MODIFIED SEROTYPE 28 ADENOVIRAL VECTORS

- GenVec, Inc.

The invention provides a replication-deficient serotype 28 adenoviral vector characterized by comprising a portion of a serotype 45 adenoviral hexon protein and/or a portion of a serotype 45 fiber protein in place of the endogenous serotype 28 hexon and/or fiber protein.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This patent application is a continuation of co-pending U.S. application Ser. No. 15/700,279, filed Sep. 11, 2017, which is a continuation of U.S. application Ser. No. 14/403,651, filed Nov. 25, 2014, now U.S. Pat. No. 9,790,519, which is a 371 of International Patent Application No. PCT/US2013/042824, filed May 28, 2013, which claims the benefit of U.S. Provisional Patent Application No. 61/652,407, filed May 29, 2012.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT

This invention was made with Government support under Grant Number 5R43AI077147-02 awarded by the National Institutes of Health. The Government has certain rights in this invention.

INCORPORATION-BY-REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY

Incorporated by reference in its entirety herein is a computer-readable nucleotide/amino acid sequence listing submitted concurrently herewith and identified as follows: One 79,166 Byte ASCII (Text) file named “753312_ST25.TXT,” created on Mar. 12, 2021.

BACKGROUND OF THE INVENTION

In vivo delivery of proteins in biologically relevant forms and amounts has been an obstacle to drug and vaccine development for decades. One solution that has proven to be a successful alternative to traditional protein delivery approaches is the use of exogenous nucleic acid sequences for production of proteins in vivo. Gene transfer vectors ideally enter a wide variety of cell types, have the capacity to accept large nucleic acid sequences, are safe, and can be produced in quantities required for treating patients. Viral vectors are gene transfer vectors with these advantageous properties (see, e.g., Thomas et al., Nature Review Genetics, 4: 346-358 (2003)). Furthermore, while many viral vectors are engineered to infect a broad range of cell types, viral vectors also can be modified to target specific cell types, which can enhance the therapeutic efficacy of the vector (see, e.g., Kay et al., Nature Medicine, 7(1): 33-40 (2001)).

Viral vectors that have been used with some success to deliver exogenous proteins to mammalian cells for therapeutic purposes include, for example, Retrovirus (see, e.g., Cavazzana-Calvo et al., Science, 288 (5466): 669-672 (2000)), Lentivirus (see, e.g., Cartier et al., Science, 326: 818-823 (2009)), Adeno-associated virus (AAV) (see, e.g., Mease et al., Journal of Rheumatology, 27(4): 692-703 (2010)), Herpes Simplex Virus (HSV) (see, e.g., Goins et al., Gene Ther., 16(4): 558-569 (2009)), Vaccinia Virus (see, e.g., Mayrhofer et al., J. Virol., 83(10): 5192-5203 (2009)), and Adenovirus (see, e.g., Lasaro and Ertl, Molecular Therapy, 17(8): 1333-1339 (2009)).

Despite their advantageous properties, widespread use of viral gene transfer vectors is hindered by several factors. In this respect, certain cells are not readily amenable to gene delivery by currently available viral vectors. For example, lymphocytes are impaired in the uptake of adenoviruses (Silver et al., Virology, 165: 377-387 (1988), and Horvath et al., J. Virology, 62(1): 341-345 (1988)). In addition, viral vectors that integrate into the host cell's genome (e.g., retroviral vectors) have the potential to cause insertion mutations in oncogenes (see, e.g., Cavazzana-Calvo et al., supra, and Hacein-Bey-Abina et al., N. Engl. J Med., 348: 255-256 (2003)).

The use of viral vectors for gene transfer also is impeded by the immunogenicity of viral vectors. A majority of the U.S. population has been exposed to wild-type forms of many of the viruses currently under development as gene transfer vectors (e.g., adenovirus). As a result, much of the U.S. population has developed pre-existing immunity to certain virus-based gene transfer vectors. Such vectors are quickly cleared from the bloodstream, thereby reducing the effectiveness of the vector in delivering biologically relevant amounts of a gene product. Moreover, the immunogenicity of certain viral vectors prevents efficient repeat dosing, which can be advantageous for “boosting” the immune system against pathogens when viral vectors are used in vaccine applications, thereby resulting in only a small fraction of a dose of the viral vector delivering its payload to host cells.

Thus, there remains a need for improved viral vectors that can be used to efficiently deliver genes to mammalian cells in vivo. The invention provides such viral vectors.

BRIEF SUMMARY OF THE INVENTION

The invention provides a replication-deficient serotype 28 adenoviral vector comprising one or both of the following: (a) at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein, and (b) at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein.

BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S)

FIG. 1 is a flow diagram of the construction of plasmid pAC28UL19.H (45) by homologous recombination. Steps 1 through 10 illustrate the homologous recombination between the Ad28 viral vector genome plasmid and the shuttle plasmid containing the Ad45 hexon followed by subsequent recombination containing the CMVTetO.UL19 expression cassette as described in Example 1.

FIG. 2 is a graph depicting experimental data of CD8+ T-cell responses induced by serotype 5 and 28 adenoviral vectors encoding an HSV2 UL19 protein (Ad5 UL19 and Ad28 UL19, respectively), as well as the mutant serotype 28 adenoviral vector encoding an HSV2 UL19 protein (Ad28 H/F UL19) described in Example 2.

DETAILED DESCRIPTION OF THE INVENTION

The invention is predicated, at least in part, on the generation of an adenoviral vector based on a serotype 28 adenovirus (Ad28) containing one or more modified capsid proteins, which can effectively be used to deliver and express nucleic acid sequences encoding therapeutic proteins. It is believed that a vector based on Ad28 will provide improved delivery to human cells because Ad28 seroprevalence is low in human populations. It is also believed that a serotype 28 adenoviral vector containing a modified hexon protein and/or a modified fiber protein as described herein can stimulate a more robust T-cell response than an Ad28 vector comprising wild-type hexon and fiber proteins when used in vaccine applications.

A serotype 28 adenovirus is a member of the group D adenoviruses. In addition to Ad28, the group D adenoviruses include the following serotypes: 8, 9, 10, 13, 15, 17, 19, 20, 22, 23, 24, 25, 26, 27, 29, 30, 32, 33, 36, 37, 38, 39, 42, 43, 44, 45, 46, 47, 48, 49, 51, 53, 54, and 56 (see, e.g., Gema et al., J. Infectious Diseases, 145(5): 678-682 (1982), and Robinson et al., Virology, 409(2): 141-147 (2011)). The genome of human Ad28 has been sequenced and is available via the GenBank database (Accession No. FJ824826.1).

