Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics

Information

  • Patent Grant
  • 11401529
  • Patent Number
    11,401,529
  • Date Filed
    Tuesday, June 30, 2020
    3 years ago
  • Date Issued
    Tuesday, August 2, 2022
    a year ago
Abstract
Recombinant adenovirus genomes that include an exogenous open reading frame (ORF) and a self-cleaving peptide coding sequence are described. Optimal placement of the exogenous genes for minimal impact on viral kinetics is further disclosed. Therapeutic applications of the recombinant adenoviruses are also described.
Description
FIELD

This disclosure concerns the optimal placement of exogenous open reading frames in recombinant adenovirus constructs and therapeutic applications of the recombinant viruses.


BACKGROUND

Adenovirus serotype 5 (Ad5) is the vector of choice in basic research applications, murine lung cancer models, and human gene therapy trials. Adenoviruses have a stable 36 kb double-stranded DNA genome protected by a protein capsid decorated with Ad fiber protein spikes that target infection to receptors on specific cell types. Adenoviruses do not integrate into host DNA, can be produced to high titers using established protocols, and have proven safety in human gene therapy and cancer applications. Thus, Ad-based vectors have enormous promise for cancer diagnostics and therapies.


SUMMARY

Disclosed herein are recombinant adenovirus genomes that include a heterologous open reading frame (ORF) and a self-cleaving peptide coding sequence. The heterologous ORF can encode, for example, a therapeutic protein. The recombinant adenovirus genomes and recombinant adenoviruses produced by the disclosed genomes can be used, for example, in therapeutic applications, such as for the treatment of cancer.


Provided herein are recombinant adenovirus genomes that include a heterologous ORF and a self-cleaving peptide coding sequence, both operably linked to and in the same reading frame as an endogenous adenovirus ORF. The self-cleaving peptide coding sequence is located between the heterologous ORF and the endogenous ORF. In some embodiments, the endogenous ORF is E1B-55k and the heterologous ORF is 3′ of E1B-55k; the endogenous ORF is DNA polymerase and the heterologous ORF is 5′ of DNA polymerase; the endogenous ORF is DNA-binding protein (DBP) and the heterologous ORF is 3′ of DBP; the endogenous ORF is adenovirus death protein (ADP) and the heterologous ORF is 5′ of ADP; the endogenous ORF is E3-14.7k and the heterologous ORF is 3′ of E3-14.7k; the endogenous ORF is E4-ORF2 and the heterologous ORF is 5′ of E4-ORF2; or the endogenous ORF is fiber and the heterologous ORF is 3′ of fiber. In some examples, the heterologous ORF encodes a therapeutic protein. Further provided herein are recombinant adenoviruses that include a recombinant adenovirus genome disclosed herein. Also provided are compositions that include a recombinant adenovirus genome or recombinant adenovirus disclosed herein and a pharmaceutically acceptable carrier.


Also provided is a method of delivering a therapeutic protein to a subject by administering to the subject a recombinant adenovirus genome or a recombinant adenovirus (or composition thereof) disclosed herein. In these methods, the heterologous ORF of the recombinant adenovirus genome or recombinant adenovirus encodes the therapeutic protein.


Further provided are methods of inhibiting tumor cell viability, inhibiting tumor cell growth, inhibiting tumor progression, reducing tumor volume and treating a subject with cancer by administering a recombinant adenovirus genome or a recombinant adenovirus (or composition thereof) disclosed herein. In these methods, the heterologous ORF of the recombinant adenovirus genome or recombinant adenovirus encodes a therapeutic protein suitable for the treatment of cancer.


The foregoing and other objects and features of the disclosure will become more apparent from the following detailed description, which proceeds with reference to the accompanying figures.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a schematic of an exemplary work-flow for testing adenoviral constructs. Whole virus genome plasmid is produced and transfected into suitable cells, such as 293-E4 cells, in a multi-well plate. As transfected cells expand, they are subjected to freeze/thaw to release viral particles, followed by centrifugation to pellet cell debris. The supernatant (containing the viral particles) is transferred to multiple, larger culture plates. Viral particles are harvested from transfected cells, CsCl purified and infectious virus titer is measured by ELISA. The cell type of interest is then infected with a known MOI of purified virus. At 48 or 72 hours post-infection, adenovirus late proteins, adenovirus genomes or plaques are measured by Western blot, q-PCR or plaque assay, respectively.



FIG. 2 is a schematic showing exponential viral growth. Oncolytic killing of all cells within a tumor requires exponential viral growth. However, in most instances, only a small percentage of tumor cells are initially infected. Thus, a small difference in the number of progeny per round of replication leads to large differences in the total number of particles after just a few rounds of replication. Shown is a comparison between a virus that produces 3 virions per cycle and a virus that produces 5 virions per cycle. As shown in the graph, after 5-6 rounds of replication, viral titers of the two viruses are significantly different.



FIG. 3 is a schematic showing an exemplary work-flow of a fluorescence-based viral kinetic (FBVK) assay. Whole virus genome plasmid is produced (such as by Adsembly or AdSLIC) and used to transfect a cell type of interest in a multi-well plate. Alternatively, cells are infected with recombinant adenovirus particles. The adenovirus genome comprises at least one open reading frame (ORF) encoding a fluorescent protein in a location within the viral genome that does not substantially alter viral replication kinetics. Fluorescence is monitored over time to calculate viral replication kinetics. Oncolytic virus candidates are those exhibiting the largest differential in virus kinetics between tumor cells and normal cells.



FIGS. 4A-4B outline the kinetic assay setup when starting with adenovirus genome plasmids. This assay does not require accurate knowledge of initial transfection efficiency. Transfection conditions are selected to result in approximately 5-10% of cells initially transfected. In the example shown, a 48-well plate is used, which allows for the testing of 14 different virus constructs in triplicate, along with three mock-infected wells and three wells with FLUORESBRITE™ beads to compensate for tool sensitivity drift. (FIG. 4A) The wells of the upper half of the 48-well plate contain cells transfected with the genome plasmids of 6 different viruses, mock-infected cells, and blanks (FLUORESBRITE™ beads), each in triplicate. (FIG. 4B) The wells of the lower half of the 48-well plate contain cells transfected with the genome plasmids of 8 different viruses in triplicate. The multi-well plate is placed on a plate reader (such as a TECAN plate reader) for continuous fluorescence monitoring.



FIG. 5 outlines the kinetic assay setup when starting with recombinant virus. This assay does not require accurate knowledge of virus titer. Recombinant virus is serially diluted and used to infect cells plated in a multi-well plate. In the example shown, a 96-well plate is used and each virus is diluted 1:100, 1:300, 1:900, 1:2700, 1:8100, 1:24,300, 1:72,900 and 1:218,700, allowing for the testing of 11 viruses simultaneously. Four wells are mock-infected and FLUORESBRITE™ beads are placed in four wells to compensate for tool sensitivity and drift. The multi-well plate is placed on a plate reader (such as a TECAN plate reader) for continuous fluorescence monitoring.



FIGS. 6A-6C provide a schematic overview of the Adsembly and AdSLIC techniques for the combinatorial assembly of recombinant adenoviruses. (FIG. 6A) The adenovirus genome is separated into four modules—E1, core, E3 and E4. (FIG. 6B) Adsembly involves genome reassembly using multi-site Gateway reactions. (FIG. 6C) AdSLIC utilizes sequence and ligation independent cloning (SLIC) to assemble adenovirus modules.



FIG. 7 is a bar graph showing ln-slope values for recombinant adenoviruses encoding a fluorescent protein in the E1 region. Shown are the values for the direct fusion construct YPet-E1A, and the YPet-P2A-E1A, E1A-P2A-mCherry and E1B-55k-P2A-YPet constructs, which each contain a P2A site. The YPet-P2A-ADP construct is shown for comparison.



FIG. 8 is a schematic of kinetic data analysis and interpretation for the fluorescence-based viral kinetic assay.



FIGS. 9A-9C are bar graphs showing ln-slope values for recombinant adenoviruses derived from Ad5, Ad9 or Ad34 and containing a heterologous ORF 3′ of the E3-14.7k ORF (or equivalent thereof in Ad9 and Ad34). Shown are the values for Ad5 (E3-14.7k-P2A-YPet; PCMN-887), Ad9 (E3-15k-P2A-YPet; PCMN-888) and Ad34 (E3-14.8k-P2A-YPet; PCMN-889) in 293 cells (FIG. 9A), A549 cells (FIG. 9B) and U2OS cells (FIG. 9C). Also shown in each figure are values for chimeric viruses comprising an Ad5 core (including E3-14.7k-P2A-YPet) and fiber shaft/knob from either Ad9 (Ad5/Ad9) or Ad34 (Ad5/Ad34).





SEQUENCE LISTING

The nucleic and amino acid sequences listed in the accompanying sequence listing are shown using standard letter abbreviations for nucleotide bases, and three letter code for amino acids, as defined in 37 C.F.R. 1.822. Only one strand of each nucleic acid sequence is shown, but the complementary strand is understood as included by any reference to the displayed strand. The Sequence Listing is submitted as an ASCII text file, created on Jun. 25, 2020, 703 KB, which is incorporated by reference herein. In the accompanying sequence listing:


SEQ ID NO: 1 is the nucleotide sequence of synthetic adenovirus genome CMBT-379 (YPet-P2A-E1A).


SEQ ID NO: 2 is the nucleotide sequence of synthetic adenovirus genome CMBT-432 (E1A-P2A-YPet).


SEQ ID NO: 3 is the nucleotide sequence of synthetic adenovirus genome CMBT-456 (E1B-55k-P2A-YPet).


SEQ ID NO: 4 is the nucleotide sequence of synthetic adenovirus genome CMBT-499 (E1B-55k-P2A-mCherry).


SEQ ID NO: 5 is the nucleotide sequence of synthetic adenovirus genome CMBT-530 (YPet-P2A-(DNA Poly)).


SEQ ID NO: 6 is the nucleotide sequence of synthetic adenovirus genome CMBT-886 (DBP-P2A-YPet).


SEQ ID NO: 7 is the nucleotide sequence of synthetic adenovirus genome CMBT-403 (YPet-P2A-ADP).


SEQ ID NO: 8 is the nucleotide sequence of synthetic adenovirus genome CMBT-429 (ADP-P2A-YPet).


SEQ ID NO: 9 is the nucleotide sequence of synthetic adenovirus genome PCMN-887 (E3-14.7k-P2A-YPet).


SEQ ID NO: 10 is the nucleotide sequence of synthetic adenovirus genome CMBT-457 (YPet-P2A-E4-ORF2).


SEQ ID NO: 11 is the nucleotide sequence of synthetic adenovirus genome CMBT-633 (mCherry-P2A-E4-ORF2).


SEQ ID NO: 12 is the nucleotide sequence of synthetic adenovirus genome CMBT-407 (YPet-P2A-Fiber).


SEQ ID NO: 13 is the nucleotide sequence of synthetic adenovirus genome CMBT-445 (Fiber-P2A-YPet).


SEQ ID NO: 14 is the amino acid sequence of P2A.


SEQ ID NO: 15 is the amino acid sequence of F2A.


SEQ ID NO: 16 is the amino acid sequence of E2A.


SEQ ID NO: 17 is the amino acid sequence of T2A.


SEQ ID NO: 18 is the amino acid sequence of a modified P2A comprising GSG at the N-terminus.


SEQ ID NO: 19 is the amino acid sequence of a modified F2A comprising GSG at the N-terminus.


SEQ ID NO: 20 is the amino acid sequence of a modified E2A comprising GSG at the N-terminus.


SEQ ID NO: 21 is the amino acid sequence of a modified T2A comprising GSG at the N-terminus.


SEQ ID NO: 22 is the nucleotide sequence of synthetic adenovirus genome PCMN-888 (Ad9 E3-15k-P2A-YPet).


SEQ ID NO: 23 is the nucleotide sequence of synthetic adenovirus genome PCMN-889 (Ad34 E3-14.8k-P2A-YPet).


DETAILED DESCRIPTION
I. Abbreviations

Ad adenovirus


ADP adenovirus death protein


BFP blue fluorescent protein


E2A equine rhinitis A virus 2A


ELISA enzyme-linked immunosorbent assay


ERAV equine rhinitis A virus


F2A foot and mouth disease virus 2A


FACS fluorescence activated cells sorting


FMDV food and mouth disease virus


GFP green fluorescent protein


MOI multiplicity of infection


OD optical density


ORF open reading frame


P2A porcine teschovirus-1 2A


pIX protein IX


PTV1 porcine teschovirus-1


RFP red fluorescent protein


SLIC sequence and ligation independent cloning


T2A Thosea asigna virus 2A


TaV Thosea asigna virus


YFP yellow fluorescent protein


II. Terms and Methods

Unless otherwise noted, technical terms are used according to conventional usage. Definitions of common terms in molecular biology may be found in Benjamin Lewin, Genes V, published by Oxford University Press, 1994 (ISBN 0-19-854287-9); Kendrew et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8).


In order to facilitate review of the various embodiments of the disclosure, the following explanations of specific terms are provided:


2A peptide: A type of self-cleaving peptide encoded by some RNA viruses, such as picornaviruses. 2A peptides function by making the ribosome skip the synthesis of a peptide bond at the C-terminus of a 2A element, leading to separation between the end of the 2A sequence and the downstream peptide (Kim et al., PLoS One 6(4):e18556, 2011). The “cleavage” occurs between the glycine and proline residues found on the C-terminus of the 2A peptide. Exemplary 2A peptides include, but are not limited to, the 2A peptides encoded by Thosea asigna virus (TaV), equine rhinitis A virus (ERAV), porcine teschovirus-1 (PTV1) and foot and mouth disease virus (FMDV), which are set forth herein as SEQ ID NOs: 14-17). In some embodiments, the 2A peptide comprises Gly-Ser-Gly at the N-terminus to improve cleavage efficiency (SEQ ID NOs: 18-21).


Adenovirus: A non-enveloped virus with a linear, double-stranded DNA genome and an icosahedral capsid. There are currently 68 known serotypes of human adenovirus, which are divided into seven species (species A, B, C, D, E, F and G). Different serotypes of adenovirus are associated with different types of disease, with some serotypes causing respiratory disease (primarily species B and C), conjunctivitis (species B and D) and/or gastroenteritis (species F and G).


Adenovirus death protein (ADP): A protein synthesized in the late stages of adenovirus infection that mediates lysis of cells and release of adenovirus to infect other cells. ADP is an integral membrane glycoprotein of 101 amino acids that localizes to the nuclear membrane, endoplasmic reticulum and Golgi. ADP was previously named E3-11.6K).


Administration: To provide or give a subject an agent, such as a therapeutic agent (e.g. a recombinant virus), by any effective route. Exemplary routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal, intratumoral, and intravenous), oral, intraductal, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.


Chimeric: Composed of at least two parts having different origins. In the context of the present disclosure, a “chimeric adenovirus” is an adenovirus having genetic material and/or proteins derived from at least two different serotypes (such as from Ad5 and a second serotype of adenovirus). In this context, a “capsid-swapped” adenovirus refers to a chimeric adenovirus in which the capsid proteins are derived from one serotype of adenovirus and the remaining proteins are derived from another adenovirus serotype. Similarly, a “chimeric fiber” is a fiber protein having amino acid sequence derived from at least two different serotypes of adenovirus. For example, a chimeric fiber can be composed of a fiber shaft from Ad5 and a fiber knob from a second serotype of adenovirus. In another example, a chimeric fiber is composed of an Ad5 tail and a fiber shaft and knob from a second serotype of adenovirus (such as Ad9 or Ad34).


Contacting: Placement in direct physical association; includes both in solid and liquid form.


Degenerate variant: In the context of the present disclosure, a “degenerate variant” refers to a polynucleotide encoding a peptide that includes a sequence that is degenerate as a result of the genetic code. There are 20 natural amino acids, most of which are specified by more than one codon. Therefore, all degenerate nucleotide sequences encoding a peptide are included as long as the amino acid sequence of the peptide encoded by the nucleotide sequence is unchanged.


Deleted: An adenovirus genome encoding a “deleted” protein (such as the E4orf1 or E4orf6/7 protein) refers to an adenovirus having a complete deletion of the protein coding sequence, or a partial deletion that results in the absence of protein expression.


