRNA molecule of any one of embodiments 100-104, wherein a poly(A) tail has been added to the 3′ end of the mRNA molecule.
107. The mRNA molecule of any one of embodiments 100-106, wherein the mRNA molecule was produced using a double-stranded DNA template encoding the poly(A) tail.
108. The mRNA molecule of claim 107, wherein the double-stranded DNA template is a plasmid.
109. The mRNA molecule of any one of embodiments 100-108, wherein the poly(A) tail is approximately 60-100 nucleotides long.
110. The mRNA molecule of any one of embodiments 100-109, wherein the poly(A) tail is 80 nucleotides long.
111. A mRNA molecule comprising a sequence according to SEQ ID NOs: 14-20, wherein each uridine is substituted with pseudouridine and each cytidine is substituted with 5-methylcytidine, and a poly(A) tail 80 nucleotides in length.
112. A mRNA molecule comprising a sequence according to SEQ ID NOs: 4-10, wherein each uridine is substituted with pseudouridine and each cytidine is substituted with 5-methylcytidine, and a poly(A) tail 80 nucleotides in length, wherein the mRNA molecule was produced using a plasmid template.
113. A mRNA molecule comprising a sequence according to SEQ ID NOs: 4-10, wherein each uridine is substituted with 5-methoxyuridine and the mRNA molecule a poly(A) tail 80 nucleotides in length, wherein the mRNA molecule was produced using a plasmid template.
114. A mRNA molecule comprising a sequence according to SEQ ID NOs: 21-27, wherein each uridine is substituted with 5-methoxyuridine and a poly(A) tail 80 nucleotides in length.
115. The mRNA molecule of embodiment 105, wherein the poly(A) tail has been added by an enzyme after transcription.
116. The mRNA molecule of embodiment 115, wherein the poly(A) tail is approximately 50-100 nucleotides long.
Transient Transfection of UL82 mRNA in Primary Fibroblasts Complements UL82-Deleted HCMV Vectors
Deletion of essential viral genes from vaccine vectors is a customary practice to ensure clinical safety. However, to produce the vector some method of complementation must be employed. Standard approaches involve creating stable cell lines that express the essential viral gene or its functional equivalent. This is complicated in situations such as HCMV that require primary normal diploid cells for virus production. An alternative approach is to utilize mRNA transfection to deliver the essential viral gene to the host cell. In this study, UL82 mRNA was transiently transfected into primary fibroblasts (MRC-5 cells) to complement HCMV vectors deleted for UL82.
HCMV UL82 expresses the major tegument protein pp71. One of the major functions of pp71 occurs at the onset of infection, where it is involved in the degradation of the cellular gene product, Daxx. In the absence of pp71, Daxx silences viral immediate-early (IE) gene expression mediated by histone deacetylases. However, this cellular protection mechanism is effectively neutralized when pp71 is transported to the nucleus where it can mediate proteasomal degradation of Daxx releasing the block to IE gene expression. HCMV vectors deleted for pp71 show a significant growth defect that prevents virus spreading and shedding in primate models. In combination with other safety modifications, pp71 deletion protects fetal rhesus macaques in a direct injection model of primary RhCMV infection.
To be able to produce HCMV vectors deleted for UL82, siRNA targeting DAXX may be transfected into host cells, in order to functionally complement for the absence of pp71. While this method is sufficient to produce virus, it primarily inhibits de novo Daxx production, and does not provide any of the other functions of pp71 likely to enhance the infection process such as cell cycle stimulation, efficient virion packaging, and virus stability.
The results demonstrate that mRNA transfection of the essential viral gene UL82 can provide functional complementation resulting in successful propagation of UL82-deleted HCMV virus vector. Use of UL82 mRNA transfection accelerates HCMV spread when compared to either mock transfection or functional complementation by anti-DAXX siRNA transfection as shown in
These results also suggest that UL82 mRNA transfection may enhance reconstitution of virus from BAC DNA. Experiments using a App71-GFP (UL82-deleted) virus construct indicated a more rapid progression of reconstitution from clonal BAC DNA.