The term “adenoviral vector,” as used herein, refers to an adenovirus in which the adenoviral genome has been manipulated to accommodate a nucleic acid sequence that is non-native with respect to the adenoviral genome. Adenovirus is a medium-sized (90-100 nm), nonenveloped icosohedral virus containing approximately 36 kb of double-stranded DNA. There are 49 human adenoviral serotypes, categorized into 6 subgenera (A through F) based on nucleic acid comparisons, fiber protein characteristics, and biological properties (Crawford-Miksza et al., J. Virol., 70: 1836-1844 (1996)). The group C viruses (e.g., serotypes 2 and 5, or Ad2 and Ad5) are well characterized, and currently are employed for gene transfer studies, including human gene therapy trials (see, e.g., Rosenfeld et al., Science, 252: 431-434 (1991); Rosenfeld et al., Cell, 68: 143-155 (1992); Zabner, Cell, 75: 207-216 (1993); Crystal et al., Nat. Gen., 8: 42-51 (1994); Yei et al., Gene Therapy, 1: 192-200 (1994); Chen et al., Proc. Natl. Acad. Sci., 91: 3054-3057 (1994); Yang et al., Nat. Gen., 7: 362-369 (1994); Zabner et al., Nat. Gen., 6: 75-83 (1994)). Typically, an adenoviral vector is generated by introducing one or more mutations (e.g., deletion, insertion, or substitution) into the adenoviral genome of the adenovirus so as to accommodate the insertion of a non-native nucleic acid sequence, for example, for gene transfer, into the adenovirus.

The adenovirus capsid mediates the key interactions of the early stages of the infection of a cell by the virus, and is required for packaging adenovirus genomes at the end of the adenovirus life cycle. The capsid comprises 252 capsomeres, which includes 240 hexons, 12 penton base proteins, and 12 fibers (Ginsberg et al., Virology, 28: 782-83 (1966)). In one embodiment of the invention, one or more capsid proteins (also referred to herein as “coat” proteins) of the adenoviral vector can be manipulated to alter the binding specificity or recognition of the vector for a viral receptor on a potential host cell. Such manipulations can include deletion of regions of the fiber or penton, insertions of various native or non-native ligands into portions of the capsid proteins, and the like. Manipulation of capsid proteins can broaden the range of cells infected by the adenoviral vector or enable targeting of the adenoviral vector to a specific cell type.

The adenoviral vector of the invention can comprise a modified hexon protein. The adenovirus hexon protein is the largest and most abundant protein in the adenovirus capsid. The hexon protein is essential for virus capsid assembly, determination of the icosahedral symmetry of the capsid (which in turn defines the limits on capsid volume and DNA packaging size), and integrity of the capsid. In addition, the hexon protein is a primary target for modification in order to reduce neutralization of adenoviral vectors (see, e.g., Gall et al., J. Virol., 72: 10260-264 (1998), and Rux et al., J. Virol., 77(17): 9553-9566 (2003)). The major structural features of the hexon protein are shared by adenoviruses across serotypes, but the hexon protein differs in size and immunological properties between serotypes (Jornvall et al., J. Biol. Chem., 256(12): 6181-6186 (1981)). A comparison of 15 adenovirus hexon proteins reveals that the predominant antigenic and serotype-specific regions of the hexon protein appear to be in loops 1 and 2 (i.e., LI or l1, and LII or l2, respectively), within which are seven to nine discrete hypervariable regions (HVR1 to HVR 7 or HVR9) varying in length and sequence between adenoviral serotypes (Crawford-Miksza et al., J. Virol., 70(3): 1836-1844 (1996), and Bruder et al., PLoS ONE, 7(4): e33920 (2012)).

The hexon protein is “modified” in that it comprises a non-native amino acid sequence in addition to or in place of a wild-type hexon amino acid sequence of the serotype 28 adenoviral vector. In this respect, at least a portion of the wild-type hexon protein (e.g., the entire hexon protein) of the inventive serotype 28 adenoviral vector desirably is removed and replaced with a corresponding portion of a hexon protein from a non-group D adenovirus (e.g., a group A, B, C, E, or F adenovirus), or an adenovirus that does not normally infect humans (e.g., a simian or gorilla adenovirus). Alternatively and preferably, at least a portion of the wild-type hexon protein of the serotype 28 adenoviral vector can be removed and replaced with a corresponding portion of a hexon protein from another group D adenovirus. For example, a portion of the wild-type hexon protein of the serotype 28 adenoviral vector can be removed and replaced with a corresponding portion of a hexon protein from any group D adenovirus (such as those described herein). Preferably, the inventive serotype 28 adenoviral vector comprises at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein. Any suitable amino acid residue(s) of a wild-type hexon protein of the serotype 28 adenoviral vector can be modified or removed, so long as viral capsid assembly is not impeded. Similarly, amino acids can be added to the hexon protein as long as the structural integrity of the capsid is maintained. In a preferred embodiment, at least 50% (e.g., at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100%) of the endogenous Ad28 hexon protein is modified or removed.

A “portion” of an amino acid sequence comprises at least three amino acids (e.g., about 3 to about 800 amino acids). Preferably, a “portion” comprises 10 or more (e.g., 10 or more, 15 or more, 20 or more, 25 or more, 30 or more, 40 or more, 50 or more, or 100 or more) amino acid residues, but less than the entire wild-type hexon protein (e.g., 900 or less, 800 or less, 700 or less, 600 or less, 500 or less, 400 or less, 300 or less, 200 or less, or 100 or less amino acid residues). For example, a portion can be about 10 to about 700 amino acids (e.g., about 10, 100, 300, 500, or 600 amino acids), about 10 to about 500 amino acids (e.g., about 20, 50, 200, or 400 amino acids), or about 10 to about 300 amino acids (e.g., about 15, 40, 60, 70, 90, 150, 250, or 290 amino acids), or a range defined by any two of the foregoing values. More preferably, a “portion” comprises no more than about 600 amino acids (e.g., about 10 to about 550 amino acids, about 10 to about 500 amino acids, or about 50 to about 300 amino acids, or a range defined by any two of the foregoing values).

A “portion” of a nucleic acid sequence comprises at least ten nucleotides (e.g., about 10 to about 5000 nucleotides). Preferably, a “portion” of a nucleic acid sequence comprises 10 or more (e.g., 15 or more, 20 or more, 25 or more, 30 or more, 35 or more, 40 or more, 45 or more, 50 or more, or 100 or more) nucleotides, but less than 5,000 (e.g., 4900 or less, 4000 or less, 3000 or less, 2000 or less, 1000 or less, 800 or less, 500 or less, 300 or less, or 100 or less) nucleotides. Preferably, a portion of a nucleic acid sequence is about 10 to about 3500 nucleotides (e.g., about 10, 20, 30, 50, 100, 300, 500, 700, 1000, 1500, 2000, 2500, or 3000 nucleotides), about 10 to about 1000 nucleotides (e.g., about 25, 55, 125, 325, 525, 725, or 925 nucleotides), or about 10 to about 500 nucleotides (e.g., about 15, 30, 40, 50, 60, 70, 80, 90, 150, 175, 250, 275, 350, 375, 450, 475, 480, 490, 495, or 499 nucleotides), or a range defined by any two of the foregoing values. More preferably, a “portion” of a nucleic acid sequence comprises no more than about 3200 nucleotides (e.g., about 10 to about 3200 nucleotides, about 10 to about 3000 nucleotides, or about 30 to about 500 nucleotides, or a range defined by any two of the foregoing values).