Deregulation of E2F: Refers to an increase in activity of the E2F transcription factor and downstream target genes, which occurs in nearly all types of human cancer. Deregulation of the E2F pathway activity and transcription can result from a variety of different mutations in any upstream component of the pathway, such as loss of function mutations and deletions in Rb, p107 and p130 tumor suppressors. Rb was the first tumor suppressor to be identified and is absent or mutated in at least one third of human tumors. In addition, p16 mutations and/or epigenetic silencing can activate E2F in tumor cells. Cyclin D and CDK4 mutations, gene amplifications or over-expression can also result in deregulated E2F activity in human tumors. In addition E2F is activated by growth factor receptor pathway mutations including EGFR, RTKs, RAS, RAF, PI-3K, PTEN, RAF, MYC. Mutations in the p16INK4a-Cyclin D:cdk4/6-RB-E2F pathway generally occur in a mutually exclusive fashion, so that one ‘hit’ (for example, p16) is unaccompanied by others (for example, Rb mutation or cyclin D:cdk over-expression). However, most current chemotherapies are proliferative poisons that inhibit E2F transcriptional targets, but are also toxic to normal cells and have often devastating iatrogenic complications. As disclosed herein, an alternative therapeutic approach is to use a virus that undergoes selective lytic replication in cancer cell lesions that have deregulated the p16-cyclin D:cdk4-RB-E2F pathway.


DNA-binding protein (DBP): This adenovirus protein binds to single-stranded DNA and RNA, as well as double-stranded DNA. DBP, a 72-kilodalton protein, is essential for replication of adenoviral DNA.


E1A: The adenovirus early region 1A (E1A) gene and polypeptides expressed from the gene. The E1A protein plays a role in viral genome replication by driving cells into the cell cycle. As used herein, the term “E1A protein” refers to the proteins expressed from the E1A gene and the term includes E1A proteins produced by any adenovirus serotype.


E3-RIDα/RIDβ and E3-14.7k: Early-expressed proteins produced from the E3 gene. The E3-RIDα, E3-RIDβ, and E3-14.7k proteins make up the receptor internalization and degradation complex (RID), which localizes to the nuclear membrane and causes the endocytosis and degradation of a variety of receptors including CD95 (FasL receptor), and TNFR1 and 2 (TNF/TRAIL receptors) to protect infected cells from host antiviral responses. The E3-RIDα, E3-RIDβ, and E3-14.7k coding sequences are next to each other, in this order.


E4orf1: An adenovirus protein produced from the E4 gene. The term “E4orf1 protein” includes E4orf1 proteins produced by the E4 gene from any adenovirus serotype.


E4orf6/7: A protein encoded by the adenovirus E4 gene. The term “E4orf6/7 protein” includes E4orf6/7 proteins produced by the E4 gene from any adenovirus serotype.


Fiber: The adenovirus fiber protein is a trimeric protein that mediates binding to cell surface receptors. The fiber protein is comprised of a long N-terminal shaft and globular C-terminal knob.


Fluorescent protein: A protein that emits light of a certain wavelength when exposed to a particular wavelength of light. Fluorescent proteins include, but are not limited to, green fluorescent proteins (such as GFP, EGFP, AcGFP1, Emerald, Superfolder GFP, Azami Green, mWasabi, TagGFP, TurboGFP and ZsGreen), blue fluorescent proteins (such as EBFP, EBFP2, Sapphire, T-Sapphire, Azurite and mTagBFP), cyan fluorescent proteins (such as ECFP, mECFP, Cerulean, CyPet, AmCyan1, Midori-Ishi Cyan, mTurquoise and mTFP1), yellow fluorescent proteins (EYFP, Topaz, Venus, mCitrine, YPet, TagYFP, PhiYFP, ZsYellow1 and mBanana), orange fluorescent proteins (Kusabira Orange, Kusabira Orange2, mOrange, mOrange2 and mTangerine), red fluorescent proteins (mRuby, mApple, mStrawberry, AsRed2, mRFP1, JRed, mCherry, HcRed1, mRaspberry, dKeima-Tandem, HcRed-Tandem, mPlum, AQ143, tdTomato and E2-Crimson), orange/red fluorescence proteins (dTomato, dTomato-Tandem, TagRFP, TagRFP-T, DsRed, DsRed2, DsRed-Express (T1) and DsRed-Monomer) and modified versions thereof.


Fusion protein: A protein containing amino acid sequence from at least two different (heterologous) proteins or peptides. Fusion proteins can be generated, for example, by expression of a nucleic acid sequence engineered from nucleic acid sequences encoding at least a portion of two different (heterologous) proteins. To create a fusion protein, the nucleic acid sequences must be in the same reading frame and contain no internal stop codons. Fusion proteins, particularly short fusion proteins, can also be generated by chemical synthesis.


Heterologous: A heterologous protein or polypeptide refers to a protein or polypeptide derived from a different source or species.


Hexon: A major adenovirus capsid protein.


Immunomodulator: An agent that alters (e.g. activates, enhances or suppresses) the immune system. Immunomodulators include, but are not limited to, cytokines (such as interleukin 2 (IL-2), IL-7, IL-12, GM-CSF, FLT3 ligand, or interferons), chemokines (such as CCL3, CCL26, CXCL7, CXCL9, and CXCL10), T cell activating ligands (such as anti-CD3 Abs or alloantigens), co-stimulatory molecules (such as B7.1/B7.2, OX40L, 4-1-BBL or CD40L), checkpoint blockade inhibitors (such as anti-PD-1 or anti-CTLA4 Abs), and small molecule immunomodulators.


Isolated: An “isolated” biological component (such as a nucleic acid molecule, protein, virus or cell) has been substantially separated or purified away from other biological components in the cell or tissue of the organism, or the organism itself, in which the component naturally occurs, such as other chromosomal and extra-chromosomal DNA and RNA, proteins and cells. Nucleic acid molecules and proteins that have been “isolated” include those purified by standard purification methods. The term also embraces nucleic acid molecules and proteins prepared by recombinant expression in a host cell as well as chemically synthesized nucleic acid molecules and proteins.


Modification: A change in the sequence of a nucleic acid or protein sequence. For example, amino acid sequence modifications include, for example, substitutions, insertions and deletions, or combinations thereof. Insertions include amino and/or carboxyl terminal fusions as well as intrasequence insertions of single or multiple amino acid residues. Deletions are characterized by the removal of one or more amino acid residues from the protein sequence. In some embodiments herein, the modification (such as a substitution, insertion or deletion) results in a change in function, such as a reduction or enhancement of a particular activity of a protein. As used herein, “Δ” or “delta” refer to a deletion. Substitutional modifications are those in which at least one residue has been removed and a different residue inserted in its place. Amino acid substitutions are typically of single residues, but can occur at a number of different locations at once. Substitutions, deletions, insertions or any combination thereof may be combined to arrive at a final mutant sequence. These modifications can be prepared by modification of nucleotides in the DNA encoding the protein, thereby producing DNA encoding the modification. Techniques for making insertion, deletion and substitution mutations at predetermined sites in DNA having a known sequence are well known in the art. A “modified” protein, nucleic acid or virus is one that has one or more modifications as outlined above.


Neoplasia, malignancy, cancer and tumor: A neoplasm is an abnormal growth of tissue or cells that results from excessive cell division. Neoplastic growth can produce a tumor. The amount of a tumor in an individual is the “tumor burden” which can be measured as the number, volume, or weight of the tumor. A tumor that does not metastasize is referred to as “benign.” A tumor that invades the surrounding tissue and/or can metastasize is referred to as “malignant.” Malignant tumors are also referred to as “cancer.”


Hematologic cancers are cancers of the blood or bone marrow. Examples of hematological (or hematogenous) cancers include leukemias, including acute leukemias (such as acute lymphocytic leukemia, acute myelocytic leukemia, acute myelogenous leukemia and myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia), chronic leukemias (such as chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high grade forms), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia and myelodysplasia. In some cases, lymphomas are considered solid tumors.


Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, and other sarcomas, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, pheochromocytomas sebaceous gland carcinoma, papillary carcinoma, human papilloma virus (HPV)-infected neoplasias, papillary adenocarcinomas, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, Wilms' tumor, cervical cancer, testicular tumor, seminoma, bladder carcinoma, melanoma, and CNS tumors (such as a glioma (such as brainstem glioma and mixed gliomas), glioblastoma (also known as glioblastoma multiforme) astrocytoma, CNS lymphoma, germinoma, medulloblastoma, Schwannoma craniopharyogioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, neuroblastoma, retinoblastoma and brain metastasis).


Oncolytic virus: A virus that selectively kills cells of a proliferative disorder, e.g., cancer/tumor cells. Killing of the cancer cells can be detected by any method established in the art, such as determining viable cell count, or detecting cytopathic effect, apoptosis, or synthesis of viral proteins in the cancer cells (e.g., by metabolic labeling, immunoblot, or RT-PCR of viral genes necessary for replication), or reduction in size of a tumor.


Operably linked: A first nucleic acid sequence is operably linked with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Generally, operably linked DNA sequences are contiguous and, where necessary to join two protein-coding regions, in the same reading frame.


Pharmaceutically acceptable carrier: The pharmaceutically acceptable carriers (vehicles) useful in this disclosure are conventional. Remington's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, Pa., 15th Edition (1975), describes compositions and formulations suitable for pharmaceutical delivery of one or more therapeutic compounds, molecules or agents (e.g. a recombinant virus disclosed herein).


In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. For solid compositions (for example, powder, pill, tablet, or capsule forms), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.


Polypeptide, peptide or protein: A polymer in which the monomers are amino acid residues which are joined together through amide bonds. When the amino acids are alpha-amino acids, either the L-optical isomer or the D-optical isomer can be used. The terms “polypeptide,” “peptide” and “protein” are used interchangeably herein. These terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. The term “residue” or “amino acid residue” includes reference to an amino acid that is incorporated into a protein, polypeptide, or peptide.


A conservative substitution in a polypeptide is a substitution of one amino acid residue in a protein sequence for a different amino acid residue having similar biochemical properties. Typically, conservative substitutions have little to no impact on the activity of a resulting polypeptide. For example, a protein or peptide including one or more conservative substitutions (for example no more than 1, 2, 3, 4 or 5 substitutions) retains the structure and function of the wild-type protein or peptide. A polypeptide can be produced to contain one or more conservative substitutions by manipulating the nucleotide sequence that encodes that polypeptide using, for example, standard procedures such as site-directed mutagenesis or PCR. In one example, such variants can be readily selected by testing antibody cross-reactivity or its ability to induce an immune response. Examples of conservative substitutions are shown below.
















Original Residue
Conservative Substitutions









Ala
Ser



Arg
Lys



Asn
Gln, His



Asp
Glu



Cys
Ser



Gln
Asn



Glu
Asp



His
Asn; Gln



Ile
Leu, Val



Leu
Ile; Val



Lys
Arg; Gln; Glu



Met
Leu; Ile



Phe
Met; Leu; Tyr



Ser
Thr



Thr
Ser



Trp
Tyr



Tyr
Trp; Phe



Val
Ile; Leu










Conservative substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.


The substitutions which in general are expected to produce the greatest changes in protein properties will be non-conservative, for instance changes in which (a) a hydrophilic residue, for example, seryl or threonyl, is substituted for (or by) a hydrophobic residue, for example, leucyl, isoleucyl, phenylalanyl, valyl or alanyl; (b) a cysteine or proline is substituted for (or by) any other residue; (c) a residue having an electropositive side chain, for example, lysyl, arginyl, or histidyl, is substituted for (or by) an electronegative residue, for example, glutamyl or aspartyl; or (d) a residue having a bulky side chain, for example, phenylalanine, is substituted for (or by) one not having a side chain, for example, glycine.


Preventing, treating or ameliorating a disease: “Preventing” a disease refers to inhibiting the full development of a disease. “Treating” refers to a therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition after it has begun to develop. “Ameliorating” refers to the reduction in the number or severity of signs or symptoms of a disease.


Promoter: A region of DNA that directs/initiates transcription of a nucleic acid (e.g. a gene). A promoter includes necessary nucleic acid sequences near the start site of transcription. Typically, promoters are located near the genes they transcribe. A promoter also optionally includes distal enhancer or repressor elements which can be located as much as several thousand base pairs from the start site of transcription. A “constitutive promoter” is a promoter that is continuously active and is not subject to regulation by external signals or molecules. In contrast, the activity of an “inducible promoter” is regulated by an external signal or molecule (for example, a transcription factor or tetracycline).


Protein IX (pIX): A minor component of the adenovirus capsid that associates with the hexon protein.


Purified: The term “purified” does not require absolute purity; rather, it is intended as a relative term. Thus, for example, a purified peptide, protein, virus, or other active compound is one that is isolated in whole or in part from naturally associated proteins and other contaminants. In certain embodiments, the term “substantially purified” refers to a peptide, protein, virus or other active compound that has been isolated from a cell, cell culture medium, or other crude preparation and subjected to fractionation to remove various components of the initial preparation, such as proteins, cellular debris, and other components.


Recombinant: A recombinant nucleic acid molecule, protein or virus is one that has a sequence that is not naturally occurring or has a sequence that is made by an artificial combination of two otherwise separated segments of sequence. This artificial combination can be accomplished by chemical synthesis or by the artificial manipulation of isolated segments of nucleic acid molecules, such as by genetic engineering techniques. The term “recombinant” also includes nucleic acids, proteins and viruses that have been altered solely by addition, substitution, or deletion of a portion of the natural nucleic acid molecule, protein or virus.


Replication defects: An adenovirus that exhibits “replication defects” in a non-tumor cell (compared to a tumor cell) refers to an adenovirus that exhibits reduced viral replication in normal cells compared to tumor cells. Replication defects are evidenced by, for example, a lack of viral late protein expression, a reduction in viral DNA synthesis, a reduced ability to induce E2F target genes (e.g. cyclin A and B), a reduced ability to elicit S phase entry and/or a reduced ability to induce cell killing in normal cells compared to tumor cells.


Replication deficient virus: A virus that preferentially inhibits cell proliferation, causes cell lysis, or induces apoptosis (collectively considered killing) in a predetermined cell population with a given phenotype (e.g., tumor cells with a deregulated E2F pathway). Such viruses are unable to or are limited in the ability to reduce or inhibit cell proliferation, cause cell lysis, induce apoptosis, or otherwise replicate in cells that do not have the predetermined cell phenotype (such as normal, non-tumor cells).


Self-cleaving peptides: Peptides that induce the ribosome to skip the synthesis of a peptide bond at the C-terminus, leading to separation of the peptide sequence and a downstream polypeptide. Virally encoded 2A peptides are a type of self-cleaving peptide. Virally encoded 2A peptides include, for example, 2A peptides from porcine teschovirus-1 (PTV1), foot and mouth disease virus (FMDV), equine rhinitis A virus (ERAV) and Thosea asigna virus (TaV).


Sequence identity: The identity or 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 can be measured in terms of percentage identity; the higher the percentage, the more identical the sequences are. Sequence similarity can be measured in terms of percentage similarity (which takes into account conservative amino acid substitutions); the higher the percentage, the more similar the sequences are. Homologs or orthologs of nucleic acid or amino acid sequences possess a relatively high degree of sequence identity/similarity when aligned using standard methods. This homology is more significant when the orthologous proteins or cDNAs are derived from species which are more closely related (such as human and mouse sequences), compared to species more distantly related (such as human and C. elegans sequences).


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, 73:237-44, 1988; Higgins & Sharp, CABIOS 5:151-3, 1989; Corpet et al., Nuc. Acids Res. 16:10881-90, 1988; Huang et al. Computer Appls. in the Biosciences 8, 155-65, 1992; and Pearson et al., Meth. Mol. Bio. 24:307-31, 1994. Altschul et al., J. Mol. Biol. 215:403-10, 1990, presents a detailed consideration of sequence alignment methods and homology calculations.


The NCBI Basic Local Alignment Search Tool (BLAST) (Altschul et al., J. Mol. Biol. 215:403-10, 1990) is available from several sources, including the National Center for Biological Information (NCBI) and on the internet, for use in connection with the sequence analysis programs blastp, blastn, blastx, tblastn and tblastx. Additional information can be found at the NCBI web site.


Serotype: A group of closely related microorganisms (such as viruses) distinguished by a characteristic set of antigens.


Subject: Living multi-cellular vertebrate organisms, a category that includes human and non-human mammals.


Synthetic: Produced by artificial means in a laboratory, for example a synthetic nucleic acid or protein can be chemically synthesized in a laboratory.


Therapeutic agent: Any agent capable of inducing a desired therapeutic or prophylactic effect when properly administered to a subject. Therapeutic agents include, but are not limited to, chemical compounds, small molecules, recombinant viruses, antisense compounds, antibodies (or antigen-binding fragments thereof), peptides or nucleic acid molecules. For example, therapeutic agents for treating cancer include agents that prevent or inhibit tumor growth, tumor development, or tumor metastasis. “Therapeutic proteins” are therapeutic agents that are proteins or peptides, including antibodies or antigen-binding fragments thereof. In some embodiments herein, the therapeutic protein is an immunomodulator. In other embodiments, the therapeutic protein comprises a toxin, Fas or FasL, a soluble death factor, a mediator of bystander destruction, a tumor antigen, a neo antigen or an alloantigen.