The use of mRNA transfection for complementation is not limited to UL82 and could be extended to other essential HCMV genes. Transient transfection of mRNA could also be used to identify functions that permit HCMV to grow to higher titers by supplementing infection with combinatorial libraries of mRNAs from laboratory strains known to grow to high titer. In addition, pp71 protein present late in infection has the potential to be packaged in the progeny virus, which could lower the required dose of the vaccine by more efficient first-round infection and establishment of persistent infection.
The utility of transfected UL82 mRNA can also be applied to cell lines for use in determining the infectious titer of viral stocks. Current methods for viral titering involve the use of cell lines created to produce pp71 when induced by the addition of exogenous chemicals. The creation of these cell lines is labor and time intensive and the ability to complement function by transfection of mRNA could potentially save development time. MRC-5 and BJ-5ta cells transfected with UL82 mRNA display viral titers constant across a wide viral dilution series (
In addition, transfection of naturally permissive MRC-5 cells with UL82 mRNA leads to much higher apparent titers as compared to either transfection of BJ-5ta cells or the pp71 BJ-5ta cell line. This likely provides a more accurate titer of the material and allows better quantification of diluted material used in dose range studies. Transfection of UL82 mRNA should allow for lower titer viruses to be tested (e.g., less than 5e4 FFU/mL) with greater assay reproducibility and confidence.
Transient Transfection of UL82 mRNA for Complementation of UL82-Deleted HCMV Vectors
Complementation of UL82(pp71)-deleted vectors by providing the protein in trans, using pp71 mRNA transfection, was evaluated in another example. This approach may significantly reduce the dose and potentially stabilize the virus product with a full complement of tegument protein. These studies demonstrated reproducibility with additional operators, demonstrated titration of the mRNA transfection amount, and evaluated the pp71 mRNA transfection in the production process with HYPERStacks®.
Accelerated growth kinetics were observed for pp71-deleted viruses when pp71 mRNA was transfected into MRC-5 cells prior to infection. These experiments were conducted by transfecting 100 ng/cm2 of pp71 mRNA into cells seeded at 6.7×103 cells/cm2. To further improve virus growth kinetics, dose ranges of pp71 mRNA from 5 ng/cm2 to 500 ng/cm2 were investigated (
After the dose range experiments concluded, the 100 ng/cm2 condition was selected to move forward into the production process using HYPERStacks®. Seven runs with pp71 mRNA were performed in either HYPERStack-12s or HYPERStack-36s to confirm process scalability (
An immunoblot study was used to evaluate pp71 protein incorporation in the virion over a range of pp71 mRNA concentrations (
To evaluate whether the pp71 mRNA-expressed protein loaded into virions was functional, titers of pp71-deleted viruses in primary MRC-5 cells (no pp71 complementation) were compared to titers in a pBJ5TA fibroblast cell line (+pp71 complementation). A tissue culture infectious dose 50 (TCID50) titer assay was used for the uncomplemented MRC-5 cells and a late antigen immunofluorescence assay (LA IFA) was used for the pp71-complemented BJ5TA fibroblast cell line. In preliminary experiments, virus stocks produced with either anti-DAXX siRNA or pp71 mRNA were evaluated in both titer assays (Table 1). For pp71-deleted viruses produced with pp71 mRNA, the uncomplemented TCID50 titers were <1 log lower than the complemented LA IFA titers, similar to wildtype TR3 virus, suggesting that functional pp71 is incorporated in the virions. In contrast, the uncomplemented TCID50 titers for the pp71-deleted viruses produced with anti-DAXX siRNA were >2 log lower than the complemented LA IFA titers, indicating the lack of functional pp71. These results suggest that this comparative titer assay can be used to confirm pp71 protein function in pp71 mRNA-produced virus stocks and used in potency assays.