Desirably, the portion of an adenovirus serotype 45 hexon protein comprises at least one hypervariable region (HVR) in place of an endogenous Ad28 HVR. Thus, at least one HVR of the hexon protein of the inventive serotype 28 adenoviral vector is removed and replaced with at least one HVR from a wild-type serotype 45 adenovirus. In one embodiment, the serotype 28 adenoviral vector can comprise one or more of HVR1, HVR2, HVR3, HVR4, HVR5, HVR6, HVR7, HVR8, or HVR9 of a wild-type serotype 45 adenovirus hexon protein in place of the corresponding endogenous Ad28 HVR. Preferably, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or 9) HVRs of the hexon protein of the serotype 28 adenoviral vector are removed and replaced with corresponding HVRs from a serotype 45 adenovirus. More preferably, the inventive serotype 28 adenoviral vector comprises all nine HVRs of a serotype 45 adenovirus hexon protein in place of the corresponding endogenous Ad28 HVRs. In one embodiment, the entire wild-type hexon protein of the serotype 28 adenoviral vector is replaced with the entire hexon protein of a serotype 45 adenovirus.

Nucleic acid sequences that encode all or a portion of a serotype 28 or 45 adenovirus hexon protein are publicly available (see, e.g., GenBank Accession Nos. DQ149626.1 and AB330126.1). Amino acid sequences that comprise a full-length serotype 28 or 45 adenovirus hexon protein, or portions thereof, also are publicly available (see, e.g., GenBank Accession Nos. ABA00010.1 and BAG48822). In one embodiment, the portion of an adenovirus serotype 45 hexon protein comprises, for example, the amino acid sequence of SEQ ID NO: 1, and a nucleic acid sequence that encodes a portion of a serotype 45 adenovirus hexon protein comprises, for example, SEQ ID NO: 2. In another embodiment, the portion of an adenovirus serotype 45 hexon protein comprises an amino acid sequence that is at least 91.4% identical (e.g., at least 91.5% identical, at least 92% identical, at least 92.5% identical, at least 93% identical, at least 93.5% identical, at least 94% identical, at least 94.5% identical, at least 95% identical, at least 95.5% identical, at least 96% identical, at least 96.5% identical, at least 97% identical, at least 97.5% identical, at least 98% identical, at least 98.5% identical, at least 99% identical, or at least 99.5% identical) to SEQ ID NO: 1.

Nucleic acid or amino acid sequence “identity,” as described herein, can be determined by comparing a nucleic acid or amino acid sequence of interest to a reference nucleic acid or amino acid sequence. The number of nucleotides or amino acid residues that have been changed and/or modified (such as, e.g., by point mutations, insertions, or deletions) in the reference sequence so as to result in the sequence of interest are counted. The total number of such changes is subtracted from the total length of the sequence of interest, and the difference is divided by the length of the sequence of interest and expressed as a percentage. A number of mathematical algorithms for obtaining the optimal alignment and calculating identity between two or more sequences are known and incorporated into a number of available software programs. Examples of such programs include CLUSTAL-W, T-Coffee, and ALIGN (for alignment of nucleic acid and amino acid sequences), BLAST programs (e.g., BLAST 2.1, BL2SEQ, and later versions thereof) and FASTA programs (e.g., FASTA3x, FASTM, and SSEARCH) (for sequence alignment and sequence similarity searches). Sequence alignment algorithms also are disclosed in, for example, Altschul et al., J. Molecular Biol., 215(3): 403-410 (1990); Beigert et al., Proc. Nat. Acad. Sci. USA, 106(10): 3770-3775 (2009); Durbin et al., eds., Biological Sequence Analysis: Probalistic Models of Proteins and Nucleic Acids, Cambridge University Press, Cambridge, U.K. (2009), Soding, Bioinformatics, 21(7): 951-960 (2005); Altschul et al., Nucleic Acids Res., 25(17): 3389-3402 (1997); and Gusfield, Algorithms on Strings, Trees and Sequences, Cambridge University Press, Cambridge, U.K. (1997)).

The adenoviral vector of the invention can comprise a modified fiber protein. The adenovirus fiber protein is a homotrimer of the adenoviral polypeptide IV that has three domains: the tail, shaft, and knob (Devaux et al., J. Molec. Biol., 215: 567-88 (1990), and Yeh et al., Virus Res., 33: 179-98 (1991)). The fiber protein mediates primary viral binding to receptors on the cell surface via the knob and the shaft domains (Henry et al., J. Virol., 68(8): 5239-46 (1994)). The amino acid sequences for trimerization are located in the knob, which appears necessary for the amino terminus of the fiber (the tail) to properly associate with the penton base (Novelli et al., Virology, 185: 365-76 (1991)). In addition to recognizing cell receptors and binding the penton base, the fiber contributes to serotype identity. Fiber proteins from different adenoviral serotypes differ considerably (see, e.g., Green et al., EMBO J., 2: 1357-65 (1983), Chroboczek et al., Virology, 186: 280-85 (1992), and Signas et al., J. Virol., 53: 672-78 (1985)). Thus, the fiber protein has multiple functions that are key to the life cycle of adenovirus.

The fiber protein is “modified” in that it comprises a non-native amino acid sequence, in addition to or in place of a wild-type fiber amino acid sequence of the inventive serotype 28 adenoviral vector. In this respect, a portion of the wild-type fiber protein (e.g., the fiber tail, the fiber shaft, the fiber knob, or the entire fiber protein) of the inventive serotype 28 adenoviral vector can be removed and replaced with a corresponding portion of a fiber protein from a non-group D adenovirus (e.g., a group A, B, C, E, or F adenovirus), or an adenovirus that does not infect humans (e.g., a simian or gorilla adenovirus). Alternatively and preferably, at least a portion of the wild-type fiber protein of the inventive serotype 28 adenoviral vector can be removed and replaced with a corresponding portion of a fiber protein from another group D adenovirus. For example, a portion of the wild-type fiber protein of the inventive serotype 28 adenoviral vector can be removed and replaced with a corresponding portion of a fiber protein from any group D adenovirus (such as those described herein). Preferably, the inventive serotype 28 adenoviral vector comprises at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein. Any suitable amino acid residue(s) of a wild-type fiber protein of the serotype 28 adenoviral vector that mediates or assists in the interaction between the fiber knob and the native cellular receptor can be modified or removed, so long as the fiber protein is able to trimerize. Similarly, amino acids can be added to the fiber knob as long as the fiber protein retains the ability to trimerize. In a preferred embodiment, at least 75% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, or 100%) of the endogenous Ad28 fiber protein is modified or removed.

Nucleic acid sequences that encode all or a portion of a serotype 28 or 45 adenovirus fiber protein are publicly available (see, e.g., GenBank Accession Nos. AB361404.1, Y14242.1, FM210554.1, and AB361421.1). Amino acid sequences that comprise a full-length serotype 28 or 45 adenovirus fiber protein, or portions thereof, also are publicly available (see, e.g., GenBank Accession Nos. ACQ91171, CAR66130.1, BAG71098.1, and CAH18767.1). In one embodiment, the portion of an adenovirus serotype 45 fiber protein comprises the amino acid sequence of SEQ ID NO: 3, and a nucleic acid sequence that encodes a portion of a serotype 45 adenovirus fiber protein comprises, for example, SEQ ID NO: 4. In another embodiment, the portion of an adenovirus serotype 45 fiber protein comprises an amino acid sequence that is at least 67% identical (e.g., at least 68% identical, at least 69% identical, at least 70% identical, at least 71% identical, at least 72% identical, at least 73% identical, at least 74% identical, at least 75% identical, at least 76% identical, at least 77% identical, at least 78% identical, at least 79% identical, at least 80% identical, at least 81% identical, at least 82% identical, at least 83% identical, at least 84% identical, at least 85% identical, at least 86% identical, at least 87% identical, at least 88% identical, at least 89% identical, at least 90% identical, at least 91% identical, at least 92% identical, at least 93% identical, at least 94% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical) to SEQ ID NO: 3.