Therapeutically effective amount: A quantity of a specified pharmaceutical or therapeutic agent (e.g. a recombinant virus) sufficient to achieve a desired effect in a subject, or in a cell, being treated with the agent. The effective amount of the agent can be dependent on several factors, including, but not limited to the subject or cells being treated, and the manner of administration of the therapeutic composition.


Uexon: An open reading frame located on the/strand (leftward transcription) between the early E3 region and the fiber gene (Tollefson et al., J Virol 81(23):12918-12926).


Unless otherwise explained, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The singular terms “a,” “an,” and “the” include plural referents unless context clearly indicates otherwise. “Comprising A or B” means including A, or B, or A and B. It is further to be understood that all base sizes or amino acid sizes, and all molecular weight or molecular mass values, given for nucleic acids or polypeptides are approximate, and are provided for description. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including explanations of terms, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.


III. Overview of Embodiments

Disclosed herein are recombinant adenovirus genomes that include a heterologous open reading frame (ORF) and a self-cleaving peptide coding sequence. The heterologous ORF can encode, for example, a therapeutic protein (such as an immunomodulator). The recombinant adenovirus genomes and recombinant adenoviruses produced by the disclosed genomes can be used, for example, in a variety of different therapeutic applications, such as the treatment of cancer.


Provided herein are recombinant adenovirus genomes that include a heterologous ORF and a self-cleaving peptide coding sequence, both operably linked to and in the same reading frame as an endogenous adenovirus ORF. The self-cleaving peptide coding sequence is located between the heterologous ORF and the endogenous ORF. In some embodiments, the endogenous ORF is E1B-55k and the heterologous ORF is 3′ of E1B-55k; the endogenous ORF is DNA polymerase and the heterologous ORF is 5′ of DNA polymerase; the endogenous ORF is DNA-binding protein (DBP) and the heterologous ORF is 3′ of DBP; the endogenous ORF is adenovirus death protein (ADP) and the heterologous ORF is 5′ of ADP; the endogenous ORF is E3-14.7k and the heterologous ORF is 3′ of E3-14.7k; the endogenous ORF is E4-ORF2 and the heterologous ORF is 5′ of E4-ORF2; or the endogenous ORF is fiber and the heterologous ORF is 3′ of fiber.


In some embodiments, the heterologous ORF encodes a therapeutic protein.


In some embodiments, the self-cleaving peptide is a 2A peptide or variant thereof. In some examples, the 2A peptide includes a porcine teschovirus-1 (PTV1) 2A (P2A) peptide, a foot and mouth disease virus (FMDV) 2A (F2A) peptide, an equine rhinitis A virus (ERAV) 2A (E2A) peptide or a Thosea asigna virus (TaV) 2A (T2A) peptide, or a variant thereof. In particular examples, the P2A peptide sequence is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to the amino acid sequence of SEQ ID NO: 14 or SEQ ID NO: 18. In some examples, the 2A peptide variant comprises additional amino acid sequence (such as GSG) at the N-terminus.


In particular examples, the F2A peptide sequence is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 19. In particular examples, the E2A peptide sequence is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to the amino acid sequence of SEQ ID NO: 16 or SEQ ID NO: 20. In particular examples, the T2A peptide sequence is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to the amino acid sequence of SEQ ID NO: 17 or SEQ ID NO: 21. In specific non-limiting examples, the self-cleaving peptide comprises or consists of the amino acid sequence of any one of SEQ ID NOs: 14-21.


In some embodiments, the adenovirus is an adenovirus type 5 (Ad5). In other embodiments, the adenovirus is an Ad2, Ad3, Ad9, Ad11, Ad12 or Ad34. In yet other embodiments, the adenovirus is a chimeric adenovirus, such as, but not limited to, an Ad5/Ad9 or Ad5/Ad34 chimeric adenovirus.


Further provided herein are recombinant adenoviruses that include a recombinant adenovirus genome disclosed herein.


The recombinant adenoviruses (and recombinant adenovirus genomes) provided herein optionally include additional modifications, such as to target the virus to specific cell types, to inhibit targeting to and replication in the liver, to allow for selective replication in tumor cells, and to evade pre-existing neutralizing antibodies to common adenovirus serotypes. The additional modifications can vary depending on the desired use of the recombinant adenovirus. Adenovirus modifications are described, for example, in PCT Application No. PCT/US2015/051745 (filed Sep. 23, 2015), WO 2012/024350, WO 2013/138505 and WO 2014/153204, which are herein incorporated by reference in their entirety.


Compositions that include a recombinant adenovirus genome, or a recombinant adenovirus, and a pharmaceutically acceptable carrier are also provided by the present disclosure.


Also provided herein are methods of delivering a therapeutic protein to a subject. In some embodiments, the method includes administering to the subject a recombinant adenovirus genome, a recombinant adenovirus, or a composition disclosed herein. In these methods, the heterologous ORF of the recombinant adenovirus or recombinant adenovirus genome encodes the therapeutic protein.


Further provided are methods of reducing or inhibiting tumor cell viability and/or tumor cell growth (for example a reduction of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, at least 90%, at least 95%, or at least 98%, as compared to an absence of the disclosed therapy). In some embodiments, the method includes contacting the tumor cell with a recombinant adenovirus genome, a recombinant adenovirus, or a composition disclosed herein. In these methods, the heterologous ORF encodes a therapeutic protein. In some examples, the method is an in vitro method. In other examples, the method is an in vivo method and contacting the tumor cell includes administering the recombinant adenovirus genome, recombinant adenovirus, or composition to a subject with a tumor.


Methods of reducing or inhibiting tumor progression, such as by reducing the number and/or size of a metastasis, or reducing tumor volume in a subject are further provided herein (for example a reduction of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, at least 90%, at least 95%, or at least 98%, as compared to an absence of the disclosed therapy). In some embodiments, the method includes administering to the subject a therapeutically effective amount of a recombinant adenovirus genome, a recombinant adenovirus, or a composition disclosed herein. In these methods, the heterologous ORF encodes a therapeutic protein.


Further provided are methods of treating cancer in a subject. In some embodiments, the method includes administering to the subject a therapeutically effective amount of a recombinant adenovirus genome, a recombinant adenovirus, or a composition disclosed herein. In these methods, the heterologous ORF encodes a therapeutic protein.


In some embodiments herein, the method further includes administering an additional therapeutic agent to the subject. For example, the additional therapeutic agent may include an anti-cancer agent, such as a chemotherapeutic agent, a biologic (such as an antibody or fragment thereof, such as a monoclonal antibody), or a nucleic acid molecule, such as an inhibitory nucleic acid molecule) or other therapeutic treatment, such as surgical resection of a tumor or irradiation of a tumor.


Also provided herein are kits that include a recombinant adenovirus genome, a recombinant adenovirus, or a composition disclosed herein; and one or more additional therapeutic agents and/or one or more diagnostic agents. In some embodiments, the one or more additional therapeutic agents include a chemotherapeutic, biologic, or combinations thereof. In some examples, the one or more diagnostic agents include one or more antibodies or nucleic acid molecules specific for a tumor marker, or an imaging probe that can be used to track the virus or tumor cells in vitro or in vivo.


IV. Optimal Placement of Exogenous ORFs

The 36 kb adenovirus genome is compact, using both the top and bottom strands for coding of various genes. At many locations within the adenovirus genome, both the top and bottom strand are used simultaneously for coding separate genes. The genome size has evolved to be optimal for insertion into its capsid. As a result, the insertion of exogenous genes is limited by the size capacity of the capsid as excessive addition of exogenous nucleic acid leads to incomplete genome loading into the capsid and reduced viral kinetics.


A solution to the challenge presented by the limited available space in the adenovirus genome is to locate exogenous open reading frames (ORFs) as fusion products within native adenovirus ORFs. This strategy makes use of adenovirus promoters, 5′UTRs, and polyA tails already encoded in the genome. However, expression of a fusion between a native adenovirus protein and an exogenous protein can be deleterious to one or both protein functions and lead to a significant decrease in adenovirus replication kinetics.


The present disclosure provides a solution to this problem by using a self-cleaving peptide sequence placed between the native (endogenous) ORF and the exogenous (heterologous) ORF. When placed between the two ORFs on a single mRNA, the presence of the self-cleaving peptide sequence leads to ribosome skipping and release of the first protein separate from the second protein. In some embodiments disclosed herein, the self-cleaving peptide is a 2A peptide (P2A).


Also disclosed herein is the identification of optimal placement sites for heterologous ORFs within the adenovirus genome. The combination of the self-cleaving peptide sequence and the judicious placement of the heterologous ORF leads to high expression and minimal to no impact on viral kinetics.


As described in Example 1 below, several sites within the adenovirus genome were identified that upon insertion of a heterologous ORF, did not inhibit adenovirus replication kinetics. In particular, it was determined that a heterologous ORF could be inserted C-terminal to the E1B-55k ORF, N-terminal to the DNA polymerase ORF, C-terminal to the DBP ORF, N-terminal to the ADP ORF, C-terminal to the E3-14.7k ORF or N-terminal to E4-ORF2. In each instance, a self-cleaving peptide sequence (P2A site) was inserted between the adenovirus ORF and the heterologous ORF. It is further disclosed herein that insertion of a heterologous ORF C-terminal to fiber produced a replication defective adenovirus; however, the recombinant virus was capable of producing extraordinarily high levels of heterologous protein in infected cells, which could prove useful in a number of therapeutic applications.


Therefore, the present disclosure contemplates the use of the following recombinant adenoviruses for therapeutic applications (where “SC” refers to a sequence encoding a self-cleaving peptide, such as P2A):


E1B-55k-SC-heterologous ORF


heterologous ORF-SC-(DNA polymerase)


DBP-SC-heterologous ORF


heterologous ORF-SC-ADP


E3-14.7k-SC-heterologous ORF


heterologous ORF-SC-E4-ORF2


fiber-SC-heterologous ORF


In some embodiments herein, the self-cleaving peptide is a virally encoded 2A peptide, or a modified version thereof as described further below.


V. Self-Cleaving Peptide Sequences

Self-cleaving peptides are peptides that induce the ribosome to skip the synthesis of a peptide bond at the C-terminus, leading to separation of the peptide sequence and a downstream polypeptide. The use of self-cleaving peptides allows for expression of multiple proteins flanking the self-cleaving peptide from a single ORF. Virally encoded 2A peptides are one type of self-cleaving peptide.


As with other self-cleaving peptides, 2A peptides function by making the ribosome skip the synthesis of a peptide bond at the C-terminus of a 2A element, leading to separation between the end of the 2A sequence and the downstream peptide (Kim et al., PLoS One 6(4):e18556, 2011). The “cleavage” occurs between the glycine and proline residues found on the C-terminus of the 2A peptide. Exemplary 2A peptides include, but are not limited to, the 2A peptides encoded by Thosea asigna virus (TaV), equine rhinitis A virus (ERAV), porcine teschovirus-1 (PTV1) and foot and mouth disease virus (FMDV), or modified versions thereof


In particular examples herein, the 2A peptide comprises PTV1 2A (P2A), FMDV 2A (F2A), ERAV 2A (E2A) or TaV 2A (T2A), the sequences of which are show below and are set forth herein as SEQ ID NOs: 14-17.









P2A:


(SEQ ID NO: 14)


ATNFSLLKQAGDVEENPGP





F2A:


(SEQ ID NO: 15)


VKQTLNFDLLKLAGDVESNPGP





E2A:


(SEQ ID NO: 16)


QCTNYALLKLAGDVESNPGP





T2A:


(SEQ ID NO: 17)


EGRGSLLTCGDVEENPGP






In some examples, the 2A peptide is modified to include Gly-Ser-Gly at the N-terminus to improve cleavage efficiency. The sequences of modified P2A, F2A, E2A and T2A are shown below and are set forth herein as SEQ ID NOs: 14-17.









Modified P2A:


(SEQ ID NO: 18)


GSGATNFSLLKQAGDVEENPGP





Modified F2A:


(SEQ ID NO: 19)


GSGVKQTLNFDLLKLAGDVESNPGP





Modified E2A:


(SEQ ID NO: 20)


GSGQCTNYALLKLAGDVESNPGP





Modified T2A:


(SEQ ID NO: 21)


GSGEGRGSLLTCGDVEENPGP






In some embodiments, the 2A polypeptide is a variant of a 2A polypeptide disclosed herein. Variants can include polypeptide sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or more, sequence identity to a wild-type or modified 2A polypeptide disclosed herein. Variants can include, for example, a deletion of at least one N-terminal amino acid from the 2A polypeptide of any one of SEQ ID NOs: 14-21, for example a deletion of 1, 2, 3, 4 or 5 amino acids, including ranges between any two of the listed values. Variants can include a deletion of at least one C-terminal amino acid from the 2A polypeptide of any one of SEQ ID NOs: 14-21, for example a deletion of 1, 2, 3, 4 or 5 amino acids, including ranges between any two of the listed values. Variants can also include, for example, at least 1, 2, 3, 4 or 5 amino acid substitutions, such as conservative amino acid substitutions.


VI. Pharmaceutical Compositions

Provided herein are compositions comprising a recombinant adenovirus or a recombinant adenovirus genome. The compositions are, optionally, suitable for formulation and administration in vitro or in vivo. Optionally, the compositions comprise one or more of the provided agents and a pharmaceutically acceptable carrier. Suitable carriers and their formulations are described in Remington: The Science and Practice of Pharmacy, 22nd Edition, Loyd V. Allen et al., editors, Pharmaceutical Press (2012). Pharmaceutically acceptable carriers include materials that are not biologically or otherwise undesirable, i.e., the material is administered to a subject without causing undesirable biological effects or interacting in a deleterious manner with the other components of the pharmaceutical composition in which it is contained. If administered to a subject, the carrier is optionally selected to minimize degradation of the active ingredient and to minimize adverse side effects in the subject.


The recombinant viruses (or one or more nucleic acids or vectors encoding the recombinant adenovirus) are administered in accord with known methods, such as intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, oral, topical, intratumoral or inhalation routes. The administration may be local or systemic. The compositions can be administered via any of several routes of administration, including topically, orally, parenterally, intravenously, intra-articularly, intraperitoneally, intramuscularly, subcutaneously, intracavity, transdermally, intrahepatically, intracranially, nebulization/inhalation, or by installation via bronchoscopy. Thus, the compositions are administered in a number of ways depending on whether local or systemic treatment is desired, and on the area to be treated.


In some embodiments, the compositions for administration will include a recombinant adenovirus (or recombinant genome) as described herein dissolved in a pharmaceutically acceptable carrier, preferably an aqueous carrier. A variety of aqueous carriers can be used, e.g., buffered saline and the like. These solutions are sterile and generally free of undesirable matter. These compositions may be sterilized by conventional, well known sterilization techniques. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate and the like. The concentration of active agent in these formulations can vary widely, and will be selected primarily based on fluid volumes, viscosities, body weight and the like in accordance with the particular mode of administration selected and the subject's needs.


Pharmaceutical formulations, particularly, of the recombinant viruses can be prepared by mixing the recombinant adenovirus (or one or more nucleic acids encoding the recombinant adenovirus) having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers. Such formulations can be lyophilized formulations or aqueous solutions.


Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used. Acceptable carriers, excipients or stabilizers can be acetate, phosphate, citrate, and other organic acids; antioxidants (e.g., ascorbic acid) preservatives, low molecular weight polypeptides; proteins, such as serum albumin or gelatin, or hydrophilic polymers such as polyvinylpyllolidone; and amino acids, monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents; and ionic and non-ionic surfactants (e.g., polysorbate); salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants. The recombinant adenovirus (or one or more nucleic acids encoding the recombinant adenovirus) can be formulated at any appropriate concentration of infectious units.


Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the recombinant adenovirus suspended in diluents, such as water, saline or PEG 400; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as liquids, solids, granules or gelatin; (c) suspensions in an appropriate liquid; and (d) suitable emulsions. Tablet forms can include one or more of lactose, sucrose, mannitol, sorbitol, calcium phosphates, corn starch, potato starch, microcrystalline cellulose, gelatin, colloidal silicon dioxide, talc, magnesium stearate, stearic acid, and other excipients, colorants, fillers, binders, diluents, buffering agents, moistening agents, preservatives, flavoring agents, dyes, disintegrating agents, and pharmaceutically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, e.g., sucrose, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin or sucrose and acacia emulsions, gels, and the like containing, in addition to the active ingredient, carriers known in the art.


The recombinant adenovirus (or one or more nucleic acids encoding the recombinant adenovirus), alone or in combination with other suitable components, can be made into aerosol formulations (i.e., they can be “nebulized”) to be administered via inhalation. Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.