2TCID50 titer is determined in uncomplemented primary MRC-5 fibroblasts.
Development of a pp71 mRNA Transfection Process for pp71-Deleted HCMV Production
HCMV pp71 (UL82 ORF) may be deleted from vaccines to attenuate virus replication for improved safety. Pp71 is a 71 kDa tegument phosphoprotein delivered to cells upon viral entry. Pp71 has several roles, including immediate early regulation of viral gene expression, promotion of protein translation, and immune evasion by inhibiting intrinsic cellular factors (Kalejta 2020). For efficient virus production in vitro, pp71-deleted vectors require either direct or functional pp71 complementation. Functional complementation approaches use siRNA transfection to inhibit cellular Daxx expression. Knockdown of DAXX compensates for the absence of pp71 inhibition of intrinsic cell defenses (Cantrell 2006, Preston 2006, Saffert 2006, Woodhall 2006). However, direct pp71 complementation is desirable for its potential to enhance immediate early (IE) activation compared to anti-DAXX siRNA knockdown, as well as complementing other functions of pp71.
As shown in this Example, growth of the pp71-deleted backbone vector is accelerated upon transfection of pp71 mRNA compared to the previous transfection process with anti-DAXX siRNA. Direct pp71 complementation also has the potential to load HCMV virions with pp71 protein which may lower vaccine dose by increasing the efficiency of the first round of vector replication.
Complementation of pp71 (UL82)-deleted vectors by providing the protein in trans has demonstrated the potential for an immunogenic dose reduction in the Rhesus CMV model (Marshall 2019). By adapting the anti-DAXX siRNA transfection process to pp71 mRNA complementation, the dose may be significantly reduced, providing manufacturing and clinical benefit, and potentially stabilizing the virus product with a full complement of tegument protein. Vaccine culture systems can utilize a pp71 complementing producer cell line, or anti-DAXX siRNA transfection can be replaced by the transient transfection of pp71 mRNA, whose development and implementation will be described in this report.
Pp71-deleted HCMV production via anti-DAXX siRNA or pp71 mRNA are both based on transfecting cells using a lipid-based transfection reagent allowing entry of the nucleic acid into cells, followed by infection. The process flow for producing pp71 deleted HCMV with anti-DAXX siRNA is as follows:
The improvement to viral replication that pp71 protein enables allows for omission of the second transfection step, which is a process improvement for production because it removes a full step of the process. Another process improvement that pp71 mRNA transfection enables is culture harvest at an earlier DPI, approximately one week. The process flow for producing pp71 deleted HCMV with pp71 mRNA is as follows:
In T-flasks, MRC-5 fibroblasts are transfected on day 3 post-seeding (>70% confluent) when cells are seeded at 6.7×103 cells per cm2. In HYPERStacks, MRC-5 fibroblasts are transfected on day 4 post-seeding (>85% confluent) when cells are seeded at 6.7×103 cells per cm2.
The protocol for transfecting pp71 mRNA is as follows:
The day after transfection, vessels are infected with pp71-deleted HCMV at a MOI of 0.01. On day 5 post-infection, the complete growth media (CGM; DMEM containing 9% FBS and 2 mM GlutaMax) is replaced with reduced serum media (RSM; DMEM containing 0.2% FBS and 2 mM GlutaMax).