The inventive serotype 28 adenoviral vector comprises the aforementioned modified hexon protein, the aforementioned modified fiber protein, or the modified hexon protein and the modified fiber protein. For example, the inventive serotype 28 adenoviral vector comprises at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein, or at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein. Alternatively, the inventive serotype 28 adenoviral vector comprises at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein, and at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein.

Modifications to adenovirus coat proteins, including methods for generating chimeric hexon and fiber proteins, are described in, for example, e.g., U.S. Pat. Nos. 5,543,328; 5,559,099; 5,712,136; 5,731,190; 5,756,086; 5,770,442; 5,846,782; 5,871,727; 5,885,808; 5,922,315; 5,962,311; 5,965,541; 6,057,155; 6,127,525; 6,153,435; 6,329,190; 6,455,314; 6,465,253; 6,576,456; 6,649,407; and 6,740,525; U.S. Patent Application Publications 2001/0047081 A1, 2002/0099024 A1, 2002/0151027 A1, 2003/0022355 A1, and 2003/0099619 A1, and International Patent Application Publications WO 1996/007734, WO 1996/026281, WO 1997/020051, WO 1998/007865, WO 1998/007877, WO 1998/040509, WO 1998/054346, WO 2000/015823, WO 2001/058940, and WO 2001/092549.

The adenoviral vector can be replication-competent, conditionally replication-competent, or replication-deficient.

A replication-competent adenoviral vector can replicate in typical host cells, i.e., cells typically capable of being infected by an adenovirus. A replication-competent adenoviral vector can have one or more mutations as compared to the wild-type adenovirus (e.g., one or more deletions, insertions, and/or substitutions) in the adenoviral genome that do not inhibit viral replication in host cells. For example, the adenoviral vector can have a partial or entire deletion of the adenoviral early region known as the E3 region, which is not essential for propagation of the adenoviral genome.

A conditionally-replicating adenoviral vector is an adenoviral vector that has been engineered to replicate under pre-determined conditions. For example, replication-essential gene functions, e.g., gene functions encoded by the adenoviral early regions, can be operably linked to an inducible, repressible, or tissue-specific transcription control sequence, e.g., promoter. In such an embodiment, replication requires the presence or absence of specific factors that interact with the transcription control sequence. Conditionally-replicating adenoviral vectors are further described in U.S. Pat. No. 5,998,205.

A replication-deficient adenoviral vector is an adenoviral vector that requires complementation of one or more gene functions or regions of the adenoviral genome that are required for replication, as a result of, for example, a deficiency in the one or more replication-essential gene function or regions, such that the adenoviral vector does not replicate in typical host cells, especially those in a human to be infected by the adenoviral vector.

A deficiency in a gene function or genomic region, as used herein, is defined as a disruption (e.g., deletion) of sufficient genetic material of the adenoviral genome to obliterate or impair the function of the gene (e.g., such that the function of the gene product is reduced by at least about 2-fold, 5-fold, 10-fold, 20-fold, 30-fold, or 50-fold) whose nucleic acid sequence was disrupted (e.g., deleted) in whole or in part. Deletion of an entire gene region often is not required for disruption of a replication-essential gene function. However, for the purpose of providing sufficient space in the adenoviral genome for one or more transgenes, removal of a majority of one or more gene regions may be desirable. While deletion of genetic material is preferred, mutation of genetic material by addition or substitution also is appropriate for disrupting gene function. Replication-essential gene functions are those gene functions that are required for adenovirus replication (e.g., propagation) and are encoded by, for example, the adenoviral early regions (e.g., the E1, E2, and E4 regions), late regions (e.g., the L1, L2, L3, L4, and L5 regions), genes involved in viral packaging (e.g., the IVa2 gene), and virus-associated RNAs (e.g., VA-RNA-1 and/or VA-RNA-2).

Whether the adenoviral vector is replication-competent or replication-deficient, the adenoviral vector retains at least a portion of a serotype 28 adenoviral genome. The adenoviral vector can comprise any portion of a serotype 28 adenoviral genome, including protein coding and non-protein coding regions. Desirably, the adenoviral vector comprises at least one nucleic acid sequence that encodes a serotype 28 adenovirus protein. The adenoviral vector can comprise any suitable adenovirus protein, or a nucleic acid sequence that encodes any suitable adenovirus protein, such as, for example, a protein of any one of the early region genes (i.e., E1A, E1B, E2A, E2B, E3, and/or E4 regions), or a protein encoded by any one of the late region genes, which encode the virus structural proteins (i.e., L1, L2, L3, L4, and L5 regions).

The adenoviral vector desirably comprises one or more amino acid sequences of the pIX protein, the penton protein, the p100 protein, the L1 52/55K protein of a serotype 28 adenovirus, or any combination of the foregoing. The adenoviral vector can comprise a full-length amino acid sequence of a serotype 28 adenovirus protein. Alternatively, the adenoviral vector can comprise a portion of a full-length amino acid sequence of a serotype 28 adenovirus protein. An amino acid sequence of a serotype 28 adenovirus pIX protein comprises, for example, SEQ ID NO: 5. An amino acid sequence of a serotype 28 adenovirus penton protein comprises, for example, SEQ ID NO: 6. An amino acid sequence of a serotype 28 adenovirus p100 protein comprises, for example, SEQ ID NO: 7. An amino acid sequence of a serotype 28 adenovirus L1 52/55K protein comprises, for example, SEQ ID NO: 8. The adenoviral vector also desirably comprises a nucleic acid sequence encoding a DNA polymerase protein of a serotype 28 adenovirus or a portion thereof. A nucleic acid sequence encoding a DNA polymerase of a serotype 28 adenovirus comprises, for example, SEQ ID NO: 9. The adenoviral vector desirably comprises one or more of SEQ ID NOs: 5-9.

In another embodiment, the invention provides a virus-like particle comprising one or more amino acid sequences of the pIX protein, the penton protein, the p100 protein, the L1 52/55K protein of a serotype 28 adenovirus, or any combination of the foregoing, as well as the serotype 45 hexon protein and/or the serotype 45 fiber protein described herein. A “virus-like particle” consists of one or more viral coat proteins that assemble into viral particles, but lacks any viral genetic material (see, e.g., Miyanohara et al., J. Virol., 59: 176-180 (1986), Gheysen et al., Cell, 59: 103-112 (1989), and Buonaguro et al., ASHI Quarterly, 29: 78-80 (2005)).

Preferably, the adenoviral vector is replication-deficient, such that the replication-deficient adenoviral vector requires complementation of at least one replication-essential gene function of one or more regions of the adenoviral genome for propagation (e.g., to form adenoviral vector particles).