Formulations suitable for parenteral administration, such as, for example, by intraarticular (in the joints), intravenous, intramuscular, intratumoral, intradermal, intraperitoneal, and subcutaneous routes, include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. In the provided methods, compositions can be administered, for example, by intravenous infusion, orally, topically, intraperitoneally, intravesically intratumorally, or intrathecally. Parenteral administration, intratumoral administration, and intravenous administration are the preferred methods of administration. The formulations of compounds can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials.


Injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. Cells transduced or infected by adenovirus or transfected with nucleic acids for ex vivo therapy can also be administered intravenously or parenterally as described above.


In some examples, the pharmaceutical preparation is in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component. Thus, the pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration. For example, unit dosage forms suitable for oral administration include, but are not limited to, powder, tablets, pills, capsules and lozenges.


VII. Methods of Treatment

The recombinant adenoviruses, recombinant adenovirus genomes and compositions disclosed herein can be administered for therapeutic or prophylactic treatment. In particular, provided are methods of reducing or inhibiting tumor cell viability or growth in a subject, reducing or inhibiting tumor progression in a subject, reducing the number of metastases, reducing the size and/or volume of a metastasis, reducing tumor size and/or volume in a subject and/or treating cancer in a subject. The methods include administering a therapeutically effective amount of a recombinant adenovirus or recombinant adenovirus genome (or composition thereof) to the subject. As described throughout, the adenovirus or pharmaceutical composition is administered in any number of ways including, but not limited to, intravenously, intravascularly, intrathecally, intramuscularly, subcutaneously, intratumorally, intraperitoneally, or orally. Optionally, the method further comprising administering to the subject one or more additional therapeutic agents, such as a chemotherapeutic agent, biologic, and/or radiation.


In some embodiments, the cancer or tumor is a lung, prostate, colorectal, breast, thyroid, renal, pancreas, bone, head and neck, or liver cancer or tumor, or is a type of leukemia. In some cases, the cancer is metastatic. In some examples, the tumor is a tumor of the mammary, pituitary, thyroid, or prostate gland; a tumor of the brain, liver, meninges, bone, ovary, uterus, or cervix; monocytic or myelogenous leukemia; adenocarcinoma, adenoma, astrocytoma, bladder tumor, brain tumor, Burkitt's lymphoma, breast carcinoma, cervical carcinoma, colon carcinoma, kidney carcinoma, liver carcinoma, lung carcinoma, ovarian carcinoma, pancreatic carcinoma, prostate carcinoma, rectal carcinoma, skin carcinoma, stomach carcinoma, testis carcinoma, thyroid carcinoma, chondrosarcoma, choriocarcinoma, fibroma, fibrosarcoma, glioblastoma, glioma, hepatoma, histiocytoma, leiomyoblastoma, leiomyosarcoma, lymphoma, liposarcoma cell, mammary tumor, medulloblastoma, myeloma, plasmacytoma, neuroblastoma, neuroglioma, osteogenic sarcoma, pancreatic tumor, pituitary tumor, retinoblastoma, rhabdomyosarcoma, sarcoma, testicular tumor, thymoma, or Wilms tumor. Tumors include both primary and metastatic solid tumors, including carcinomas of breast, colon, rectum, lung, oropharynx, hypopharynx, esophagus, stomach, pancreas, liver, gallbladder and bile ducts, small intestine, urinary tract (including kidney, bladder and urothelium), female genital tract, (including cervix, uterus, and ovaries as well as choriocarcinoma and gestational trophoblastic disease), male genital tract (including prostate, seminal vesicles, testes and germ cell tumors), endocrine glands (including the thyroid, adrenal, and pituitary glands), and skin, as well as hemangiomas, melanomas, sarcomas (including those arising from bone and soft tissues as well as Kaposi's sarcoma) and tumors of the brain, nerves, eyes, and meninges (including astrocytomas, gliomas, glioblastomas, retinoblastomas, neuromas, neuroblastomas, Schwannomas, and meningiomas). In some aspects, solid tumors may be treated that arise from hematopoietic malignancies such as leukemias (i.e. chloromas, plasmacytomas and the plaques and tumors of mycosis fungoides and cutaneous T-cell lymphoma/leukemia) as well as in the treatment of lymphomas (both Hodgkin's and non-Hodgkin's lymphomas). In addition, treatments may be useful in the prevention of metastases from the tumors described herein.


In therapeutic applications, recombinant adenoviruses or compositions thereof are administered to a subject in a therapeutically effective amount or dose. Amounts effective for this use will depend upon the severity of the disease and the general state of the patient's health. Single or multiple administrations of the compositions may be administered depending on the dosage and frequency as required and tolerated by the patient. A “patient” or “subject” includes both humans and other animals, particularly mammals. Thus, the methods are applicable to both human therapy and veterinary applications, such as mice, rats, rabbits, cats, dogs, cows, horses, pigs, chickens, and the like.


An effective amount of an adenovirus having a modified sequence is determined on an individual basis and is based, at least in part, on the particular recombinant adenovirus used; the individual's size, age, gender; and the size and other characteristics of the proliferating cells. For example, for treatment of a human, at least 103 plaque forming units (PFU) of a recombinant virus is used, such as at least 104, at least 105, at least 106, at least 107, at least 108, at least 109, at least 1010, at least 1011, or at least 1012 PFU, for example approximately 103 to 1012 PFU of a recombinant virus is used, depending on the type, size and number of proliferating cells or neoplasms present. The effective amount can be from about 1.0 pfu/kg body weight to about 1015 pfu/kg body weight (e.g., from about 102 pfu/kg body weight to about 1013 pfu/kg body weight). A recombinant adenovirus is administered in a single dose or in multiple doses (e.g., two, three, four, six, or more doses). Multiple doses can be administered concurrently or consecutively (e.g., over a period of days or weeks).


In some embodiments, the provided methods include administering to the subject one or more additional therapeutic agents, such as an anti-cancer agent or other therapeutic treatment (such as surgical resection of the tumor). Exemplary anti-cancer agents include, but are not limited to, chemotherapeutic agents, such as, for example, mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, anti-survival agents, biological response modifiers, anti-hormones (e.g. anti-androgens), anti-angiogenesis agents and CDK inhibitors. Other anti-cancer treatments include radiation therapy and other antibodies that specifically target cancer cells.


Non-limiting examples of alkylating agents include nitrogen mustards (such as mechlorethamine, cyclophosphamide, melphalan, uracil mustard or chlorambucil), alkyl sulfonates (such as busulfan), nitrosoureas (such as carmustine, lomustine, semustine, streptozocin, or dacarbazine).


Non-limiting examples of antimetabolites include folic acid analogs (such as methotrexate), pyrimidine analogs (such as 5-FU or cytarabine), and purine analogs, such as mercaptopurine or thioguanine.


Non-limiting examples of natural products include vinca alkaloids (such as vinblastine, vincristine, or vindesine), epipodophyllotoxins (such as etoposide or teniposide), antibiotics (such as dactinomycin, daunorubicin, doxorubicin, bleomycin, plicamycin, or mitomycin C), and enzymes (such as L-asparaginase).


Non-limiting examples of miscellaneous agents include platinum coordination complexes (such as cis-diamine-dichloroplatinum II also known as cisplatin), substituted ureas (such as hydroxyurea), methyl hydrazine derivatives (such as procarbazine), and adrenocrotical suppressants (such as mitotane and aminoglutethimide).


Non-limiting examples of hormones and antagonists include adrenocorticosteroids (such as prednisone), progestins (such as hydroxyprogesterone caproate, medroxyprogesterone acetate, and magestrol acetate), estrogens (such as diethylstilbestrol and ethinyl estradiol), antiestrogens (such as tamoxifen), and androgens (such as testerone proprionate and fluoxymesterone).


Examples of the most commonly used chemotherapy drugs include Adriamycin, Alkeran, Ara-C, BiCNU, Busulfan, CCNU, Carboplatinum, Cisplatinum, Cytoxan, Daunorubicin, DTIC, 5-FU, Fludarabine, Hydrea, Idarubicin, Ifosfamide, Methotrexate, Mithramycin, Mitomycin, Mitoxantrone, Nitrogen Mustard, Taxol (or other taxanes, such as docetaxel), Velban, Vincristine, VP-16, while some more newer drugs include Gemcitabine (Gemzar), Herceptin, Irinotecan (Camptosar, CPT-11), Leustatin, Navelbine, Rituxan STI-571, Taxotere, Topotecan (Hycamtin), Xeloda (Capecitabine), Zevelin and calcitriol.


Non-limiting examples of immunomodulators that can be used include AS-101 (Wyeth-Ayerst Labs.), bropirimine (Upjohn), gamma interferon (Genentech), GM-CSF (granulocyte macrophage colony stimulating factor; Genetics Institute), IL-2 (Cetus or Hoffman-LaRoche), human immune globulin (Cutter Biological), IMREG (from Imreg of New Orleans, La.), SK&F 106528, and TNF (tumor necrosis factor; Genentech).


CDK (Cyclin-dependent kinase) inhibitors are agents that inhibit the function of CDKs. Non-limiting examples of CDK inhibitors for use in the provided methods include AG-024322, AT7519, AZD5438, flavopiridol, indisulam, P1446A-05, PD-0332991, and P276-00 (see e.g., Lapenna et al., Nature Reviews, 8:547-566, 2009). Other CDK inhibitors include LY2835219, Palbociclib, LEE011 (Novartis), pan-CDK inhibitor AT7519, seliciclib, CYC065, butyrolactone I, hymenialdisine, SU9516, CTNK4, PD0183812 or fascaplysin.


In some examples, the CDK inhibitor is a broad-range inhibitor (such as flavopiridol, olomoucine, roscovitine, kenpaullone, SNS-032, AT7519, AG-024322, (S)-Roscovitine or R547). In other examples, the CDK inhibitor is a specific inhibitor (such as fascaplysin, ryuvidine, purvalanol A, NU2058, BML-259, SU 9516, PD0332991 or P-276-00).


In some embodiments, the provided methods further include administering to the subject a therapeutically effective amount of an immunotherapy. Non-limiting examples of immunomodulators that can be used include AS-101 (Wyeth-Ayerst Labs.), bropirimine (Upjohn), gamma interferon (Genentech), GM-CSF (granulocyte macrophage colony stimulating factor; Genetics Institute), IL-2 (Cetus or Hoffman-LaRoche), human immune globulin (Cutter Biological), IMREG (from Imreg of New Orleans, La.), SK&F 106528, and TNF (tumor necrosis factor; Genentech). The immunotherpautic agent can be a PD-1 antagonist or a PD-L1 antagonist, such as an antibody that specifically binds PD-1 or PD-L1, such as Atezolizumab, MPDL3280A, BNS-936558 (Nivolumab), Pembrolizumab, Pidilizumab, CT011, AMP-224, AMP-514, MEDI-0680, BMS-936559, BMS935559, MEDI-4736, MPDL-3280A, MSB-0010718C. The immunotherpautic agent can also be a CTLA-4, LAG-3, or B7-H3 antagonist, such as Tremelimumab, BMS-986016, and MGA271.


In some embodiments, the provided methods further include administering to the subject a therapeutically effective amount of one or more anti-angiogenic agents, such as proteins, enzymes, polysaccharides, oligonucleotides, DNA, RNA, and recombinant vectors, and small molecules that function to reduce or even inhibit blood vessel growth. Examples of suitable angiogenesis inhibitors include, without limitation, angiostatin K1-3, staurosporine, genistein, fumagillin, medroxyprogesterone, suramin, interferon-alpha, metalloproteinase inhibitors, platelet factor 4, somatostatin, thromobospondin, endostatin, thalidomide, and derivatives and analogs thereof. For example, in some embodiments the anti-angiogenesis agent is an antibody that specifically binds to VEGF (e.g., AVASTIN®, Roche) or a VEGF receptor (e.g., a VEGFR2 antibody). In one example the anti-angiogenic agent includes a VEGFR2 antibody, or DMXAA (also known as Vadimezan or ASA404; available commercially, e.g., from Sigma Corp., St. Louis, Mo.) or both. The anti-angiogenic agent can be bevacizumab, sunitinib, an anti-angiogenic tyrosine kinase inhibitors (TKI), such as sunitinib, xitinib and dasatinib. These can be used individually or in any combination.


In some embodiments, the provided methods further include administering to the subject a therapeutically effective amount of one or more kinase inhibitors, such as GLEEVAC®, IRESSA®, and TARCEVA®, sunitinib, sorafenib, anitinib, and dasatinib that prevent phosphorylation and activation of growth factors. Antibodies that can be used include HERCEPTIN® and AVASTIN® that block growth factors and the angiogenic pathway. These can be used individually or in combination.


In some embodiments, the provided methods further include administering to the subject a therapeutically effective amount of one or more therapeutic monoclonal antibodies, for example, 3F8, Abagovomab, Adecatumumab, Afutuzumab, Alacizumab, Alemtuzumab, Altumomab pentetate, Anatumomab mafenatox, Apolizumab, Arcitumomab, Bavituximab, Bectumomab, Belimumab, Besilesomab, Bevacizumab, Bivatuzumab mertansine, Blinatumomab, Brentuximab vedotin, Cantuzumab mertansine, Capromab pendetide, Catumaxomab, CC49, Cetuximab, Citatuzumab bogatox, Cixutumumab, Clivatuzumab tetraxetan, Conatumumab, Dacetuzumab, Detumomab, Ecromeximab, Eculizumab, Edrecolomab, Epratuzumab, Ertumaxomab, Etaracizumab, Farletuzumab, Figitumumab, Galiximab, Gemtuzumab ozogamicin, Girentuximab, Glembatumumab vedotin, Ibritumomab tiuxetan, Igovomab, Imciromab, Intetumumab, Inotuzumab ozogamicin, Ipilimumab, Iratumumab, Labetuzumab, Lexatumumab, Lintuzumab, Lorvotuzumab mertansine, Lucatumumab, Lumiliximab, Mapatumumab, Matuzumab, Mepolizumab, Metelimumab, Milatuzumab, Mitumomab, Morolimumab, Nacolomab tafenatox, Naptumomab estafenatox, Necitumumab, Nimotuzumab, Nofetumomab merpentan, Ofatumumab, Olaratumab, Oportuzumab monatox, Oregovomab, Panitumumab, Pemtumomab, Pertuzumab, Pintumomab, Pritumumab, Ramucirumab, Rilotumumab, Rituximab, Robatumumab, Satumomab pendetide, Sibrotuzumab, Sonepcizumab, Tacatuzumab tetraxetan, Taplitumomab paptox, Tenatumomab, TGN1412, Ticilimumab (tremelimumab), Tigatuzumab, TNX-650, Trastuzumab, Tremelimumab, Tucotuzumab celmoleukin, Veltuzumab, Volociximab, Votumumab, or Zalutumumab. In some examples, the heterologous ORF encodes one of these therapeutic antibodies.


Another common treatment for some types of cancer is surgical treatment, for example surgical resection of the cancer or a portion of it. Another example of a treatment is radiotherapy, for example administration of radioactive material or energy (such as external beam therapy) to the tumor site to help eradicate the tumor or shrink it prior to surgical resection.


The choice of agent and dosage can be determined readily by one of skill in the art based on the given disease being treated. Combinations of agents or compositions can be administered either concomitantly (e.g., as a mixture), separately but simultaneously (e.g., via separate intravenous lines) or sequentially (e.g., one agent is administered first followed by administration of the second agent). Thus, the term combination is used to refer to concomitant, simultaneous or sequential administration of two or more agents or compositions.


According to the methods disclosed herein, the subject is administered an effective amount of one or more of the agents provided herein. The effective amount is defined as any amount necessary to produce a desired physiologic response (e.g., killing of a cancer cell). Therapeutic agents are typically administered at the initial dosage of about 0.001 mg/kg to about 1000 mg/kg daily. A dose range of about 0.01 mg/kg to about 500 mg/kg, or about 0.1 mg/kg to about 200 mg/kg, or about 1 mg/kg to about 100 mg/kg, or about 10 mg/kg to about 50 mg/kg, can be used. The dosages, however, may be varied depending upon the requirements of the subject, the severity of the condition being treated, and the compound being employed. For example, dosages can be empirically determined considering the type and stage of cancer diagnosed in a particular subject. The dose administered to a subject, in the context of the provided methods should be sufficient to affect a beneficial therapeutic response in the patient over time. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Thus, effective amounts and schedules for administering the agent may be determined empirically by one skilled in the art. The dosage should not be so large as to cause substantial adverse side effects, such as unwanted cross-reactions, anaphylactic reactions, and the like. The dosage can be adjusted by the individual physician in the event of any contraindications. Guidance can be found in the literature for appropriate dosages for given classes of pharmaceutical products.


Provided herein is a method of inhibiting tumor cell viability or growth by contacting the tumor cell with a recombinant adenovirus, a recombinant adenovirus genome, or composition thereof, as disclosed herein. In some embodiments, the method is an in vitro method. In other embodiments, the method is an in vivo method and contacting the tumor cell comprises administering the recombinant adenovirus, recombinant adenovirus genome or composition to a subject with a tumor.