The GMP produced pp71 mRNA construct to be used in manufacturing is fully substituted with pseudouridine (pseudoU) and 5-methylcytidine (5meC), contains the natural HCMV UL82 ORF 5′ and 3′ UTRs, and is produced with a plasmid template 80 nucleotide poly-A tail (see
5.1 Generation of V5-Tagged pp71 mRNA Constructs and Detection of V5 Expression in Cell Lysates
A pp71 mRNA construct was designed from the start codon of the HCMV UL82 open reading frame to the stop codon with a V5 tag, a synthetic 5′UTR and a mouse α-globin 3′UTR (
To determine if pp71 protein was detectable in MRC-5 fibroblasts after transfection of pp71-V5 mRNA, immunoblot and immunofluorescence analyses were performed. MRC-5 fibroblasts were transfected with 50 ng/cm2 of pp71-V5 mRNA using the transfection reagent Lipofectamine 2000 (used for anti-DAXX siRNA transfection). For immunoblots, a cell pellet time-course was collected from day 1 to day 6 post-transfection and resuspended in the lysis buffer modified RIPA buffer plus protease inhibitors. A BCA protein assay was performed to normalize protein loading, and 50 μg total protein was loaded per sample on a NuPAGE™ 4-12% Bis-Tris gel. An MRC-5 cell lysate was used as a negative cell control sample. Immunoblot analysis of the cell lysates using a V5 antibody shows expression of pp71-V5 protein out to day 6 post-transfection (
An immunofluorescence assay (IFA) was performed on MRC-5 fibroblasts transfected with 50 ng/cm2 of pp71-V5 mRNA at 48 hours post-transfection. Cells were fixed with 4% formaldehyde in PBS and stained with a primary anti-V5 tag antibody followed by a Cy-5 labeled secondary antibody. Pp71-V5 protein is expressed and localized to the nuclear region, which is counterstained with the nuclear stain DAPI (
After confirming that the transfected pp71-V5 mRNA expressed pp71-V5 protein in MRC-5 fibroblasts, functional analyses of the pp71-V5 protein were performed via pp71-deleted HCMV replication assessment. Preliminary infection data indicated that viral cytopathic effect (CPE) progressed faster with transfection of pp71 mRNA compared to anti-DAXX siRNA. The pp71-deleted HCMV production process using anti-DAXX siRNA for complementation consists of two 10 μM anti-DAXX siRNA transfections, one the day before infection (DPI-1) and the second anti-DAXX siRNA transfection at DPI 10. Considering the immunoblot analysis showed pp71 protein levels decreasing at day 6 post-transfection (
To further evaluate and quantitate the effect of pp71 mRNA complementation to viral replication, viral growth curves were performed. Briefly, MRC-5 fibroblasts were transfected with 10 μM anti-DAXX siRNA (−1 and 10 DPI) or transfected with 40 ng/cm2 pp71-V5 mRNA (−1 and 6 DPI) prior to infection with pp71-deleted HCMV at MOI 0.01. Viral supernatant was harvested at multiple days post-infection and titered using the HCMV LA Immunofluorescence Titering Assay (TMD-QC-0061 v4.0). Virus was detected earlier with pp71 mRNA complementation (7 DPI vs 9 DPI), and the peak titer occurred earlier (14 DPI vs 17 DPI) when compared to virus grown using anti-DAXX siRNA transfection (
Next, whether pp71 protein from transfection was being incorporated into HCMV virions was determined. To assay for protein contained in virions, infected cell supernatant is collected at full CPE. The viral supernatant is first centrifuged at low speed (2,500×g, 15 min) to remove cellular debris, followed by high-speed centrifugation (24,000 RPM, 1 hr) to pellet virions through a 20% sorbitol cushion. The virion pellet is resuspended in lysis buffer (modified RIPA buffer plus protease inhibitors) and assayed by immunoblot. Previous data showed a lack of pp71-V5 protein expression when MRC-5 fibroblasts were transfected with 40 ng/cm2 pp71-V5 mRNA at −1 and 6 DPI and infected with pp71-deleted HCMV at MOI 0.01, potentially due to the reduced expression of pp71-V5 protein by day 6 post-transfection shown in
To investigate the lack of pp71 protein expression in cell lysates at late times during viral infection, the following actions were taken:
Several transfection agents were evaluated for transfection efficiency in MRC-5 fibroblasts. An EGFP mRNA similarly constructed to the pp71-V5 mRNA was used for testing. Cells were transfected with three amounts of mRNA (low, medium, high) using four transfection reagents, including Lipofectamine 2000 (ThermoFisher), MessengerMax (ThermoFisher), Jet-mRNA (PolyPlus) and Trans-IT (Mirus Bio) at four lipid levels and evaluated by flow cytometry for EGFP expression (
All transfections were performed according to manufacturer instructions. All subsequent mRNA transfections in MRC-5 fibroblasts for virus production used the MessengerMax transfection reagent.