The replication-deficient adenoviral vector can be modified in any suitable manner to cause the deficiencies in the one or more replication-essential gene functions in one or more regions of the adenoviral genome for propagation. The complementation of the deficiencies in the one or more replication-essential gene functions of one or more regions of the adenoviral genome refers to the use of exogenous means to provide the deficient replication-essential gene functions. Such complementation can be effected in any suitable manner, for example, by using complementing cells and/or exogenous DNA (e.g., helper adenovirus) encoding the disrupted replication-essential gene functions.

The adenoviral vector can be deficient in one or more replication-essential gene functions of only the early regions (i.e., E1-E4 regions) of the adenoviral genome, only the late regions (i.e., L1-L5 regions) of the adenoviral genome, both the early and late regions of the adenoviral genome, or all adenoviral genes (i.e., a high capacity adenovector (HC-Ad)). See Morsy et al., Proc. Natl. Acad. Sci. USA, 95: 965-976 (1998); Chen et al., Proc. Natl. Acad. Sci. USA, 94: 1645-1650 (1997); and Kochanek et al., Hum. Gene Ther., 10: 2451-2459 (1999). Examples of replication-deficient adenoviral vectors are disclosed in U.S. Pat. Nos. 5,837,511; 5,851,806; 5,994,106; 6,127,175; 6,482,616; and 7,195,896, and International Patent Application Publications WO 1994/028152, WO 1995/002697, WO 1995/016772, WO 1995/034671, WO 1996/022378, WO 1997/012986, WO 1997/021826, and WO 2003/022311.

The early regions of the adenoviral genome include the E1, E2, E3, and E4 regions. The late regions of the adenoviral genome include the L1, L2, L3, L4, and L5 regions. The adenoviral vector also can have a mutation in the major late promoter (MLP), as discussed in International Patent Application Publication WO 2000/000628, which can render the adenoviral vector replication-deficient if desired.

The E1 region comprises the E1A and E1B subregions, and one or more deficiencies in replication-essential gene functions in the E1 region can include one or more deficiencies in replication-essential gene functions in either or both of the E1A and E1B subregions, thereby requiring complementation of the deficiency in the E1A subregion and/or the E1B subregion of the adenoviral genome for the adenoviral vector to propagate (e.g., to form adenoviral vector particles).

The E2 region comprises the E2A and E2B subregions, and one or more deficiencies in replication-essential gene functions in the E2 region can include one or more deficiencies in replication-essential gene functions in either or both of the E2A and E2B subregions, thereby requiring complementation of the deficiency in the E2A subregion and/or the E2B subregion of the adenoviral genome for the adenoviral vector to propagate (e.g., to form adenoviral vector particles).

The E3 region does not include any replication-essential gene functions, such that a deletion of the E3 region in part or in whole does not require complementation of any gene functions in the E3 region for the adenoviral vector to propagate (e.g., to form adenoviral vector particles). In the context of the invention, the E3 region is defined as the region that initiates with the open reading frame that encodes a protein with high homology to the 12.5K protein from the E3 region of human adenovirus 28 (NCBI reference sequence FJ824826) and ends with the open reading frame that encodes a protein with high homology to the 14.7K protein from the E3 region of human adenovirus 28 (NCBI reference sequence FJ824826). The E3 region may be deleted in whole or in part, or retained in whole or in part. The size of the deletion may be tailored so as to retain an adenoviral vector whose genome closely matches the optimum genome packaging size. A larger deletion will accommodate the insertion of larger heterologous nucleic acid sequences in the adenoviral genome. In one embodiment of the invention, the L4 polyadenylation signal sequences, which reside in the E3 region, are retained.

The E4 region comprises multiple open reading frames (ORFs). An adenoviral vector with a deletion of all of the open reading frames of the E4 region except ORF6, and in some cases ORF3, does not require complementation of any gene functions in the E4 region for the adenoviral vector to propagate (e.g., to form adenoviral vector particles). Conversely, an adenoviral vector with a disruption or deletion of ORF6, and in some cases ORF3, of the E4 region (e.g., with a deficiency in a replication-essential gene function based in ORF6 and/or ORF3 of the E4 region), with or without a disruption or deletion of any of the other open reading frames of the E4 region or the native E4 promoter, polyadenylation sequence, and/or the right-side inverted terminal repeat (ITR), requires complementation of the deficiency in the E4 region (specifically, of ORF6 and/or ORF3 of the E4 region) for the adenoviral vector to propagate (e.g., to form adenoviral vector particles).

The one or more regions of the adenoviral genome that contain one or more deficiencies in replication-essential gene functions desirably are one or more early regions of the adenoviral genome, i.e., the E1, E2, and/or E4 regions, optionally with the deletion in part or in whole of the E3 region. In other words, the adenoviral vector requires, at most, complementation of a deficiency in one or more early regions of the adenoviral genome for propagation.

The replication-deficient adenoviral vector also can have one or more mutations as compared to the wild-type adenovirus (e.g., one or more deletions, insertions, and/or substitutions) in the adenoviral genome that do not inhibit viral replication in host cells. Thus, in addition to one or more deficiencies in replication-essential gene functions, the adenoviral vector can be deficient in other respects that are not replication-essential. For example, the adenoviral vector can have a partial or entire deletion of the adenoviral early region known as the E3 region, which is not essential for propagation of the adenoviral genome.

In one embodiment, the adenoviral vector is replication-deficient and requires, at most, complementation of the E1 region of the adenoviral genome, for propagation (e.g., to form adenoviral vector particles). Thus, the replication-deficient adenoviral vector requires complementation of at least one replication-essential gene function of the E1A subregion and/or the E1B subregion of the adenoviral genome (denoted an E1-deficient adenoviral vector) for propagation (e.g., to form adenoviral vector particles). The adenoviral vector can be deficient in at least one replication-essential gene function (desirably all replication-essential gene functions) of the E1 region of the adenoviral genome and at least one gene function of the nonessential E3 region of the adenoviral genome (denoted an E1/E3-deficient adenoviral vector). Such an adenoviral vector requires, at most, complementation of a deficiency in the E1 region of the adenoviral genome for propagation.

In one embodiment, the adenoviral vector is replication-deficient and requires, at most, complementation of the E2 region, preferably the E2A subregion, of the adenoviral genome, for propagation (e.g., to form adenoviral vector particles). Thus, the replication-deficient adenoviral vector requires complementation of at least one replication-essential gene function of the E2A subregion of the adenoviral genome (denoted an E2A-deficient adenoviral vector) for propagation (e.g., to form adenoviral vector particles). The adenoviral vector can be deficient in at least one replication-essential gene function (desirably all replication-essential gene functions) of the E2A region of the adenoviral genome and at least one gene function of the nonessential E3 region of the adenoviral genome (denoted an E2A/E3-deficient adenoviral vector). Such an adenoviral vector requires, at most, complementation of a deficiency in the E2 region of the adenoviral genome for propagation.

In one embodiment, the adenoviral vector is replication-deficient and requires, at most, complementation of the E4 region of the adenoviral genome, for propagation (e.g., to form adenoviral vector particles). Thus, the replication-deficient adenoviral vector requires complementation of at least one replication-essential gene function of the E4 region of the adenoviral genome (denoted an E4-deficient adenoviral vector) for propagation (e.g., to form adenoviral vector particles). The adenoviral vector can be deficient in at least one replication-essential gene function (desirably all replication-essential gene functions) of the E4 region of the adenoviral genome and at least one gene function of the nonessential E3 region of the adenoviral genome (denoted an E3/E4-deficient adenoviral vector). Such an adenoviral vector requires, at most, complementation of a deficiency in the E4 region of the adenoviral genome for propagation.

