Tumor-targeting synthetic adenoviruses and uses thereof

Information

  • Patent Grant
  • 11813337
  • Patent Number
    11,813,337
  • Date Filed
    Friday, June 7, 2019
    4 years ago
  • Date Issued
    Tuesday, November 14, 2023
    5 months ago
Abstract
Synthetic adenoviruses with liver detargeting mutations and expressing an adenovirus type 34 (Ad34) fiber protein, or a chimeric fiber protein with an Ad34 knob domain, are described. The synthetic adenoviruses traffic to sites of tumors. Use of the synthetic adenoviruses for delivering diagnostic or therapeutic transgenes to tumors are also described.
Description
FIELD

This disclosure concerns synthetic adenoviruses having chimeric fiber proteins and liver detargeting mutations that traffic to sites of tumors. This disclosure further concerns use of the synthetic adenoviruses to express diagnostic or therapeutic transgenes in tumors.


BACKGROUND

Adenovirus (Ad) is a natural multi-gene expression vehicle. Certain coding regions of the virus, such as the E1, E3 and E4 regions, are either not necessary for replication in culture or can be complemented with available cell lines. Therefore, each of these regions can be replaced with non-viral genes to drive the expression of multiple transgenes from a single virus. There are 68 different human adenovirus serotypes, each of which has different properties. Ad5 has been the predominant Ad vector used in basic research, gene therapy and oncolytic virus therapy. However, Ad5 has a limited tropism and only infects epithelial cells that have the coxsackie adenovirus receptor (CAR) receptor for viral uptake. Furthermore, when injected intravenously, Ad5 binds to blood factors that cause it to be sequestered in the liver where it can trigger potentially limiting inflammation and toxicity. Thus, a need remains for modified adenovirus vectors capable of infecting specific cell types following intravenous administration.


SUMMARY

Described herein is the finding that a liver-detargeted synthetic adenovirus expressing a fiber protein with an adenovirus type 34 (Ad34) knob domain is capable of homing to sites of tumors. The synthetic adenoviruses can be used to deliver and express diagnostic or therapeutic transgenes in tumor cells, including tumor stromal cells.


Provided herein is a method of expressing a transgene in tumor cells of a subject. In some embodiments, the method includes administering to the subject a synthetic adenovirus that includes the transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an adenovirus type 5 (Ad5) shaft domain and an Ad34 knob domain. The transgene can be, for example, a diagnostic transgene or a therapeutic transgene.


Also provided herein is a method of diagnosing a subject as having a tumor. In some embodiments, the method includes administering to the subject a synthetic adenovirus that includes a diagnostic transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain. In some examples, the diagnostic transgene is a positron emission tomography (PET) reporter gene. In other examples, the diagnostic transgene encodes a fluorescent protein or an enzyme.


Further provided herein is a method of treating a tumor in a subject. In some embodiments, the method includes administering to the subject a synthetic adenovirus that includes a therapeutic transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain. In some examples, the therapeutic transgene encodes an anti-cancer agent or an agent that disrupts or kills tumor stromal cells.


Synthetic adenovirus genomes having at least 95% sequence identity to SEQ ID NO: 2 or SEQ ID NO: 5 are also provided by the present disclosure.


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


FIGS. 1A-1B. AdSyn-CO176 homes to pancreatic tumors. (FIG. 1A) Cre-LoxP KrasG12D/p53 pancreatic tumor model overview. Mice designated as “Kras; p53/p53” encode the KrasG12D oncogene downstream of the sequence encoding LoxP-stop codon-LoxP. The stop codon blocks the expression of KrasG12D in the absence of Cre recombinase. However, in the presence of Cre recombinase, the stop codon is removed and allows for expression of the KrasG12D oncogene. In these same mice, both alleles of the p53 gene are flanked by LoxP sites (LoxP-p53-LoxP). Mice designated “p53/p53; Cre” have both alleles of the p53 gene flanked by LoxP sites (LoxP-p53-LoxP) and also express the Cre recombinase transgene driven by the pancreatic and duodenal homeobox 1 (Pdx1) promoter. Pdx1 is a gene that is expressed specifically in the pancreatic cells, and thus both copies of p53 are deleted in the pancreatic cells. Breeding between the strains gives rise to offspring in which the Pdx1 promoter-driven Cre mediates the deletion of both alleles of the tumor suppressor p53 and activation of the mutant KrasG12D in pancreatic cells. Homozygous mice designated “Kras; p53/p53; Cre” develop pancreatic tumors in 5-7 weeks. (FIG. 1B) AdSyn-CO176, a synthetic virus with a chimeric fiber protein comprising the Ad34 knob domain, was injected intravenously into Kras; p53/p53 and Kras; p53/p53; Cre mice. Seventy-two hours after the injection of virus, tissues were collected, incubated for 5 minutes with luciferin, and then scanned for 5 minutes using the IVIS™ imaging system. The Kras; p53/p53 mouse had a normal pancreas, and the luciferase signal was mainly from the spleen. The Kras; p53/p53; Cre mouse had pancreatic tumors, and the signal was mainly from the pancreatic tumor.