5.3 Additional Pp71 mRNA Constructs
To address the lack of detectable pp71 protein at late times during HCMV infection, even after transfecting pp71 mRNA multiple times, the possibility that HCMV infection inhibits lipid-based mRNA transfection was considered. Utilizing an EGFP mRNA similarly constructed to the pp71 mRNA also containing a Cy5 tag allowed the visualization of the transfected mRNA as well as EGFP protein. MRC-5 fibroblasts were transfected with 50 ng/cm2 EGFP-Cy5 mRNA at DPI-1 and DPI 6 and infected with WT TR3 at a MOI of 0.01 at DPI 0. The Cy5 signal was present during infection, but EGFP signal was lost at late times during infection (
Transfection of cells with unmodified RNAs can lead to cell death due to activation of innate immune pathways (Devoldere 2016). The following experiment addressed whether modification of the pp71 mRNA construct can stabilize the mRNA for better expression in MRC-5 fibroblasts. Stabilizers can include (Devoldere 2016):
For example, it has been shown that substitution of uridine and cytidine residues with pseudouridine and 5-methylcytidine reduces innate immune recognition and that pseudouridine modified RNA is translated more efficiently (McCaffrey 2017, Kariko 2008). All pp71 mRNA constructs used in
Utilizing EGFP mRNA, but with different nucleoside modifications, the expression of EGFP was compared in transfected cells with and without HCMV infection (WT TR3 at MOI 0.01). The following EGFP mRNA constructs were transfected at DPI-1 in MRC-5 fibroblasts.
Note the Cy5-UTP: 5moU is Cy-5 labeled uridine triphosphate at a 1:3 ratio to 5moU and translation efficiency correlates inversely with Cy5-UTP substitution. EGFP mRNA was transfected at 100 ng/cm2, doubling the amount previously used (
To explore the possibility that the lack of pp71 protein incorporation into the virion is due to exogenous pp71 not localizing to the correct cellular location additional pp71 constructs were designed (
All constructs were transfected at 100 ng/cm2 in MRC-5 fibroblasts with and without infection using a pp71-deleted vector at a MOI of 0.01 and cell lysates were assayed for pp71 protein expression by immunoblot (
There are two production methods (TriLink Biotechnologies) to add the poly-A tail to the pp71 mRNA construct-either via (1) a plasmid template encoded tail designed to add 80 nucleotides, or (2) enzymatically, which variably adds approximately 60-100 nucleotides. A poly(A) tail was added via plasmid template to the start-to-stop pp71 mRNA construct, Construct A, with and without the V5 tag. Due to a manufacturing error the first lot of full-length pp71 mRNA with the viral 5′ and 3′ UTRs modified with pseudoU/5meC was produced with no poly-A tail. This lot of mRNA was tailed in-house enzymatically synthesizing an unknown tail length but functional protein by immunoblot (
Multiple growth curves were also performed comparing pp71 mRNA lots produced with poly-A tails by template or enzymatically (
5.4 pp71 mRNA Titration
Considering the changes made to the pp71 mRNA transfection process as well as the positive pp71 protein signal visualized by immunoblot in pp71-deleted infected cell lysates (
To further improve pp71-deleted virus production, growth curves were performed comparing a range of pp71 mRNA transfected from 5 ng/cm2 to 500 ng/cm2 as well as comparing the start-to-stop construct (SS) to the full-length construct with the viral 5′ and 3′ UTRs (FT) (
2TCID50 titer is determined in uncomplemented primary MRC-5 fibroblasts.