In one embodiment, the adenoviral vector requires complementation of the E1 and E2 (e.g., E2A) regions of the adenoviral genome for propagation (denoted an E1/E2-deficient adenoviral vector), wherein the adenoviral vector also can be deficient in at least one gene function of the E3 region (denoted an E1/E2/E3-deficient adenoviral vector). Such an adenoviral vector requires, at most, complementation of a deficiency in the E1 region and a deficiency in the E2 region of the adenoviral genome for propagation.

In one embodiment, the adenoviral vector is replication-deficient and requires, at most, complementation of the E1 and E4 regions of the adenoviral genome for propagation (e.g., to form adenoviral vector particles). Thus, the replication-deficient adenoviral vector requires complementation of at least one replication-essential gene function of both the E1 and E4 regions of the adenoviral genome (denoted an E1/E4-deficient adenoviral vector) for propagation (e.g., to form adenoviral vector particles). The adenoviral vector can be deficient in at least one replication-essential gene function (desirably all replication-essential gene functions) of the E1 region of the adenoviral genome, at least one replication-essential gene function of the E4 region of the adenoviral genome, and at least one gene function of the nonessential E3 region of the adenoviral genome (denoted an E1/E3/E4-deficient adenoviral vector). Such an adenoviral vector requires, at most, complementation of a deficiency in the E1 region and a deficiency in the E4 region of the adenoviral genome for propagation.

In a preferred embodiment, the adenoviral vector requires, at most, complementation of a deficiency in the E1 region of the adenoviral genome for propagation, and does not require complementation of any other deficiency of the adenoviral genome for propagation. In another preferred embodiment, the adenoviral vector requires, at most, complementation of a deficiency in both the E1 and E4 regions of the adenoviral genome for propagation, and does not require complementation of any other deficiency of the adenoviral genome for propagation.

The adenoviral vector, when deficient in multiple replication-essential gene functions of the adenoviral genome (e.g., an E1/E4-deficient adenoviral vector), can include a spacer sequence to provide viral growth in a complementing cell line similar to that achieved by adenoviruses or adenoviral vectors deficient in a single replication-essential gene function (e.g., an E1-deficient adenoviral vector). The spacer sequence can contain any nucleotide sequence or sequences which are of a desired length, such as sequences at least about 15 base pairs (e.g., between about 15 nucleotides and about 12,000 nucleotides), preferably about 100 nucleotides to about 10,000 nucleotides, more preferably about 500 nucleotides to about 8,000 nucleotides, even more preferably about 1,500 nucleotides to about 6,000 nucleotides, and most preferably about 2,000 to about 3,000 nucleotides in length, or a range defined by any two of the foregoing values. The spacer sequence can be coding or non-coding and native or non-native with respect to the adenoviral genome, but does not restore the replication-essential function to the deficient region. The spacer also can contain an expression cassette. More preferably, the spacer comprises a polyadenylation sequence and/or a gene that is non-native with respect to the adenovirus or adenoviral vector. The use of a spacer in an adenoviral vector is further described in, for example, U.S. Pat. No. 5,851,806 and International Patent Application Publication WO 1997/021826.

The replication-deficient adenoviral vector of the invention can be produced in complementing cell lines that provide gene functions not present in the replication-deficient adenovirus or adenoviral vector, but required for viral propagation, at appropriate levels in order to generate high titers of viral vector stock. Such complementing cell lines are known and include, but are not limited to, 293 cells (described in, e.g., Graham et al., J. Gen. Virol., 36: 59-72 (1977)), PER.C6 cells (described in, e.g., International Patent Application Publication WO 1997/000326, and U.S. Pat. Nos. 5,994,128 and 6,033,908), and 293-ORF6 cells (described in, e.g., International Patent Application Publication WO 1995/34671 and Brough et al., J. Virol., 71: 9206-9213 (1997)). Other suitable complementing cell lines to produce the replication-deficient adenoviral vector of the invention include complementing cells that have been generated to propagate adenoviral vectors encoding transgenes whose expression inhibits viral growth in host cells (see, e.g., U.S. Patent Application Publication 2008/0233650). Additional suitable complementing cells are described in, for example, U.S. Pat. Nos. 6,677,156 and 6,682,929, and International Patent Application Publication WO 2003/020879. In some instances, the cellular genome need not comprise nucleic acid sequences, the gene products of which complement for all of the deficiencies of a replication-deficient adenoviral vector. One or more replication-essential gene functions lacking in a replication-deficient adenoviral vector can be supplied by a helper virus, e.g., an adenoviral vector that supplies in trans one or more essential gene functions required for replication of the replication-deficient adenoviral vector. Alternatively, the inventive adenoviral vector can comprise a non-native replication-essential gene that complements for the one or more replication-essential gene functions lacking in the inventive replication-deficient adenoviral vector. For example, an E1/E4-deficient adenoviral vector can be engineered to contain a nucleic acid sequence encoding E4 ORF 6 that is obtained or derived from a different adenovirus (e.g., an adenovirus of a different serotype than the inventive adenoviral vector, or an adenovirus of a different species than the inventive adenoviral vector).

An example of an E1/E3-deficient serotype 28 adenoviral vector comprising a serotype 45 hexon protein and a serotype 45 fiber protein as described herein comprises the nucleic acid sequence of SEQ ID NO: 10. Using the publicly available genome information for Ad28, however, one of ordinary skill in the art would be able generate other Ad28 vectors with similar deficiencies and/or modifications using routine methods known in the art and/or described herein.

The adenoviral vector can further comprise one or more exogenous or non-native nucleic acids, which can be positioned at any suitable place in the adenoviral vector. By removing all or part of the adenoviral genome, for example, the E1, E3, and E4 regions of the adenoviral genome, the resulting adenoviral vector is able to accept inserts of exogenous nucleic acid sequences while retaining the ability to be packaged into adenoviral capsids. An exogenous nucleic acid sequence can be inserted at any position in the adenoviral genome so long as insertion in the position allows for the formation of adenovirus or the adenoviral vector particle. The exogenous nucleic acid sequence preferably is positioned in the E1 region, the E3 region, or the E4 region of the adenoviral genome. In embodiments where the adenoviral vector comprises multiple exogenous nucleic acid sequences (e.g., 2, 3, 4 or more exogenous nucleic acid sequences), at least one exogenous nucleic acid sequence is positioned in the E1 region, and at least one exogenous nucleic acid sequence is positioned in the E4 region. For example, when the adenoviral vector comprises three exogenous nucleic acid sequences, two exogenous nucleic acid sequences can be positioned in the E1 region, and one exogenous nucleic acid sequence can be positioned in the E4 region.