FIGS. 2A-2B. AdSyn-CO176 infected pancreatic tumor after the tail vein injection in a Cre-mediated genetic manipulation heterozygous model. (FIG. 2A) Kras; p53/p53 mice are as described for FIG. 1A. Mice designated as “p53/+; Cre” mice have one wild type p53 allele and one p53 allele flanked by LoxP sites (LoxP-p53-LoxP). Breeding between these two strains gives rise to offspring in which the Pdx1 promoter-driven Cre recombinase mediates the deletion of a single allele of the tumor suppressor p53 and activation of the mutant KrasG12D in pancreatic cells. These heterozygous mice, designated “Kras; p53/+; Cre,” develop tumors later in life (at 4-9 months of age) due to the fact that they have one wild type allele of p53. This wild type allele must be lost or mutated in order for pancreatic tumors to develop. (FIG. 2B) AdSyn-CO176 was injected intravenously into p53/+; Cre and Kras; p53/+; Cre mice (4 months old). Seventy-two hours after the injection of virus, tissues were collected, incubated with luciferin for 5 minutes, and then scanned for 1 minute using the IVIS™ imaging system. The p53/+; Cre mouse had a normal pancreas, and the signal was mainly from spleen. The Kras; p53/+; Cre mouse at 4 months of age had a pancreatic tumor, and the signal was mainly from the tumor and liver.



FIGS. 3A-3C. AdSyn-CO176 can infect and diagnose a pancreatic tumor at an early stage after tail vein injection. The heterozygous Kras; p53/+; Cre mice develop pancreatic tumors in 4-9 months. To test whether AdSyn-CO176 can infect pancreatic tumors at a very early stage of tumor development (before tumors are visible), AdSyn-176 was injected into Kras; p53/+; Cre mice at 2 months of age and luciferase expression was measured. (FIG. 3A) AdSyn-CO176 was injected intravenously into Kras; p53/+; Cre mice at 2 months of age. Seventy-two hours after the injection of virus, tissues were collected, incubated with luciferin for 5 minutes, and scanned for 4 minutes using the IVIS™ imaging system. The pancreas of Kras; p53/+; Cre mouse at 2 months old looked normal but luciferase signal was found in this tissue. (FIG. 3B) H&E staining showing the typical histology of normal pancreas (a) and pancreatic tumor (b). (FIG. 3C) H&E staining of the pancreas from a Kras; p53/+; Cre mouse at 2 months of age showing that a small part of the pancreas was developing the tumor (as shown in the polygon). Most of the pancreas appeared normal. This result indicates that AdSyn-CO176 can infect pancreatic tumors at a very early stage.



FIGS. 4A-4B. AdSyn-CO176 infects stromal cells in the pancreatic tumor. In pancreatic tumors, only 10% of the cells are cancer cells; the remaining 90% are stroma cells. As determined by immunohistochemistry (IHC) and immunofluorescence (IF) staining, the cells that were infected by AdSyn-CO176 were stromal cells. (FIG. 4A) IHC staining of a pancreatic tumor infected with AdSyn-CO176. CK19 is a marker of tumor cells while smooth muscle actin (SMA) is a marker of stromal cells. The staining of GFP, which was expressed from AdSyn-CO176 overlapped with SMA staining, indicating that AdSyn-CO176 targets stromal cells. (FIG. 4B) IF staining of the pancreatic tumor infected by AdSyn-CO176. GFP staining overlapped with SMA staining, confirming AdSyn-CO176 infection of stromal cells.