To confirm that the pp71 mRNA transfection method at 100 ng/cm2 was scalable into HYPERStack format, seven runs were performed in either HYPERStack-12s or HYPERStack-36s. Three growth curves were performed in HYPERStack-12s following the process developed in T-flasks (
Four growth curves performed in HYPERStack-12s and HYPERStack-36s transfected on day 4 post-seeding or greater than 85% confluency and infected on day 5 post-seeding with a pp71 deleted virus at a MOI of 0.01 resulted in LA IFA titers greater than 1×106 FFU/mL by 12 to 14 DPI (
In some HCMV vectors used to express heterologous genes, such as an antigen, the gene of interest replaces the UL82 ORF. An immunoblot was performed to investigate if the process change (producing pp71-deleted vectors via pp71 mRNA transfection), affects antigen expression from the UL82 (pp71) locus (
The next set of experiments were designed to assess whether the pp71 mRNA expressed protein loaded into virions is functional. Four virus stocks were produced by transfecting MRC-5 fibroblasts with pp71 mRNA at 25, 50, 100 and 200 ng/cm2 and infecting with a pp71-deleted virus at a MOI of 0.01. The virus stock used to infect these cultures was produced with the anti-DAXX siRNA process, therefore virions used for infection did not contain any pp71 protein.
First, uncomplemented MRC-5 fibroblasts were infected over a range of MOIs with a pp71-deleted virus grown either in the presence of 10 μM anti-DAXX siRNA or pp71 mRNA at 200 ng/cm2 (
Concurrently, a tissue culture infectious dose 50 (TCID50) titer assay in uncomplemented MRC-5 fibroblasts was evaluated using the virus stocks produced with 25, 50, 100 or 200 ng/cm2 pp71 mRNA compared to a virus stock produced with the anti-DAXX siRNA process as well as WT TR3 (Table 2). TCID50 titers were compared to titers from the late antigen immunofluorescence assay (TMD-QC-0061) in the pp71-complemented BJ-5TA fibroblast cell line. The log difference between the titer assays shows that as the amount of pp71 mRNA transfected is reduced the functional complementation is also reduced. The log difference for a virus stock produced with 25 ng/cm2 pp71 mRNA is more similar to the log difference for a virus stock produced with anti-DAXX siRNA (>2 logs), whereas the log difference for virus stocks produced with at least 100 ng/cm2 pp71 mRNA is less than 2 logs.
To further test the TCID50 assay results, 5 replicate TCID50s were performed for two viruses produced with DAXX anti-siRNA and two viruses produced with 100 ng/cm2 pp71 mRNA. The two vectors were TB deleted: TR3 mir124 ΔUL128-130 ΔUL146-147 ΔUL82 Ag85A-ESAT-6-Rv3407-Rv2626c-RpfA-RpfD and Vector 2: TR3 Δ146-147 Δ128-130 ΔUL82 M conserved gag/nef/pol fusion episensus 1 ΔUL18. Average titers were compared between TCID50s in uncomplemented MRC-5 fibroblasts and the current LA IFA using the pp71-complemented BJ5TA fibroblast cell line. In pp71-deleted viruses produced with pp71 mRNA, the uncomplemented TCID50 titers were less than 1 log lower than the complemented LA IFA, similar to WT TR3 (see Table 1), suggesting that functional pp71 is incorporated in the virions. In contrast, the uncomplemented TCID50 titers for the pp71-deleted viruses produced with anti-DAXX siRNA were greater than 2 logs lower than the complemented LA IFA titers, indicating the lack of functional pp71. These results suggest that this comparative titer assay can be used to confirm pp71 protein function in pp71 mRNA produced virus stocks. The range of TCID50 titers for pp71-produced TB deleted was 3.5×105 to 7.4×105 FFU/mL and for pp71-produced Vector 2 was 4.1×105 to 7.4×105 FFU/mL. While the range of TCID50 titers for DAXX-produced TB deleted was 8.7×103 to 1.1×104 FFU/mL and for DAXX-produced Vector 2 was 1.2×103 to 1.9×103 FFU/mL.