An “exogenous” or “non-native” nucleic acid sequence is any nucleic acid sequence (e.g., DNA, RNA, or cDNA sequence) that is not a naturally occurring nucleic acid sequence of an adenovirus in a naturally occurring position. Thus, the non-native nucleic acid sequence can be naturally found in an adenovirus, but located at a non-native position within the adenoviral genome and/or operably linked to a non-native promoter. The terms “non-native nucleic acid sequence,” “heterologous nucleic acid sequence,” and “exogenous nucleic acid sequence” are synonymous and can be used interchangeably in the context of the invention. The non-native nucleic acid sequence preferably is DNA and preferably encodes a protein (i.e., one or more nucleic acid sequences encoding one or more proteins).

The non-native nucleic acid can be in the form of a transgene. The term “transgene” is defined herein as a non-native nucleic acid sequence that is operably linked to appropriate regulatory elements (e.g., a promoter), such that the non-native nucleic acid sequence can be expressed to produce a protein (e.g., peptide or polypeptide). The regulatory elements (e.g., promoter) can be native or non-native to the adenovirus or adenoviral vector.

The non-native nucleic acid sequence can encode a therapeutic protein that can be used to prophylactically or therapeutically treat a mammal for a disease. Examples of suitable therapeutic proteins include cytokines, toxins, tumor suppressor proteins, growth factors, hormones, receptors, mitogens, immunoglobulins, neuropeptides, neurotransmitters, and enzymes. Alternatively, the non-native nucleic acid sequence can encode an antigen of a pathogen (e.g., a bacterium or a virus), and the adenoviral vector can be used as a vaccine. For example, the non-native nucleic acid sequence can encode an antigen of a Herpes Simplex Virus-2, including, but not limited to an HSV-2 UL47 protein (e.g., SEQ ID NO: 11 or SEQ ID NO: 13), and/or an HSV-2 UL19 protein (e.g., SEQ ID NO: 12).

The invention provides a composition comprising the adenoviral vector described herein and a carrier therefor (e.g., a pharmaceutically acceptable carrier). The composition desirably is a physiologically acceptable (e.g., pharmaceutically acceptable) composition, which comprises a carrier, preferably a physiologically (e.g., pharmaceutically) acceptable carrier, and the adenoviral vector. Any suitable carrier can be used within the context of the invention, and such carriers are well known in the art. The choice of carrier will be determined, in part, by the particular use of the composition (e.g., administration to an animal) and the particular method used to administer the composition. Ideally, in the context of replication-deficient adenoviral vectors, the pharmaceutical composition preferably is free of replication-competent adenovirus. The pharmaceutical composition optionally can be sterile.

Suitable compositions include aqueous and non-aqueous isotonic sterile solutions, which can contain anti-oxidants, buffers, and bacteriostats, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The composition can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, immediately prior to use. Extemporaneous solutions and suspensions can be prepared from sterile powders, granules, and tablets. Preferably, the carrier is a buffered saline solution. More preferably, the adenovirus or adenoviral vector is part of a composition formulated to protect the adenovirus or adenoviral vector from damage prior to administration. For example, the composition can be formulated to reduce loss of the adenovirus or adenoviral vector on devices used to prepare, store, or administer the adenovirus or adenoviral vector, such as glassware, syringes, or needles. The composition can be formulated to decrease the light sensitivity and/or temperature sensitivity of the adenovirus or adenoviral vector. To this end, the composition preferably comprises a pharmaceutically acceptable liquid carrier, such as, for example, those described above, and a stabilizing agent selected from the group consisting of polysorbate 80, L-arginine, polyvinylpyrrolidone, trehalose, and combinations thereof. Use of such a composition will extend the shelf life of the adenovirus or adenoviral vector, and facilitate its administration. Formulations for adenovirus or adenoviral vector-containing compositions are further described in, for example, U.S. Pat. Nos. 6,225,289, 6,514,943, and International Patent Application Publication WO 2000/034444.

The composition also can be formulated to enhance transduction efficiency. In addition, one of ordinary skill in the art will appreciate that the adenoviral vector can be present in a composition with other therapeutic or biologically-active agents. For example, factors that control inflammation, such as ibuprofen or steroids, can be part of the composition to reduce swelling and inflammation associated with in vivo administration of the adenoviral vector. If the adenoviral vector is used to deliver an antigen-encoding nucleic acid sequence to a host, immune system stimulators or adjuvants, e.g., interleukins, lipopolysaccharide, double-stranded RNA, and/or TNFSF14/LIGHT (see, e.g., Zhang et al., J. Virol. Methods, 153(2): 142-148 (2008)) can be administered to enhance or modify any immune response to the antigen. Antibiotics, i.e., microbicides and fungicides, can be utilized to treat existing infection and/or reduce the risk of future infection, such as infection associated with gene transfer procedures.

The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.

Example 1

This example demonstrates the construction of a serotype 28 adenoviral vector comprising a serotype 45 adenovirus hexon protein and a serotype 45 adenovirus fiber protein, which encodes a Herpes Simplex Virus-2 (HSV) antigen.

A human serotype 28 adenoviral vector was prepared with a deletion in the E1 region, a portion of the hexon protein replaced with a portion of the hexon protein of a human serotype 45 adenovirus, and an insertion of a codon-optimized nucleic acid sequence encoding a wild-type HSV-2 UL19 protein into the E1 region (Ad28UL19 H(Ad45)). Ad28UL19 H(Ad45) is replication-deficient due to the deletion of the essential replication function provided by E1. In place of the E1 sequences, a CMVtetO UL19 expression cassette was introduced to provide the HSV-2 UL19 nucleic acid sequence. The expression cassette, located at the E1 region deletion junction, was oriented right-to-left with respect to the viral genome.

Ad28UL19 H(Ad45) has an E1 region deletion of nucleotides 462 through 3111. The deletion includes the E1A protein and part of the E1B protein, which renders the vector replication-incompetent in noncomplementing cell lines.

The construction of plasmid pAC28UL19.H(45) to provide Ad28UL19 H(Ad45) is schematically depicted in FIG. 1. Specifically, wild-type Ad28 virus DNA was rescued into a plasmid backbone by homologous recombination by using pAd28RSQ plasmid to make an Ad28 genome-containing plasmid (pACE28) (see FIG. 1, Step 3). A CMV tetO EGFP expression plasmid was introduced into the Ad28 genome-containing plasmid by homologous recombination to make an E1-deleted Ad28 genome-containing plasmid (pAC28EGFP) (462-3111 t ef) (see FIG. 1, Steps 4 and 5). PCR amplification and subcloning of the Ad45 hexon sequence into a shuttle plasmid was carried out by standard restriction enzyme subcloning. The Ad45 hexon nucleic acid sequence was inserted into pAC28EGFP plasmid by homologous recombination in E. coli to generate the Ad45 hexon-containing Ad28 adenoviral genome in plasmid form (pAC28EGFP.H(45)) (see FIG. 1, Steps 7 and 8). The HSV-2 UL19 gene was inserted into an adenoviral vector expression shuttle plasmid by standard restriction enzyme subcloning. The CMVTetO.UL19 gene was inserted into the pAC28EGFP.H(45) plasmid by homologous recombination in E. coli to generate the UL19-expressing Ad28 adenovector genome containing the Ad45 hexon sequence in plasmid form (pAC28UL19.H(45)) (see FIG. 1, Steps 9 and 10).