Further provided is a method of inhibiting tumor progression or reducing tumor volume in a subject, by administering to the subject a therapeutically effective amount of a recombinant adenovirus or recombinant adenovirus genome (or composition thereof) disclosed herein.


Also provided is a method of treating cancer in a subject, by administering to the subject a therapeutically effective amount of a recombinant adenovirus or recombinant adenovirus genome (or composition thereof) disclosed herein.


VIII. Adsembly and AdSLIC

The adenovirus genome is organized into several functional groups, labeled E1, E2, E3, E4, and L1-5. The E1 region encodes proteins that control the transcription of all other viral genes and induces S-phase in the host cell. The E2 region encodes proteins that drive viral DNA replication. The E3 region proteins modulate host cell immune response and are dispensable in cell culture. The E4 region contains genes for a disparate set of functions. And the L1-5 region encodes the viral particle structural proteins.


Taking advantage of this natural segregation of functionality, the inventors previously developed a method of recombinant adenovirus assembly that allows quick and easy manipulation of the 36 kb Ad genome by separating it into 4 plasmids, E1, E3, E4, and Core, as shown in FIG. 6A (Adsembly and AdSLIC; see WO 2012/024351, which is incorporated herein by reference). Because of their more reasonable size, manipulation of these smaller plasmids is straightforward using standard techniques.


Adsembly and AdSLIC enable the combinatorial in vitro assembly of adenoviruses with novel properties from compatible genomic library parts in 4 hours. Adsembly and AdSLIC provide a common genome design platform that enables synthetic viruses with novel properties to be assembled using four libraries of functional parts (FIG. 6A). These libraries of parts can be re-assembled in all possible combinations using either multi-site specific recombination sites (Adsembly; FIG. 6B) or sequence independent seamless cloning (AdSLIC; FIG. 6C).


The Adsembly and AdSLIC technologies enable the modular design and production of adenoviruses with unique capabilities. Developing the capability to design, manufacture, and test viruses in an automated, high-throughput manner will accelerate and expand the development of new viruses for therapeutic, diagnostic, and research studies.


While the cloning step was once the bottleneck for producing new viral constructs, the advent of Adsembly and AdSLIC have made it such that the ability to construct viral genomes has outpaced the ability to test them. An equally high throughput kinetics assay is critical to exploit the full potential and high content assembly of synthetic and personalized viral therapies and diagnostics using the Adsembly and AdSLIC methods.


The following examples are provided to illustrate certain particular features and/or embodiments. These examples should not be construed to limit the disclosure to the particular features or embodiments described.


EXAMPLES
Example 1: Identification of Optimal Locations in the Adenovirus Genome for Exogenous ORFs

This example describes the identification of specific locations within the adenovirus genome where exogenous ORFs can be inserted, along with a self-cleaving peptide sequence, without disrupting virus kinetics.


The insertion of exogenous genes in adenovirus vectors is limited by the size capacity of the adenovirus capsid. Excessive addition of exogenous nucleic acid leads to incomplete genome loading into the capsid and reduced viral kinetics. A solution to the challenge presented by the limited available space in the adenovirus genome is to locate exogenous open reading frames (ORFs) as fusion products within native adenovirus ORFs. This strategy makes use of adenovirus promoters, 5′UTRs, and polyA tails already encoded in the genome. However, expression of a fusion between a native adenovirus protein and an exogenous protein can be deleterious to one or both protein functions and lead to a significant decrease in adenovirus replication kinetics. In fact, studies disclosed herein demonstrate that direct fusion of an exogenous ORF to the adenovirus E1A, DNA polymerase or ADP ORFs significantly inhibits adenovirus replication kinetics. In addition, the inventors previously tried using an internal ribosomal entry site (IRES) to insert exogenous ORFs, which also failed to produce recombinant virus with wild-type kinetics.


This example describes a solution to this problem by using a self-cleaving peptide sequence placed between the native adenovirus ORF and the exogenous ORF. When placed between the two ORFs on a single mRNA, the presence of the self-cleaving peptide sequence leads to ribosome skipping and release of the first protein separate from the second protein. The adenovirus constructs generated in this example use the self-cleaving peptide P2A and a fluorescent protein (e.g. YPet, mCherry) as the heterologous ORF.


The table below provides a list of the constructs that were generated and indicates the expression level of the exogenous ORF (low, medium or high) and the level of virus replication kinetics (low, medium or high) in two different cells lines (293-E4 cells and A549 cells).

















Construct
Designation
SEQ ID NO:
Expression Level
Kinetics in 293-E4 cells
Kinetics in A549 cells




















YPet-GS-ElA
CMBT-352

Low
High
Low


YPet-P2A-E1A
CMBT-379
1
High
High
Medium


E1A-P2A-YPet
CMBT-432
2
Medium
High
Medium


E1A-P2A-YPet-PEST
CMBT-569

Medium
High
Medium


E1A-P2A-mCherry
CMBT-455

Medium
High
Medium


E1B-55k-P2A-YPet
CMBT-456
3
High
High
High


E1B-55k-P2A-mCherry
CMBT-499
4
High
High
High


YPet-P2A-(DNA Poly)
CMBT-530
5
Medium
High
High


YPet-(DNA Poly)
CMBT-590

Medium
None
Not tested


DBP-GS-BFP
CMBT-612

High
High
Not tested


DBP-P2A-YPet
CMBT-886
6
High
High
High


mCherry-GS-ADP
CMBT-402

High
Medium
Not tested


ΔADP[mCherry]
CMBT-599

High
High
Medium


YPet-P2A-ADP
CMBT-403
7
High
High
High


ADP-P2A-YPet
CMBT-429
8
High
Low
None


E3-14.7k-P2A-YPet
PCMN-887
9
High
High
High


Fiber-GS-mCherry
CMBT-368

High
Medium
None


Fiber-GS-tdTomato
CMBT-369

High
None
Not tested


YPet-P2A-Fiber
CMBT-407
12
High
None
Not tested


Fiber-P2A-YPet
CMBT-445
13
Very High
Medium
None


Fiber-P2A-BFP
CMBT-632

Very High
Medium
None


Fiber-GS-P2A-YPet
CMBT-446

Very High
Medium
None


YPet-P2A-E4-ORF2
CMBT-457
10
Medium
High
High


mCherry-P2A-E4-ORF2
CMBT-633
11
Medium
High
High









Constructs exhibiting “high” replication kinetics (i.e. replication kinetics that are comparable to wild-type adenovirus) in both cell types are considered candidates for generating therapeutic adenovirus constructs (show in bold). In addition, the fiber-P2A-heterologous ORF constructs exhibited significant defects in viral replication, but generated extraordinarily high levels of expression of the heterologous protein. Thus, these fiber constructs also can be used in therapeutic applications for production of high levels of a therapeutic protein in the context of a replication defective adenovirus.


Comparison of Direct Fusion and Insertion of a P2A Site


Several constructs were generated in which a fluorescent protein was fused directly to an adenovirus ORF. In particular, the following direct fusions were generated: YPet-E1A, YPet-(DNA polymerase) and mCherry-ADP.


YPet-E1A adenovirus exhibited a significant impairment in virus kinetics. Insertion of the P2A site between YPet and E1A (YPet-P2A-E1A) improved virus kinetics, but did not restore virus kinetics to wild-type level. Another construct was then generated to test fusion of P2A and YPet to the C-terminal end of E1A (E1A-P2A-YPet). This construct further improved virus kinetics, but again did not restore kinetics to the level of wild-type adenovirus.


Multiple attempts at transfecting the YPet-(DNA-poly) genome plasmid failed to produce viable virus (no plaques were formed). However, fusion of YPet-P2A to the N-terminus of DNA polymerase (YPet-P2A-(DNA poly)) produced a virus with wild-type kinetics, as shown in the table above.


In addition, the direct fusion of mCherry to ADP (mCherry-ADP) produced a virus with significantly impaired kinetics. However, insertion of the P2A site between the mCherry ORF and the ADP ORF resulted in a virus with wild-type kinetics (mCherry-P2A-ADP). The same result was obtained using a different fluorescent protein; the YPet-P2A-ADP construct exhibited wild-type virus kinetics. However, placement of P2A and the heterologous ORF on the C-terminal side of ADP produced a virus that did not replicate. Thus, for ADP, the heterologous ORF must be placed at the N-terminus.


Both direct fusion to fiber and insertion of a P2A site between fiber and a heterologous ORF produced viruses with significantly impaired replication kinetics. However, the recombinant adenoviruses comprising fiber-P2A-heterologous ORF exhibited extraordinarily high expression levels of the heterologous protein (YPet or blue fluorescent protein (BFP) in this example).


Additional Constructs with Wild-Type Virus Kinetics



FIG. 7 shows a comparison of Ln-Slope of six different constructs: YPet-E1A, YPet-P2A-E1A, E1A-P2A-mCherry, E1B-55k-P2A-YPet, YPet-P2A-ADP and Fiber-P2A-YPet. As discussed above, direct fusion of YPet to E1A produced a virus with significantly impaired kinetics, and addition of the P2A site at either the N-terminus (YPet-P2A-E1A) or the C-terminus (E1A-P2A-mCherry) improved virus kinetics but not to wild-type levels. However, inserting the P2A site and a heterologous ORF at the C-terminus of E1B-55k (E1B-55k-P2A-YPet) or the N-terminus of ADP (YPet-P2A-ADP) generated a recombinant virus with wild-type virus kinetics.


Evaluation of viral kinetics for constructs having a P2A site and heterologous ORF on the C-terminus of DBP (DBP-P2A-YPet) or the C-terminus of E3-14.7k (E3-14.7k-P2A-YPet), or having a P2A site and heterologous ORF on the N-terminus of E4-ORF2 (YPet-P2A-E4-ORF2 and mCherry-P2A-E4-ORF2) produced viruses with wild-type replication kinetics.


The results of these data demonstrate that at least the following adenovirus genome constructs can be used to develop therapeutic adenovirus constructs:


E1B-55k-P2A-heterologous ORF


heterologous ORF-P2A-(DNA polymerase)


DBP-SC-heterologous ORF


heterologous ORF-SC-ADP


E3-14.7k-SC-heterologous ORF


heterologous ORF-P2A-E4-ORF2


fiber-P2A-heterologous ORF


For therapeutic applications, the heterologous ORF encodes a therapeutic protein.


Other Adenovirus Serotypes


Previously described methods of measuring viral kinetics are all highly dependent upon cell-type specific assays and are thus serotype specific due to the divergent tropism of each adenovirus serotype. The adenovirus kinetic assay disclosed herein is not dependent upon any one cell type and so can be extended to serotypes other than Ad5. All adenovirus serotypes contain an ORF equivalent to Ad5 E3-14.7k. Therefore, viruses equivalent to Ad5 E3-14.7k-P2A-YPet (PCMN-887; SEQ ID NO: 9) were generated using Ad9 (containing E3-15k) and Ad34 (containing E3-14.8k): PCMN-888 (Ad9 E3-15k-P2A-YPet; SEQ ID NO: 22) and PCMN-889 (Ad34 E3-14.8k-P2A-YPet; SEQ ID NO: 23). Chimeric viruses containing the Ad5 core and a fiber shaft and knob from either Ad9 or Ad34 were also generated. The four recombinant viruses were then tested in the FBVK assay using 293 cells (FIG. 9A), A549 cells (FIG. 9B) and U2OS cells (FIG. 9C). All four recombinant viruses exhibited high levels of YPet expression with minimal impact on viral kinetics resulting from insertion of the exogenous ORF.


Example 2: Methods for Evaluating Adenovirus Replication Kinetics

The Adsembly and AdSLIC methods for assembling recombinant adenoviruses provide a means for generating large numbers of recombinant virus genomes and viruses in a short period of time. However, a need exists for a rapid and high-throughput method for evaluating replication kinetics of recombinant adenoviruses designed for clinical and therapeutic use. This example describes a fluorescence-based viral kinetics assay that can be used to test virus replication kinetics of recombinant adenoviruses (FIG. 3). The assay can be performed with either recombinant adenovirus genome plasmids or recombinant adenovirus particles as the starting material.


When starting with a recombinant adenovirus genome, the assay includes transfecting cells with adenovirus genome plasmids (such as those described above in Example 1) and monitoring fluorophore expression over time (FIGS. 4A-4B). Transfection conditions are selected such that about 5-10% of the cells are initially transfected. Cells that are not initially transfected are available for secondary infection by virus particles produced from the initial transfection. Log-slope is used as a measure of kinetics based on secondary, tertiary, and quarternary (etc.) infections, thus it is not necessary to know the percentage of cells that are initially transfected. FIGS. 4A and 4B show an exemplary virus-based kinetics assay starting with recombinant adenovirus genome plasmids. In this example, a 48-well plate is used, which allows for testing of 14 different virus constructs (in triplicate) simultaneously. The upper half of the 48-well plate (FIG. 4A) includes triplicate wells of six different viruses, 3 mock-infected wells and 3 “blank” wells with FLUORESBRITE™ beads, which compensate for tool sensitivity drift. The lower half of the 48-well plate (FIG. 4B) includes triplicate wells of eight different virus constructs. Once cells are transfected, the plate is placed in a TECAN™ plate reader for continuous fluorescence monitoring. The data collected is used to calculate ln-slope for each construct (FIG. 8).


The assay can also be carried out by infecting cells with recombinant virus particles. In this version of the assay, cells are infected with recombinant virus particles and fluorophore expression is monitored over time (FIG. 5). As with the genome plasmid version of the assay, it is not necessary to know the exact titer of the starting virus stock. Typically, a dilution series is used for initial infection, such as a dilution series ranging from 1:100 to 1:218,700, as shown in FIG. 5. A dilution of 1:100 generally leads to infection of all cells, whereas a dilution of 1:218,700 generally leads to initial infection of very few cells. In this example, a 96-well plate is used and 11 different virus constructs are tested simultaneously at eight different dilutions (1:100, 1:300, 1:900, 1:2700, 1:8100, 1:24,300, 1:72,900 and 1:218,700). The plate also includes four wells of mock-infected cells and four wells of FLUORESBRITE™ beads. Once the cells are infected, the plate is placed in a TECAN™ plate reader for continuous fluorescence monitoring. The data collected is used to calculate ln-slope for each construct (FIG. 8).


The TECAN™ plate readers also provide incubation functions (maintaining an appropriate temperature as well as CO2 and O2 levels). Data points are taken every 15 minutes to calculate the ln-slope. Using these methods, it is possible to rapidly and efficiently compare the kinetics between a number of different viruses and between different cell types. For example, to evaluate whether particular recombinant adenoviruses could be used therapeutically as oncolytic viruses, this assay could be employed to find viruses that exhibit high replication kinetics in tumor cells, but slow virus kinetics in non-tumor cells. Furthermore, the virus kinetics of the recombinant viruses can be evaluated by infecting or transfecting the tumor cell type of interest in this assay.


Calculating Log-Slope


To measure log-slope, the linear plot of fluorescence intensity versus time is converted to a semi-log plot by taking the natural logarithm of the measured fluorescence intensity at each time point. Since the fluorescence intensity exhibits exponential growth during viral replication, this conversion results in a straight line when plotting ln(fluorescence intensity) vs. time. This straight line is then fit using standard least-squares methods. The resulting slope produced by this fit is the ln-slope of the fluorescence vs. time and thus the ln-slope of the viral growth vs. time. Equations are shown below.

FI(t)=F0eα(t−t0);

where FI is measured fluorescence intensity, t is time, F0 is the initial fluorescence intensity at time=t0, and α is the ln-slope.


Take natural logarithm of both sides:

ln[FI(t)]=ln[F0eα(t−t0)]=ln(F0)+α(t−t0)


The right hand side is now a linear equation with a ln-slope of α.


In view of the many possible embodiments to which the principles of the disclosure may be applied, it should be recognized that the illustrated embodiments are only examples of the invention and should not be taken as limiting the scope of the invention. Rather, the scope of the invention is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.