A series of experiments were performed to find controls and conditions for an immunoblot assay to detect exogenous pp71 protein in HCMV vector virions. Virion pp71 protein detected by immunoblot thus far could not be distinguished from cell membrane fragments or vesicles pelleted with virions by ultracentrifugation after transfection of MRC-5 fibroblasts with pp71 mRNA. Several attempts were made to create an ultracentrifuged non-infected transfection control sample to confirm pp71 protein incorporation into virions.
Negligible protein was released from pp71 mRNA transfected MRC-5 monolayers that were not been infected. Three freeze/thaw cycles were used to mimic an infected lysed MRC-5 monolayer. An alternative approach to isolate virions prior to immunoblot analysis was developed.
A two-step sucrose gradient was used to isolate virions from the T-flask production process along with transfection control samples and samples for an assay for pp71 protein by immunoblot (Dai 2014).
pp71 mRNA transfection control gradient purified lysate was obtained by following the pp71 mRNA production process in T-flasks but omitting infection. The monolayer was scraped in the media at DPI 14, collected, and subjected to three freeze/thaw cycles. The infected sample was obtained by following the pp71 mRNA production process in T-flasks, the supernatant was collected separately from the cell pellet at DPI 14. Three T150s per sample type were combined to allow for enough material to be loaded onto the gradient. Both supernatant samples were clarified by centrifugation at 5,000×g for 15 min. The clarified supernatants were ultracentrifuged at 21,000 RPM for 1 h at 15° C. and resuspended in 2 mL of PBS at pH 7.4.
The sucrose gradient protocol was as follows. In a SW41 Ti Beckman ultra-rotor, 5 mL of 50% sucrose in PBS was overlayed with 5 ml of 15% sucrose in PBS. Next, 2 mL of each sample type in PBS was overlayed. Samples were spun at 21,000 RPM for 1 hour at 15° C. and the ultracentrifuge deacceleration was set to coast. Two bands were seen in the infected sample gradient at the interface between the sucrose layers. The lower band was narrower than the upper band and the bands were too close together to collect separately. The upper band was predicted to contain more virions, while the lower band likely contained more dense bodies, but the majority of particles in both bands were likely virions with DNA-filled capsids (Dai 2014). Visually there was one band in the transfection control gradient purified sample, which was also at the interface of the sucrose layers. Approximately 1 mL was collected from each gradient and diluted with PBS to a total volume of 12 mL. A final spin to concentrate the samples was performed in the SW41 Ti at 21,000 RPM for 1 h at 15 C. The pellet was resuspended in 100 uL lysis buffer.
An immunoblot showed pp71 protein present in the transfection-only cell lysate control (
The gradient immunoblot was repeated with increased clarification speed at 10,000×g (data not shown). This resulted in less total protein recovered from the control transfection only gradient purified sample. While these experiments did not provide evidence that pp71 was incorporated in the virion, optimizing the separation of cellular debris from virions and/or using an alternative assay could provide further information.
While specific embodiments have been illustrated and described, it will be readily appreciated that the various embodiments described above can be combined to provide further embodiments, and the various embodiments described above can be combined to provide further embodiments.
All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications, and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, including U.S. Provisional Patent Applications No. 63/239,269 filed Aug. 31, 2021 is incorporated herein by reference, in its entirety, unless explicitly stated otherwise. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications, and publications to provide yet further embodiments.
These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2022/075647 | 8/30/2022 | WO |
Number | Date | Country | |
---|---|---|---|
63239269 | Aug 2021 | US |