Plasmid pAC28UL19.H(45) was linearized with the restriction endonuclease PmeI. Adherent 293-ORF6 cells were transfected with the linearized plasmid, thereby resulting in conversion of the adenoviral genome (i.e., DNA) of the linearized plasmid into the viral vector Ad28UL19H(45).

The Ad28UL19H(45) adenoviral vector was expanded by serial passaging to generate high titer cell-virus lysate. The adenoviral vector was purified from the lysates by density-equilibrium centrifugation in cesium chloride gradients. The purified adenoviral vector was dialyzed into Final Formulation Buffer (FFB). The structural integrity of the genome of the adenoviral vector was confirmed by PCR, and the expression of the HSV-2 UL19 nucleic acid sequence was confirmed by Western blot.

A similar method was used to incorporate a portion of the Ad45 fiber nucleic acid sequence in place of a portion of the Ad28 fiber nucleic acid sequence.

The results of this example confirm the production of a serotype 28 adenoviral vector in accordance with the invention.

Example 2

This example demonstrates that the administration of the inventive serotype 28 adenoviral vector encoding one or more HSV antigens induces an immune response against HSV.

A serotype 28 adenoviral vector comprising portions of the hexon and fiber proteins from a serotype 45 adenovirus vector (Ad28 H/F) was produced using the methods described in Example 1. A first such adenoviral vector comprised a nucleic acid sequence encoding a wild-type HSV-2 UL47 protein (SEQ ID NO: 11). A second such adenoviral vector comprised a codon-optimized nucleic acid sequence encoding a wild-type HSV-2 UL19 protein (SEQ ID NO: 12).

T-cell response following a single intramuscular administration of each of the adenoviral vector-delivered antigens (1×109 PU) was assessed in a mouse model and compared to natural infection (1×106 PFU of HSV administered intravaginally) and control (i.e., formulation buffer (FFB)). A single administration of the adenoviral vector encoding either HSV-2 antigen induced a T-cell response that was greater than the T-cell response induced by a natural HSV infection.

The results of this example confirm that administration of the inventive serotype 28 adenoviral vector comprising a nucleic acid sequence encoding an HSV antigen induces an immune response against HSV.

Example 3

This example demonstrates that the administration of the inventive serotype 28 adenoviral vector encoding one or more HSV antigens induces an immune response against HSV.

Mice were divided into groups of four, and each group received a single intramuscular administration of one of the following: (a) an E1-deleted serotype 5 adenoviral vector encoding an HSV2 UL19 protein, (b) an E1-deleted serotype 28 adenoviral vector encoding an HSV2 UL19 protein, (c) the Ad28 H/F vector of the invention as described in Example 2, which encodes an HSV2 UL19 protein, and (d) formulation buffer (FFB; negative control). CD8+ T-cell responses following vaccination with the adenoviral vectors (1×109 PU) were assessed. The elicited T-cell responses are plotted in the graph of FIG. 2.

As is apparent from the results depicted in FIG. 2, a single administration of the Ad28H/F vector elicited a strong CD8+ T-cell response that was comparable to the CD8+ T-cell response elicited by the Ad5 vector.

The results of this example confirm that administration of the inventive serotype 28 adenoviral vector can induce a strong immune response against HSV.

All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.

The use of the terms “a” and “an” and “the” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.

Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

1. A replication-deficient serotype 28 adenoviral vector comprising one or both of the following:

(a) at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein, and
(b) at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein.

2. The adenoviral vector of claim 1, which comprises at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein, or at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein.

3. The adenoviral vector of claim 1, which comprises at least a portion of an adenovirus serotype 45 hexon protein in place of at least a portion of the endogenous serotype 28 hexon protein, and at least a portion of an adenovirus serotype 45 fiber protein in place of at least a portion of the endogenous serotype 28 fiber protein.

4. The adenoviral vector of claim 1, wherein the portion of an adenovirus serotype 45 hexon protein comprises at least one hypervariable region (HVR).

5. The adenoviral vector of claim 4, wherein the portion of an adenovirus serotype 45 hexon protein comprises nine HVRs.

6. The adenoviral vector of claim 1, wherein the portion of an adenovirus serotype 45 hexon protein comprises the amino acid sequence of SEQ ID NO: 1, or an amino acid sequence that is at least 91.4% identical to SEQ ID NO: 1.

7. The adenoviral vector of claim 6, wherein the portion of an adenovirus serotype 45 hexon protein is encoded by the nucleic acid sequence of SEQ ID NO: 2.

8. The adenoviral vector of claim 1, wherein the portion of an adenovirus serotype 45 fiber protein comprises the amino acid sequence of SEQ ID NO: 3, or an amino acid sequence that is at least 67% identical to SEQ ID NO: 3.

9. The adenoviral vector of claim 8, wherein the portion of an adenovirus serotype 45 fiber protein is encoded by the nucleic acid sequence of SEQ ID NO:

4.

10. The adenoviral vector of claim 1, which comprises:

(a) an amino acid sequence of a serotype 28 adenovirus penton protein,
(b) an amino acid sequence of a serotype 28 adenovirus pIX protein,
(c) an amino acid sequence of a serotype 28 adenovirus p100 protein,
(d) an amino acid sequence of a serotype 28 adenovirus L1 52/55K protein, or
(e) any combination of (a)-(d).

11. The adenoviral vector of claim 1, wherein the adenoviral vector requires complementation of a deficiency in one or more early regions of the adenoviral genome for propagation and does not require complementation of any other deficiency of the adenoviral genome for propagation.

12. The adenoviral vector of claim 1, wherein the one or more early regions are selected from the group consisting of the E1 region, the E2 region, and the E4 region of the adenovirus genome.

13. The adenoviral vector of claim 12, wherein the adenoviral vector requires complementation of a deficiency in the E1 region of the adenoviral genome for propagation and does not require complementation of any other deficiency of the adenoviral genome for propagation.

14. The adenoviral vector of claim 12, wherein the adenoviral vector requires complementation of a deficiency in the E1A region or the E1B region of the adenoviral genome for propagation and does not require complementation of any other deficiency of the adenoviral genome for propagation.

15. The adenoviral vector of claim 12, wherein the adenoviral vector requires at most complementation of a deficiency in the E4 region of the adenoviral genome for propagation and does not require complementation of any other deficiency of the adenoviral genome for propagation.

16. The adenoviral vector of claim 12, wherein the adenoviral vector requires complementation of a deficiency in the E1 region of the adenoviral genome and a deficiency in the E4 region of the adenoviral genome for propagation and does not require complementation of any other deficiency of the adenoviral genome for propagation.

17. The adenoviral vector of claim 1, which comprises the nucleic acid sequence of SEQ ID NO: 10.

18. The adenoviral vector of claim 1 further comprising a transgene.

19. A composition comprising the adenoviral vector of claim 1 and a pharmaceutically acceptable carrier.

Patent History
Publication number: 20210269827
Type: Application
Filed: Mar 12, 2021
Publication Date: Sep 2, 2021
Applicant: GenVec, Inc. (Gaithersburg, MD)
Inventors: Lisa Wei (Gaithersburg, MD), Douglas E. Brough (Gaithersburg, MD), C. Richter King (New York, NY)
Application Number: 17/200,012
Classifications
International Classification: C12N 15/86 (20060101); A61K 48/00 (20060101);