Claims
  • 1. A recombinant adenovirus genome, comprising a heterologous open reading frame (ORF) and a self-cleaving peptide coding sequence, both operably linked to and in the same reading frame as an endogenous adenovirus ORF, wherein the self-cleaving peptide coding sequence is located between the heterologous ORF and the endogenous ORF, and wherein: the endogenous ORF is E1B-55k and the heterologous ORF is 3′ of E1B-55k;the endogenous ORF is DNA polymerase and the heterologous ORF is 5′ of DNA polymerase;the endogenous ORF is DNA-binding protein (DBP) and the heterologous ORF is 3′ of DBP;the endogenous ORF is adenovirus death protein (ADP) and the heterologous ORF is 5′ of ADP;the endogenous ORF is E3-14.7k and the heterologous ORF is 3′ of E3-14.7k;the endogenous ORF is E4-ORF2 and the heterologous ORF is 5′ of E4-ORF2; orthe endogenous ORF is fiber and the heterologous ORF is 3′ of fiber.
  • 2. The recombinant adenovirus genome of claim 1, wherein the self-cleaving peptide is a 2A peptide or variant thereof.
  • 3. The recombinant adenovirus genome of claim 2, wherein the 2A peptide comprises a porcine teschovirus-1 (PTV1) 2A (P2A) peptide, a foot and mouth disease virus (FMDV) 2A (F2A) peptide, an equine rhinitis A virus (ERAV) 2A (E2A) peptide or a Thosea asigna virus (TaV) 2A (T2A) peptide, or a variant thereof.
  • 4. The recombinant adenovirus genome of claim 3, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 5. The recombinant adenovirus genome of claim 3, wherein the self-cleaving peptide comprises the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 6. The recombinant adenovirus genome of claim 1, wherein the heterologous ORF encodes a fluorescent protein.
  • 7. The recombinant adenovirus genome of claim 6, wherein the fluorescent protein is a green fluorescent protein (GFP) a yellow fluorescent protein (YFP), a red fluorescent protein (RFP) or a blue fluorescent protein (BFP).
  • 8. The recombinant adenovirus genome of claim 7, wherein the YFP is YPet.
  • 9. The recombinant adenovirus genome of claim 7, wherein the RFP is mCherry.
  • 10. The recombinant adenovirus genome of claim 6, comprising in the 5′ to 3′ direction: E1B-55K-P2A-YPet;E1B-55K-P2A-mCherry;YPet-P2A-(DNA polymerase);DBP-P2A-YPet;YPet-P2A-ADP;E3-14.7k-P2A-YPet;YPet-P2A-E4-ORF2;mCherry-P2A-E4-ORF2; orFiber-P2A-YPet.
  • 11. The recombinant adenovirus genome of claim 10, comprising the nucleotide sequence of any one of SEQ ID NOs: 3-7, 9-11 and 13.
  • 12. A composition comprising the recombinant adenovirus genome of claim 1 and a pharmaceutically acceptable carrier.
  • 13. A recombinant adenovirus comprising the recombinant adenovirus genome of claim 1.
  • 14. A kit comprising: (i) the recombinant adenovirus genome of claim 1; and(ii) one or more diagnostic agents.
  • 15. The kit of claim 14, wherein the one or more diagnostic agents comprises one or more antibodies that bind a tumor marker.
  • 16. The kit of claim 14, wherein the one or more diagnostic agents comprises an imaging probe.
  • 17. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is E1B-55k and the heterologous ORF is 3′ of E1B-55k.
  • 18. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is DNA polymerase and the heterologous ORF is 5′ of DNA polymerase.
  • 19. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is DBP and the heterologous ORF is 3′ of DBP.
  • 20. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is ADP and the heterologous ORF is 5′ of ADP.
  • 21. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is E3-14.7k and the heterologous ORF is 3′ of E3-14.7k.
  • 22. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is E4-ORF2 and the heterologous ORF is 5′ of E4-ORF2.
  • 23. The recombinant adenovirus genome of claim 1, wherein the endogenous ORF is fiber and the heterologous ORF is 3′ of fiber.
  • 24. The recombinant adenovirus genome of claim 17, wherein the heterologous ORF encodes a fluorescent protein.
  • 25. The recombinant adenovirus genome of claim 18, wherein the heterologous ORF encodes a fluorescent protein.
  • 26. The recombinant adenovirus genome of claim 19, wherein the heterologous ORF encodes a fluorescent protein.
  • 27. The recombinant adenovirus genome of claim 20, wherein the heterologous ORF encodes a fluorescent protein.
  • 28. The recombinant adenovirus genome of claim 21, wherein the heterologous ORF encodes a fluorescent protein.
  • 29. The recombinant adenovirus genome of claim 22, wherein the heterologous ORF encodes a fluorescent protein.
  • 30. The recombinant adenovirus genome of claim 23, wherein the heterologous ORF encodes a fluorescent protein.
  • 31. The recombinant adenovirus genome of claim 24, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 32. The recombinant adenovirus genome of claim 25, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 33. The recombinant adenovirus genome of claim 26, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 34. The recombinant adenovirus genome of claim 27, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 35. The recombinant adenovirus genome of claim 28, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 36. The recombinant adenovirus genome of claim 29, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 37. The recombinant adenovirus genome of claim 30, wherein the amino acid sequence of the self-cleaving peptide is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 38. The recombinant adenovirus genome of claim 27, wherein the self-cleaving peptide is a 2A peptide or variant thereof.
  • 39. The recombinant adenovirus genome of claim 38, wherein the 2A peptide comprises a porcine teschovirus-1 (PTV1) 2A (P2A) peptide, a foot and mouth disease virus (FMDV) 2A (F2A) peptide, an equine rhinitis A virus (ERAV) 2A (E2A) peptide or a Thosea asigna virus (TaV) 2A (T2A) peptide.
  • 40. The recombinant adenovirus genome of claim 27, wherein the amino acid sequence of the self-cleaving peptide comprises the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 41. The recombinant adenovirus genome of claim 27, wherein the amino acid sequence of the self-cleaving peptide consists of the amino acid sequence of any one of SEQ ID NOs: 14-21.
  • 42. The recombinant adenovirus genome of claim 27, wherein the self-cleaving peptide is P2A.
  • 43. The recombinant adenovirus genome of claim 1, wherein the recombinant adenovirus genome comprises in the 5′ to 3′ direction YPet-P2A-ADP.
  • 44. The recombinant adenovirus genome of claim 27, wherein the recombinant adenovirus genome comprises the YPet-P2A-ADP sequence of adenovirus CMBT-403, wherein adenovirus CMBT-403 has a genome of SEQ ID NO: 7.
  • 45. The recombinant adenovirus genome of claim 1, wherein the recombinant adenovirus genome is an Ad5 genome.
  • 46. The recombinant adenovirus genome of claim 43, wherein the recombinant adenovirus genome is an Ad5 genome.
  • 47. The recombinant adenovirus genome of claim 44, wherein the recombinant adenovirus genome is an Ad5 genome.
  • 48. The recombinant adenovirus genome of claim 43, wherein the genome codes for a chimeric fiber protein comprising a fiber shaft from Ad5 and a fiber knob from a second serotype of adenovirus.
  • 49. The recombinant adenovirus genome of claim 45, wherein the genome codes for a chimeric fiber protein comprising a fiber shaft from Ad5 and a fiber knob from a second serotype of adenovirus.
  • 50. The recombinant adenovirus genome of claim 48, wherein the second serotype of adenovirus is Ad9 or Ad34.
  • 51. The recombinant adenovirus genome of claim 49, wherein the second serotype of adenovirus is Ad9 or Ad34.
  • 52. The recombinant adenovirus genome of claim 43, wherein the E4orf6/7 protein coding sequence is deleted.
  • 53. The recombinant adenovirus genome of claim 45, wherein the E4orf6/7 protein coding sequence is deleted.
  • 54. The recombinant adenovirus genome of claim 43, wherein the recombinant adenovirus genome is an Ad5 genome, wherein the genome codes for a chimeric fiber protein comprising a fiber shaft from Ad5 and a fiber knob from adenovirus Ad9 or Ad34, and wherein the E4orf6/7 protein coding sequence is deleted.
  • 55. A recombinant adenovirus comprising the recombinant adenovirus genome of claim 43.
  • 56. A recombinant adenovirus comprising the recombinant adenovirus genome of claim 46.
  • 57. A recombinant adenovirus comprising the recombinant adenovirus genome of claim 54.
  • 58. A composition comprising the recombinant adenovirus genome of claim 1 and a pharmaceutically acceptable carrier.
  • 59. A composition comprising the recombinant adenovirus genome of claim 43 and a pharmaceutically acceptable carrier.
  • 60. A composition comprising the recombinant adenovirus genome of claim 46 and a pharmaceutically acceptable carrier.
  • 61. A composition comprising the recombinant adenovirus genome of claim 54 and a pharmaceutically acceptable carrier.
  • 62. The recombinant adenovirus genome of claim 27, wherein the genome codes for a chimeric fiber protein comprising a fiber shaft from Ad5 and a fiber knob from a second serotype of adenovirus.
  • 63. The recombinant adenovirus genome of claim 62, wherein the second serotype of adenovirus is Ad9 or Ad34.
  • 64. The recombinant adenovirus genome of claim 63, wherein the E4orf6/7 protein coding sequence is deleted.
  • 65. The recombinant adenovirus genome of claim 64, wherein the recombinant adenovirus genome comprises in the 5′ to 3′ direction YPet-P2A-ADP.
  • 66. A recombinant adenovirus comprising the recombinant adenovirus genome of claim 65.
  • 67. A composition comprising the recombinant adenovirus genome of claim 65 and a pharmaceutically acceptable carrier.
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 16/110,207, filed Aug. 23, 2018, issued as U.S. Pat. No. 10,738,325 on Aug. 11, 2020, which is a continuation of PCT International Application No. PCT/US2017/019086, filed Feb. 23, 2017, published in English under PCT Article 21(2), which claims the benefit of U.S. Provisional Application No. 62/298,653, filed Feb. 23, 2016. The above-referenced applications are herein incorporated by reference in their entirety.

US Referenced Citations (362)
Number Name Date Kind
5559099 Wickham et al. Sep 1996 A
5670488 Gregory et al. Sep 1997 A
5677178 McCormick Oct 1997 A
5731190 Wickham et al. Mar 1998 A
5801029 McCormick Sep 1998 A
5846782 Wickham et al. Dec 1998 A
5846945 McCormick Dec 1998 A
5856181 McCormick Jan 1999 A
5922315 Roy Jul 1999 A
5945335 Colosi Aug 1999 A
5962311 Wickham et al. Oct 1999 A
5965541 Wickham et al. Oct 1999 A
5972706 McCormick Oct 1999 A
6020172 Both Feb 2000 A
6069134 Roth et al. May 2000 A
6127525 Crystal et al. Oct 2000 A
6133243 Kirn Oct 2000 A
6153435 Crystal et al. Nov 2000 A
6296845 Sampson et al. Oct 2001 B1
6329190 Wickham et al. Dec 2001 B1
6410010 Zhang et al. Jun 2002 B1
6455314 Wickham et al. Sep 2002 B1
6465253 Wickham et al. Oct 2002 B1
6475480 Mehtali et al. Nov 2002 B1
6506379 Clackson et al. Jan 2003 B1
6506602 Stemmer Jan 2003 B1
6569677 Legrand et al. May 2003 B1
6596268 Coffey et al. Jul 2003 B1
6635466 Davidson et al. Oct 2003 B2
6635476 Murphy Oct 2003 B1
6649157 Coffey et al. Nov 2003 B2
6737234 Freimuth May 2004 B1
6740525 Roelvink et al. May 2004 B2
6797702 Roth et al. Sep 2004 B1
6811774 Haddada et al. Nov 2004 B2
6824771 Curiel et al. Nov 2004 B1
6838285 Farmer et al. Jan 2005 B2
6841540 Curiel et al. Jan 2005 B1
6849446 Tikoo et al. Feb 2005 B2
6867022 Imperiale Mar 2005 B1
6869936 Vogels et al. Mar 2005 B1
6878549 Vogels et al. Apr 2005 B1
6905678 Havenga et al. Jun 2005 B2
6911199 Vigne et al. Jun 2005 B2
6911200 Yu et al. Jun 2005 B2
6913922 Bout et al. Jul 2005 B1
6929946 Vogels et al. Aug 2005 B1
6951755 Wickham et al. Oct 2005 B2
6984635 Schreiber et al. Jan 2006 B1
7001596 Johnson et al. Feb 2006 B1
7045347 Graham et al. May 2006 B2
7094398 Lieber et al. Aug 2006 B1
7094399 Otto Aug 2006 B2
7109179 Roth et al. Sep 2006 B2
7157266 Freimuth et al. Jan 2007 B2
7232899 Von Seggern et al. Jun 2007 B2
7235233 Havenga et al. Jun 2007 B2
7247472 Wilson et al. Jul 2007 B2
7252817 Coffey et al. Aug 2007 B2
7252989 Zhang et al. Aug 2007 B1
7256036 Legrand et al. Aug 2007 B2
7291498 Roy et al. Nov 2007 B2
7297542 Curiel et al. Nov 2007 B2
7306793 Haddada et al. Dec 2007 B2
7332337 van Es et al. Feb 2008 B2
7344711 Bonastre et al. Mar 2008 B2
7344872 Gao et al. Mar 2008 B2
7364727 Li et al. Apr 2008 B2
7410954 Davidson et al. Aug 2008 B2
7456008 Lindholm et al. Nov 2008 B2
7473418 Yu et al. Jan 2009 B2
7482156 Arroyo et al. Jan 2009 B2
7491508 Roy et al. Feb 2009 B2
7510868 Harden et al. Mar 2009 B2
7589069 Wold et al. Sep 2009 B1
7611868 Monaci et al. Nov 2009 B2
7741099 Havenga et al. Jun 2010 B2
7749493 Havenga et al. Jul 2010 B2
7754201 Wang et al. Jul 2010 B2
7906113 Bout et al. Mar 2011 B2
7943373 Fujiwara et al. May 2011 B2
7951585 Ke May 2011 B2
7968333 Yu et al. Jun 2011 B2
8105574 Wilson et al. Jan 2012 B2
8168168 Fueyo et al. May 2012 B2
8231880 Roy et al. Jul 2012 B2
8470310 Roy et al. Jun 2013 B2
8524219 Roy et al. Sep 2013 B2
8603459 Wilson et al. Dec 2013 B2
8685387 Roy et al. Apr 2014 B2
8715642 Kochanek et al. May 2014 B2
8765146 Bruder et al. Jul 2014 B2
8765463 Harden et al. Jul 2014 B2
8808986 Jacobson et al. Aug 2014 B2
8815563 Davis et al. Aug 2014 B2
8834863 Roy et al. Sep 2014 B2
8846031 Roy et al. Sep 2014 B2
8865182 Mayall et al. Oct 2014 B2
8920813 Bruder et al. Dec 2014 B2
8940290 Roy et al. Jan 2015 B2
8974777 Cascallo et al. Mar 2015 B2
9017672 Yu et al. Apr 2015 B2
9018182 Koh et al. Apr 2015 B2
9056090 Colloca et al. Jun 2015 B2
9061055 Fueyo et al. Jun 2015 B2
9133483 Wilson et al. Sep 2015 B2
9163261 Kollipara et al. Oct 2015 B2
9187733 O'Shea et al. Nov 2015 B2
9200041 Lieber et al. Dec 2015 B2
9206238 Roy et al. Dec 2015 B2
9217159 Roy et al. Dec 2015 B2
9217160 O'Shea et al. Dec 2015 B2
9267153 Curiel Feb 2016 B2
9315827 Wang et al. Apr 2016 B2
9359618 Roy et al. Jun 2016 B2
9382551 Roy et al. Jul 2016 B2
9410129 Ranki et al. Aug 2016 B2
9476061 Baker et al. Oct 2016 B2
9493745 Lee et al. Nov 2016 B2
9555089 Shiratsuchi et al. Jan 2017 B2
9593346 Roy et al. Mar 2017 B2
9597363 Roy et al. Mar 2017 B2
9682133 Crystal et al. Jun 2017 B2
9688727 Lieber et al. Jun 2017 B2
9714435 Dicks et al. Jul 2017 B2
9718863 Colloca et al. Aug 2017 B2
9790519 Wei et al. Oct 2017 B2
9885090 O'Shea et al. Feb 2018 B2
9913866 O'3 Shea et al. Mar 2018 B2
10016470 Bonastre et al. Jul 2018 B2
10034905 Seymour et al. Jul 2018 B2
10046067 Yun et al. Aug 2018 B2
10066215 Lee et al. Sep 2018 B2
10071126 Kumon et al. Sep 2018 B2
10077430 Lee et al. Sep 2018 B2
10080774 Fueyo et al. Sep 2018 B2
10113182 Roy et al. Oct 2018 B2
10149873 Roy et al. Dec 2018 B2
10150798 Lieber et al. Dec 2018 B2
10155930 Holm Dec 2018 B2
10232053 Hicklin et al. Mar 2019 B2
10272162 McVey et al. Apr 2019 B2
10294493 Wang et al. May 2019 B2
10316065 Carrió et al. Jun 2019 B2
10376549 Shayakhmetov et al. Aug 2019 B2
10391183 Fueyo-Margareto et al. Aug 2019 B2
10501757 Roy et al. Dec 2019 B2
10538744 Holm Jan 2020 B2
10544192 Colloca et al. Jan 2020 B2
10604549 Alemany Bonastre et al. Mar 2020 B2
10611803 Lieber et al. Apr 2020 B2
10617729 Dobbins Apr 2020 B2
10738325 O'Shea et al. Aug 2020 B2
20010039046 Yeh et al. Nov 2001 A1
20020037274 Williams et al. Mar 2002 A1
20020086411 Holm et al. Jul 2002 A1
20020106382 Young et al. Aug 2002 A1
20020142989 Alemany et al. Oct 2002 A1
20020151069 Korokhov Oct 2002 A1
20020168343 Curiel et al. Nov 2002 A1
20020187128 Imperiale Dec 2002 A1
20020193327 Nemerow Dec 2002 A1
20020193328 Ketner Dec 2002 A1
20030017138 Havenga et al. Jan 2003 A1
20030021768 Shen Jan 2003 A1
20030027338 Freimuth Feb 2003 A1
20030073072 Havenga et al. Apr 2003 A1
20030082146 van Es May 2003 A1
20030082150 Boon Falleur et al. May 2003 A1
20030082811 Orlando et al. May 2003 A1
20030092162 Shankara et al. May 2003 A1
20030095989 Irving et al. May 2003 A1
20030099615 Tikoo May 2003 A1
20030099619 Wickham et al. May 2003 A1
20030104625 Cheng et al. Jun 2003 A1
20030138405 Fueyo et al. Jul 2003 A1
20030143730 Blanche et al. Jul 2003 A1
20030166286 Wickham et al. Sep 2003 A1
20030170899 McVey et al. Sep 2003 A1
20030175244 Curiel et al. Sep 2003 A1
20030175245 Brough et al. Sep 2003 A1
20030215948 Kaleko et al. Nov 2003 A1
20030219899 Korokhov Nov 2003 A1
20030220284 Yotnda et al. Nov 2003 A1
20040002060 Kaleko et al. Jan 2004 A1
20040038205 Van Raaij et al. Feb 2004 A1
20040091456 Nakai et al. May 2004 A1
20040102382 Schughart et al. May 2004 A1
20040146489 Yu et al. Jul 2004 A1
20040175362 Curiel et al. Sep 2004 A1
20040185555 Emini et al. Sep 2004 A1
20040191222 Emini et al. Sep 2004 A1
20040191761 Routes Sep 2004 A1
20040213764 Wold et al. Oct 2004 A1
20040219516 Bennett et al. Nov 2004 A1
20040219543 Wirtz Nov 2004 A1
20040265277 Holm Dec 2004 A1
20050032045 Tikoo et al. Feb 2005 A1
20050036989 Shen et al. Feb 2005 A1
20050079158 Zhou et al. Apr 2005 A1
20050095231 Curiel et al. May 2005 A1
20050095705 Kadan et al. May 2005 A1
20050169891 Vogels et al. Aug 2005 A1
20050181507 Havenga et al. Aug 2005 A1
20050186178 Ennist Aug 2005 A1
20050201936 Wold et al. Sep 2005 A1
20050201978 Lipton Sep 2005 A1
20050232900 Vogels et al. Oct 2005 A1
20050238622 Axelrod et al. Oct 2005 A1
20050260162 Fueyo et al. Nov 2005 A1
20050271622 Zhou et al. Dec 2005 A1
20050277193 Wickham et al. Dec 2005 A1
20050287120 Fisher et al. Dec 2005 A1
20060002893 Vigne et al. Jan 2006 A1
20060034804 Gregory et al. Feb 2006 A1
20060099178 Holm May 2006 A1
20060104953 Havenga et al. May 2006 A1
20060140910 Gregory et al. Jun 2006 A1
20060147420 Fueyo et al. Jul 2006 A1
20060182718 Roth et al. Aug 2006 A1
20060211115 Roy et al. Sep 2006 A1
20060228334 Rosa Calatrava et al. Oct 2006 A1
20060257370 Hermiston et al. Nov 2006 A1
20060281090 Lieber et al. Dec 2006 A1
20060286121 Gall et al. Dec 2006 A1
20060292122 Hermiston et al. Dec 2006 A1
20060292682 Hawkins et al. Dec 2006 A1
20070003923 Nemerow Jan 2007 A1
20070110719 Holm May 2007 A1
20070122817 Church et al. May 2007 A1
20070202080 Yun et al. Aug 2007 A1
20070202524 Murphy Aug 2007 A1
20070253932 Gregory et al. Nov 2007 A1
20070254357 Gregory et al. Nov 2007 A1
20070292396 Fueyo et al. Dec 2007 A1
20070292954 Elledge Dec 2007 A1
20080069836 Nabel et al. Mar 2008 A1
20080089864 Bonastre et al. Apr 2008 A1
20080108129 Pitcovski et al. May 2008 A1
20080112929 Kovesdi et al. May 2008 A1
20080118470 Ennist et al. May 2008 A1
20080124360 Seggern May 2008 A1
20080213220 Fisher et al. Sep 2008 A1
20080242608 Bonni et al. Oct 2008 A1
20080247996 Yu et al. Oct 2008 A1
20080254059 Bett et al. Oct 2008 A1
20090074810 Roy et al. Mar 2009 A1
20090111144 Bebbington Apr 2009 A1
20090202565 Labow et al. Aug 2009 A1
20090232800 Holm Sep 2009 A1
20090280089 Benihoud et al. Nov 2009 A1
20090311219 Bonastre et al. Dec 2009 A1
20100008977 Boulikas et al. Jan 2010 A1
20100034774 Vogels et al. Feb 2010 A1
20100047208 Ke Feb 2010 A1
20100075951 Cardin et al. Mar 2010 A1
20100075998 Vanotti et al. Mar 2010 A1
20100098668 Seth Apr 2010 A1
20100151576 Li et al. Jun 2010 A1
20100233125 Tagawa Sep 2010 A1
20100272753 Ketner et al. Oct 2010 A1
20100292166 Lee et al. Nov 2010 A1
20100310554 Holm Dec 2010 A1
20100311145 Holm Dec 2010 A1
20110053249 Bonastre et al. Mar 2011 A1
20110059135 Kovesdi et al. Mar 2011 A1
20110086063 Crystal et al. Apr 2011 A1
20110104788 Baker et al. May 2011 A1
20110189234 Van Beusechem et al. Aug 2011 A1
20110256524 Lee et al. Oct 2011 A1
20110275093 Holm Nov 2011 A1
20110286999 Holm Nov 2011 A1
20120020924 Nakai et al. Jan 2012 A1
20120039877 Holm Feb 2012 A1
20120207711 Fueyo et al. Aug 2012 A1
20130058897 Lee et al. Mar 2013 A1
20130101557 Yun et al. Apr 2013 A1
20130130292 Szalay May 2013 A1
20130231267 O'Shea et al. Sep 2013 A1
20130243729 O'Shea et al. Sep 2013 A1
20130243731 Dias et al. Sep 2013 A1
20130323205 Diaconu et al. Dec 2013 A1
20130345295 Wang et al. Dec 2013 A1
20140023619 Kosai et al. Jan 2014 A1
20140199688 Mizuguchi et al. Jul 2014 A1
20140294890 Ketner et al. Oct 2014 A1
20140341857 Bressy et al. Nov 2014 A1
20140348791 Barouch et al. Nov 2014 A1
20140377294 Fueyo-Margareto et al. Dec 2014 A1
20140377295 Ertl et al. Dec 2014 A1
20150005397 O'Shea et al. Jan 2015 A1
20150017127 O'Shea et al. Jan 2015 A1
20150071881 Bonastre et al. Mar 2015 A1
20150086579 Mayall et al. Mar 2015 A1
20150202324 Hemminki et al. Jul 2015 A1
20150232880 Hemminki et al. Aug 2015 A1
20150246949 Lieber et al. Sep 2015 A1
20150352203 Wilson et al. Dec 2015 A1
20150374766 O'Shea et al. Dec 2015 A1
20160017294 Reid et al. Jan 2016 A1
20160051603 Roy et al. Feb 2016 A1
20160053235 O'Shea et al. Feb 2016 A1
20160082100 Ranki et al. Mar 2016 A1
20160090574 Fisher et al. Mar 2016 A1
20160102295 Roy et al. Apr 2016 A1
20160143967 Fueyo-Margareto et al. May 2016 A1
20160208287 Hemminki et al. Jul 2016 A1
20160244783 Roy et al. Aug 2016 A1
20160289645 Tufaro et al. Oct 2016 A1
20170035818 Seymour et al. Feb 2017 A1
20170073647 Fisher et al. Mar 2017 A1
20170080069 Cerullo et al. Mar 2017 A1
20170096646 Roy et al. Apr 2017 A1
20170137786 Hemminki et al. May 2017 A1
20170183636 Roy et al. Jun 2017 A1
20170190752 Holm Jul 2017 A1
20170202893 O'Shea et al. Jul 2017 A1
20170252443 Holm Sep 2017 A1
20170314044 Davydova et al. Nov 2017 A1
20170348405 Shiratsuchi et al. Dec 2017 A1
20180000966 Dicks et al. Jan 2018 A1
20180051301 Rentschler et al. Feb 2018 A1
20180072809 Hemminki et al. Mar 2018 A1
20180100164 Wei et al. Apr 2018 A1
20180104288 Galili et al. Apr 2018 A1
20180163190 Gerardy-Schahn et al. Jun 2018 A1
20180216081 Colloca et al. Aug 2018 A1
20180221423 O'Shea et al. Aug 2018 A1
20180318365 Yeung et al. Nov 2018 A1
20180346929 Kosai et al. Dec 2018 A1
20180355374 O'Shea et al. Dec 2018 A1
20180355379 O'Shea et al. Dec 2018 A1
20180369417 Yun et al. Dec 2018 A1
20190055522 Holm Feb 2019 A1
20190062395 Merchant et al. Feb 2019 A1
20190070233 Yeung et al. Mar 2019 A1
20190093085 Tufaro et al. Mar 2019 A1
20190136204 Reid et al. May 2019 A1
20190142967 Hicklin et al. May 2019 A1
20190175716 Gilbert et al. Jun 2019 A1
20190183946 Bonastre et al. Jun 2019 A1
20190201462 Tufaro et al. Jul 2019 A1
20190201551 Curiel Jul 2019 A1
20190233845 Maloveste et al. Aug 2019 A1
20190247452 Lan et al. Aug 2019 A1
20190269794 McVey et al. Sep 2019 A1
20190275092 Tufaro et al. Sep 2019 A1
20190275093 Aboody et al. Sep 2019 A1
20190300905 Ammendola et al. Oct 2019 A1
20190314523 O'Shea et al. Oct 2019 A1
20190314525 O'Shea et al. Oct 2019 A1
20190345204 Carrió et al. Nov 2019 A1
20190350992 Cascallo Piqueras et al. Nov 2019 A1
20190352616 Reid et al. Nov 2019 A1
20190352669 Reid et al. Nov 2019 A1
20190374589 Suzuki et al. Dec 2019 A1
20190388487 Shayakhmetov et al. Dec 2019 A1
20200014798 Hicklin et al. Jan 2020 A1
20200032223 Reid et al. Jan 2020 A1
20200078415 Reid et al. Mar 2020 A1
20200095560 Holm Mar 2020 A1
20200102352 Colloca et al. Apr 2020 A1
Foreign Referenced Citations (171)
Number Date Country
1330715 Jan 2002 CN
1380420 Nov 2002 CN
102191245 Sep 2011 CN
0689447 Apr 1999 EP
0931830 Mar 2001 EP
0760675 Aug 2001 EP
1167533 Jan 2002 EP
1284294 Feb 2003 EP
1413586 Apr 2004 EP
1196616 Dec 2004 EP
1185279 Jan 2005 EP
0851769 Feb 2005 EP
0861329 Mar 2005 EP
1181382 Mar 2005 EP
1121137 Jul 2005 EP
0991763 Sep 2005 EP
1294918 Oct 2005 EP
0889969 Nov 2005 EP
1498129 Nov 2005 EP
1593742 Nov 2005 EP
0920524 Dec 2005 EP
1307573 Jan 2006 EP
0978566 May 2006 EP
0778889 Jul 2006 EP
1070118 Oct 2006 EP
1214098 Nov 2006 EP
1230378 Jun 2007 EP
1550722 Jun 2007 EP
1187919 Nov 2007 EP
0863987 Jan 2008 EP
0920514 Jan 2008 EP
1159438 Jul 2008 EP
1266022 Oct 2008 EP
1678193 Dec 2008 EP
1054064 Dec 2009 EP
2012822 Jan 2010 EP
1816204 Oct 2010 EP
1749098 Dec 2010 EP
1799836 Dec 2010 EP
1816205 Aug 2011 EP
1818408 Aug 2011 EP
1409748 Oct 2011 EP
1180932 Jan 2012 EP
1466001 Apr 2012 EP
1743041 Jun 2012 EP
1446479 Aug 2012 EP
1649028 Aug 2012 EP
1990418 Aug 2012 EP
2311499 Aug 2012 EP
1636370 Apr 2014 EP
1767642 Apr 2014 EP
1689445 Feb 2015 EP
2350269 Sep 2015 EP
2403951 Sep 2015 EP
2643465 May 2016 EP
2428229 Aug 2016 EP
2459716 Aug 2016 EP
2220241 Sep 2016 EP
2325298 Oct 2016 EP
2379586 Nov 2016 EP
2220242 Dec 2016 EP
2774985 Dec 2016 EP
2163260 Mar 2017 EP
2580234 Mar 2017 EP
2798069 Mar 2017 EP
2855685 Mar 2017 EP
2900818 Jun 2017 EP
2301582 Jul 2017 EP
3049520 Jul 2017 EP
1453543 Aug 2017 EP
2463362 Nov 2017 EP
2558481 Dec 2017 EP
2682459 Dec 2017 EP
2714916 Jan 2018 EP
2391638 Jun 2018 EP
2563919 Jun 2018 EP
2971008 Jul 2018 EP
2606137 Aug 2018 EP
2855669 Oct 2018 EP
2986311 Nov 2018 EP
3145537 Dec 2018 EP
2654786 Feb 2019 EP
3280798 Jun 2019 EP
3029144 Jul 2019 EP
3150706 Jul 2019 EP
2809788 Sep 2019 EP
3071697 Oct 2019 EP
3274363 Oct 2019 EP
3460052 Oct 2019 EP
2005-525779 Sep 2005 JP
2008-517627 May 2008 JP
2010-527324 Aug 2010 JP
2011-524904 Sep 2011 JP
WO 9618418 Jun 1996 WO
WO 9854346 Dec 1998 WO
WO 9855641 Dec 1998 WO
WO 9944423 Sep 1999 WO
WO 0003029 Jan 2000 WO
WO 0022137 Apr 2000 WO
WO 0042208 Jul 2000 WO
WO 0102431 Jan 2001 WO
WO 01004282 Jan 2001 WO
WO 0121217 Mar 2001 WO
WO 0123004 Apr 2001 WO
WO 0190392 Nov 2001 WO
WO 0198513 Dec 2001 WO
WO 0246372 Jun 2002 WO
WO 03064666 Aug 2003 WO
WO 03076605 Sep 2003 WO
WO 03092579 Nov 2003 WO
WO 03104467 Dec 2003 WO
WO 2004018627 Mar 2004 WO
WO 2004031357 Apr 2004 WO
WO 2005001103 Jan 2005 WO
WO 2005023848 Mar 2005 WO
WO 2005030261 Apr 2005 WO
WO 2005065348 Jul 2005 WO
WO 2005075506 Aug 2005 WO
WO 2005107474 Nov 2005 WO
WO 2005113781 Dec 2005 WO
WO 2005117993 Dec 2005 WO
WO 2006086357 Aug 2006 WO
WO 2006119449 Nov 2006 WO
WO 2007124065 Nov 2007 WO
WO 2008095168 Aug 2008 WO
WO 2008150496 Dec 2008 WO
WO 2009065800 May 2009 WO
WO 2010024483 Mar 2010 WO
WO 2010037027 Apr 2010 WO
WO 2011133040 Oct 2011 WO
WO 2012003287 Jan 2012 WO
WO 2012022496 Feb 2012 WO
WO 2012024350 Feb 2012 WO
WO 2012024351 Feb 2012 WO
WO 2012083297 Jun 2012 WO
WO 2013036791 Mar 2013 WO
WO 2013135615 Sep 2013 WO
WO 2013138505 Sep 2013 WO
WO 2014000026 Jan 2014 WO
WO 2014153204 Sep 2014 WO
WO 2014170389 Oct 2014 WO
WO 2015155370 Oct 2015 WO
WO 2016049201 Mar 2016 WO
WO 2017062511 Apr 2017 WO
WO 2017147265 Aug 2017 WO
WO 2017147269 Aug 2017 WO
WO 2018078220 May 2018 WO
WO 2018083257 May 2018 WO
WO 2018083258 May 2018 WO
WO 2018083259 May 2018 WO
WO 2018104919 Jun 2018 WO
WO 2018201017 Nov 2018 WO
WO 2018204677 Nov 2018 WO
WO 2018218083 Nov 2018 WO
WO 2019016756 Jan 2019 WO
WO 2019057745 Mar 2019 WO
WO 2019073059 Apr 2019 WO
WO 2019086450 May 2019 WO
WO 2019086456 May 2019 WO
WO 2019086461 May 2019 WO
WO 2019086466 May 2019 WO
WO 2019158914 Aug 2019 WO
WO 2019179977 Sep 2019 WO
WO 2019179979 Sep 2019 WO
WO 2019191494 Oct 2019 WO
WO 2019199859 Oct 2019 WO
WO 2019202118 Oct 2019 WO
WO 2019239311 Dec 2019 WO
WO 2020014539 Jan 2020 WO
WO 2020046130 Mar 2020 WO
WO 2020076820 Apr 2020 WO
Non-Patent Literature Citations (89)
Entry
Finke et al., “Tracking Fluorescence-Labeled Rabies Virus: Enhanced Green Fluorescent Protein-Tagged Phosphoprotein P Supports Virus Gene Expression and Formation of Infectious Particles,” J. Virol., vol. 78(22): 12333-12343, 2004.
Funston et al., “Expression of Heterologous Genes in Oncolytic Adenoviruses using Picornaviral 2A Sequences that Trigger Ribosome Skipping,” J. Gen. Virol., vol. 89:389-396, 2008.
Ketzer et al., “Synthetic riboswitches for external regulation of genes transferred by replication-deficient and oncolytic adenoviruses,” Nucleic Acids Res 40(21):e167 (10 pages), 2012.
Kim et al., “High Cleavage Efficiency of a 2A Peptide Derived from Porcine Teschovirus-1 in Human Cell Lines, Zebrafish and Mice,” PLoS One, vol. 64:e18556, 2011.
Pelka et al., “Adenovirus E1A Directly Targets the E2F/DP-1 Complex,” J. Virol., vol. 85:8841-8851, 2011.
Szymczak et al., “Correction of Multi-Gene Deficiency in vivo using a Single ‘self-cleaving’ 2A Peptide-Based Retroviral Vector,” Nature Biotech., vol. 22:589-594, 2004.
Tan et al., “Coexpression of double or triple copies of the rabies virus glycoprotein gene using a ‘self-cleaving’ 2A peptide-based replication-defective human adenovirus serotype 5 vector,” Biologicals, vol. 38:586-593, 2010.
Chopra, “Recombinant Adenovirus with Enhanced Green Fluorescent Protein,” Molecular Imaging and Contrast Agent Database (MICAD), Bethesda, MD: National Center for Biotechnology Information (US) (2004-2013): (Dec. 9, 2007, updated Jan. 2, 2008), 5 pp.
Liu et al., “Oncolytic Adenoviral Vector Carrying the Cytosine Deaminase Gene for Melanoma Gene Therapy,” Cancer Gene Ther., vol. 13:845-855, 2006.
Minskaia and Ryan, “Protein Coexpression Using FMDV 2A: Effect of “Linker” Residues,” BioMed Research International, vol. 2013, 12 pp.
Mohr, “To replicate or not to replicate: achieving selective oncolytic virus replication in cancer cells through translational control,” Oncogene, vol. 24:7697-7709, 2005.
Ono et al., “Noninvasive Visualization of Adenovirus Replication with a Fluorescent Reporter in the E3 Region,” Cancer Res., vol. 65:10154-10158, 2005.
Alba et al., “Gutless adenovirus: last-generation adenovirus for gene therapy,” Gene Ther 12:S18-S27, 2005.
Alba et al., “Identification of coagulation factor (F)X binding sites on the adenovirus serotype 5 hexon: effect of mutagenesis on FX interactions and gene transfer,” Blood 114(5): 965-971, 2009.
Alonso et al., “Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo,” Cancer Gene Ther 14:756-761, 2007.
Barton, et al., “Second-Generation Replication-Competent Oncolytic Adenovirus Armed with Improved Suicide Genes and ADP Gene Demonstrates Greater Efficacy without Increased Toxicity”, Molecular Therapy, 2006, 13(2):347-356.
Batzer et al., “Enhanced evolutionary PCR using oligonucleotides with inosine at the 3′-terminus”, Nucleic Acid Research, 1991, 19(18):5081.
Bauerschmitz et al., “Tissue-Specific Promoters Active in CD44+CD24-llow Breast Cancer Cells,” Cancer Res 68(14):5533-5539, 2008.
Bayle et al., “Rapamycin Analogs with Differential Binding Specificity Permit Orthogonal Control of Protein Activity,” Chem Biol 13:99-107, 2006.
Behar et al., “Llama Single-Domain Antibodies Directed against Nonconventional Epitopes of Tumor-Associated Carcinoembryonic Antigen Absent from Nonspecific Cross-Reacting Antigen,” FEBS J., vol. 276:3881-3893, 2009.
Belousova et al., “Modulation of Adenovirus Vector Tropism via Incorporation of Polypeptide Ligands into the Fiber Protein,” J Virol 76(17):8621-8631, 2002.
Berge et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66: 1-19.
Bett et al., “DNA sequence of the deletion/insertion in early region 3 of Ad5 dl309,” Virus Res 39: 75-82, 1995.
Binkowski et al., “Ligand-Regulated Peptides: A General Approach for Modulating Protein-Peptide Interactions with Small Molecules,” Chem. Biol., vol. 12: 847-855, 2005.
Bradshaw et al., “Biodistribution and inflammatory profiles of novel pen ton and hexon double-mutant serotype 5 adenoviruses,” J Control Release 164(3): 394-402, 2012.
Bremnes et al., “The Role of Tumor Stroma in Cancer Progression and Prognosis,” J. Thorac. Oneal., vol. 6:209-217, 2011.
Card et al., “MicroRNA silencing improves the tumor specificity of adenoviral transgene expression,” Cancer Gene Ther 19: 451-459, 2012.
Chen et al., “Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue,” Proc Natl Acad Sci USA 92:4947-4951, 1995.
Cheo et al., “Concerted Assembly and Cloning of Multiple DNA Segments Using In Vitro Site-Specific Recombination: Functional Analysis of Multi-Segment Expression Clones,” Genome Res 14:2111-2120, 2004.
Chong et al., “A System for Small-Molecule Control of Conditionally Replication-Competent Adenoviral Vectors,” Mal Ther 5(2):195-203, 2002.
Doronin et al., “Overexpression of ADP (E3-11.6K) Protein Increases Cell Lysis and Spread of Adenovirus,” Virology 305: 378-387, 2003.
Doronin et al., “Tumor-Specific, Replication-Competent Adenovirus Vectors Overexpressing the Adenovirus Death Protein,” J. Viral., vol. 74:6147-6155, 2000.
Evans et al., “Relocalization of the Mre11-Rad50-Nbs1 Complex by the Adenovirus E4 ORF3 Protein Is Required for Viral Replication”, Journal of Virology, 2005, 79(10):6207-6215.
Extended European Search Report dated Dec. 11, 2013 for European Application No. 11818698.0, 10 pages.
Extended European Search Report for European Application No. 13760821.2, dated Sep. 30, 2015.
Fang et al., “An Antibody Delivery System for Regulated Expression of Therapeutic Levels of Monoclonal Antibodies In Vivo,” Mal. Ther., vol. 15:1153-1159, 2007.
Frese et al., “Selective PDZ protein-dependent stimulation of phosphatidylinositol 3-kinase by the adenovirus E4-ORF1 oncoprotein,” Oncogene 22: 710-721, 2003.
Fuerer et al., “Adenoviruses with Tcf binding sites in multiple early promoters show enhanced selectivity for tumour cells with constitutive activation of the wnt signalling pathway,” Gene Ther 9:270-281, 2002.
Gall et al., “Construction and Characterization of Hexon-Chimeric Adenoviruses: Specification of Adenovirus Serotype,” J Virol 72(12): 10260-10264, 1998.
Gibson et al., “Enzymatic Assembly of DNA Molecules up to Several Hundred Kilobases,” Nature Meth., vol. 6:343-360, 2009.
Glasgow et al., “A Strategy for Adenovirus Vector Targeting with a Secreted Single Chain Antibody,” PLoS One, vol. 4:e8355, 2009.
Havenga et al., “Novel Replication-Incompetent Adenoviral B-group Vectors: High Vector Stability and Yield in PER.C6 Cells,” J. Gen. Viral., vol. 87:2135-2143, 2006.
Hawkins et al., “Gene delivery from the E3 region of replicating human adenovirus: evaluation of the E3B region,” Gene Therapy 8, 1142-1148, 2001.
Heise et al., “An Adenovirus ElA Mutant that Demonstrates Potent and Selective Systemic Anti-Tumoral Efficacy,” Nat Med. 6: 1134-1139, 2000.
Helin et al., “Heterodimerization of the Transcription Factors E2F-1 and DP-1 is required for Binding to the Adenovirus E4 (ORF6/7) Protein,” J Virol 68: 5027-5035, 1994.
Henikoff et al., “Amino acid substitution matrices from protein blocks”, Proc. Natl. Acad. Sci. USA , 1992, 89:10915-10919.
Hernandez-Aya et al. “Targeting the Phosphatidylinositol 3-Kinase Signaling Pathway in Breast Cancer”, The Oncologist, 16, pp. 404-414, 2011.
Holm et al., “Multidrug-resistance Cancer Cells Facilitate El-independent Adenovirus Replication: Impact for Cancer Gene Therapy,” Cancer Res 64:322-328, 2004.
International Preliminary Report on Patentability and Written Opinion dated Feb. 19, 2013 for International Application No. PCT/US2011/048005, 5 pages.
International Search Report dated Mar. 23, 2012 for International Application No. PCT/US2011 /048005, 6 pages.
International Search Report and Written Opinion for PCT/US2019/026626, dated Jun. 24, 2019 (12 pages).
Javier, “Cell polarity proteins: common targets for tumorigenic human viruses,” Oncogene 27:7031-7046, 2008.
Johnson et al., “Selectively replicating adenoviruses targeting deregulated E2F activity are potent, systemic antitumor agents,” Cancer Cell 1:325-337, 2012.
Kirn, “Clinical research results with dl1520 (Onyx-015, a replication-selective adenovirus for the treatment of cancer: what have we learned?”, Gene Therapy, 2001, 8(2):89-98.
Kovesdi et al., “Role of an Adenovirus E2 Promoter Binding Factor in ElA Mediated Coordinate Gene Control,” Proc Nat Acad Sci USA 84: 2180-2184, 1987.
Leicher et al., “Coexpression of the KCNA3B Gene Product with Kv1 .5 Leads to a Novel A-type Potassium Channel*”, The Journal of Biological Chemistry, 1998, 273(52):35095-35101.
Leppard et al., “Adenovirus type 5 E4 Orf3 protein targets promyelocytic leukaemia (PML) protein nuclear domains for disruption via a sequence in PML isoform II that is predicted as protein as a protein interaction site of bioinformatics anaylsis”, Journal of General Virology 2009, 90(1):95-104.
Li et al., “Harnessing homologous recombination in vitro to generate recombinant DNA via SLIC,” Nat Methods 4(3):251-256, 2007.
Lopez et al., “A Tumor-stroma Targeted Oncolytic Adenovirus Replicated in Human Ovary Cancer Samples and Inhibited Growth of Disseminated Solid Tumors in Mice,” Mal Ther 20(12):2222-2233, 2012.
McCormick, “Cancer Gene Therapy: Fringe or Cutting Edge?,” Nature Rev. Cancer, vol. 1:130-141, 2001.
Murakami et al., “Chimeric Adenoviral Vectors Incorporating a Fiber of Human Adenovirus 3 Efficiently Mediate Gene Transfer into Prostrate Cancer Cells,” The Prostate, vol. 70:362-376, 2009.
NCBI Accession No. CVl 10986, Jan. 11, 2011, 3 pages.
Nevels et al., “The Adenovirus E4orf6 can Promote ElA/ElB-induced Focus Formation by Interfering with p53 Tumor Suppressor Function,” Proc. Natl. Acad. Sci. USA, vol. 94:1206-1211, 1997.
Office Action and Search Report from China Application No. 201380014047.7, dated Aug. 5, 2015 (English translation).
O'Shea et al., “Adenoviral Proteins Mimic Nutrient/Growth Signals to Activate the mTOR Pathway for Viral Replication,” EMBO J., vol. 24:1211-1221, 2005.
O'Shea et al., “Adenovirus Overrides Cellular Checkpoints for Protein Translation,” Cell Cycle 4(7):883-888, 2005.
O'Shea et al., “DNA Tumor Viruses—the Spies who Lyse Us,” Curr. Opin. Genet. Dev., vol. 15:18-26, 2005.
O'Shea et al., “Viruses—seeking and destroying the tumor program,” Oncogene 24: 7640-7655, 2005.
Pearson et al., “Improved tools for biological sequence comparison”, Proc. Nat'/. Acad. Sci. USA , 1988, 85:2444-2448.
Rossolini et al., “Use of deoxyinosine-containing primers vs degenerate primers for polymerase chain reaction based on ambiguous sequence information”, Mo/. Cell. Probes, 1994, 8:91-98.
Roy et al., “Rescue of chimeric adenoviral vectors to expand the serotype repertoire,” J Viral Methods 14:41-21, 2007.
Shapiro et al., “Recombinant Adenoviral Vectors Can Induce Expression of p73 via the E4-orf6/7 Protein,” J Viral 80(11):5349-5360, 2006.
Shepard et al., “E4orf13 Is Necessary for Enhanced S-Phase Replication of Cell Cycle-Restricted Subgroup C Adenoviruses,” J Virol 77(15):8593-8595, 2003.
Smith et al., “Comparison of Biosequences”, Advances in Applied Mathematics, 1981, 2:482-489.
Soria et al., “Heterochromatin silencing of p53 target genes by a small viral protein”, Nature, 2010, 466(7310):1076-1083.
Stanton et al. “Re-engineering adenovirus vector systems to enable high-throughput analyses of gene function” Bio Techniques 45: 659-668 (Dec. 2008).
Ullman et al., “Adenovirus E4 ORF3 Protein Inhibits the Interferon-Mediated Antiviral Response”, Journal of Virology, 2007, 81(9):47 44-4752.
Verheije et al., “Retargeting of Viruses to Generate Oncolytic Agents,” Adv. Viral., vol. 2012:1-15, 2012.
Volk et al., “Enhanced Adenovirus Infection of Melanoma Cells by Fiber-Modification,” Cancer Biol Ther 2(5): 511-515, 2003.
Waehler et al., “Engineering targeted viral vectors for gene therapy,” Nat Rev Genet 8(8):573-587, 2007.
Wang et al., “Identification of Specific Adenovirus ElA-N-Terminal Residues Critical to the Binding of Cellular Proteins and to the Control of Cell Growth,” J. Viral., vol. 67:476-488, 1993.
Warram et al., “A Genetic Strategy for Combined Screening and Localized Imaging of Breast Cancer,” Mal Imaging Biol 13:452-461, 2011.
Whyte et al., “Association between an Oncogene and an Anti-Oncogene: the Adenovirus ElA Proteins Bind to the Retinoblastoma Gene Product,” Nature 334:124-129, 1988.
Yaghoubi et al., “Positron Emission Tomography Reporter Genes and Reporter Probes: Gene and Cell Therapy Applications,” Theranostics, vol. 2:374-391, 2012.
Yount et al., “Strategy for Systematic Assembly of Large RNA and DNA Genomes: Transmissible Gastroenteritis Virus Model,” J. Virol., vol. 74: 10600-10611, 2000.
Li, “Self-Cleaving Fusion Tags for Recombinant Protein Production,” Biotechnol. Lett., vol. 33:869-881, 2011.
Shimizu et al., “Development of a Novel Adenovirus Vector Exhibiting MicroRNA-mediated Suppression of the Leaky Expression of Adenovirus Genes,” Yakugaku Zasshi, vol. 132:1407-1412, 2012 (in Japanese) (English abstract).
Suzuki et al., “miR-122A-Regulated Expression of a Suicide Gene Prevents Hepatotoxicity Without Altering Antitumor Effects in Suicide Gene Therapy,” Mol. Ther., vol. 16:1719-1726, 2008.
Wu et al., “The application of intein in the research of membrane protein,” Chemistry of Life 35(2):200-205, 2015.
Related Publications (1)
Number Date Country
20200325492 A1 Oct 2020 US
Provisional Applications (1)
Number Date Country
62298653 Feb 2016 US
Continuations (2)
Number Date Country
Parent 16110207 Aug 2018 US
Child 16916764 US
Parent PCT/US2017/019086 Feb 2017 US
Child 16110207 US