FIGS. 5A-5D. AdSyn-CO176 infected glioblastoma after tail vein injection. Synthetic adenovirus AdSyn-CO176 was injected into mice with glioblastoma by tail vein and the luciferase signal was found in the glioblastoma. (FIG. 5A) Schematic of a Cre-mediated genetic manipulation glioblastoma model. Lentiviruses were injected directly into the brain of GFAP-Cre mice. Glial fibrillary acidic protein (GFAP) promoter-driven Cre recombinase cleaves out RFP from the lentivirus-encoded gene and induces the expression of HRasV12 and GFP primarily in astrocytes. Expression of lentivirus-encoded U6-p53 shRNA knocks down the expression of p53 in the brain cells that take up the virus. The expression of HRasV12 and the knock down of p53 induces tumorigenesis in the brain from 1 week after the injection. GFP signal is used to indicate the formation of glioblastoma. (FIG. 5B) Saline, AdSyn-CO171, or AdSyn-CO176 were injected via intravenous (IV) administration into GFAP-Cre mice that had received the tumor-inducing lentiviruses 4 weeks earlier. Forty-eight hours after the injection of virus, mice were scanned for 1 minute using the IVIS™ imaging system 5 minutes after the intraperitoneal injection of luciferin. The luciferase signal was detected in AdSyn-CO176-infected mice (arrow), while no signal was detected in saline-treated or AdSyn-CO171-injected mice. (FIG. 5C) Wild type mice (normal brain) and GFAP-Cre mice with the injection of lentiviruses (develop brain tumors) were injected with AdSyn-CO171 or AdSyn-CO176. Brain tissues were collected 72 hours after the injection of synthetic adenoviruses, incubated with luciferin for 5 minutes, and scanned for 5 minutes using the IVIS™ imaging system. Only the GFAP-Cre mouse injected with the tumor-inducing lentiviruses showed a luciferase signal from AdSyn-CO176. This demonstrates that AdSyn-CO176 will traffic to the brain tissue only when a tumor is present. (FIG. 5D) Brain tissues were also scanned for the GFP signal. The GFP signal is used to identify the glioblastoma. Both of the GFAP-Cre mice that received the tumor-inducing lentiviruses had the GFP signal in the brain, while no GFP was detected in mice that did not receive lentivirus. The GFP signal overlapped with the luciferase signal perfectly in the GFAP-Cre mouse that received the tumor-inducing lentiviruses and AdSyn-CO176.



FIGS. 6A-6B. Trafficking of AdSyn-CO176 to glioblastoma is driven by the tumor, not injury. The injection of the tumor-inducing lentiviruses causes temporary injury to the brain at the injection site. Although the synthetic adenoviruses (AdSyn-CO171 and AdSyn-CO176) were injected 4 weeks after the initial injection of lentiviruses, it was still unclear whether trafficking of AdSyn-CO176 to the glioblastoma was driven by the tumor or by the injection site injury. To answer this question, GFAP-Cre mice were injected with synthetic adenovirus 4 weeks after either no injection, sham-injection or injection with tumor-inducing lentivirus. (FIG. 6A) GFAP-Cre mice were injected with either Hanks' balanced salt solution (HBSS) or tumor-inducing lentiviruses. After 4 weeks, AdSyn-CO171 was injected intravenously. There was no luciferase signal from AdSyn-CO171 in the brain in either group of mice. (FIG. 6B) GFAP-Cre mice were injected with HBSS or tumor-inducing lentiviruses, or received no injection. After 4 weeks, mice were injected intravenously with AdSyn-CO176. The luciferase signal was detected only in the brain of the mouse that received the tumor-inducing lentiviruses, while the mouse that received HBSS or no injection produced no signal. These results demonstrate that the specificity of AdSyn-CO176 is driven by the tumor and not the injection site injury.



FIGS. 7A-7B. AdSyn-CO176 can traffic to human glioblastoma xenograft tumors. (FIG. 7A) Schematic of the human glioblastoma xenograft model. Human glioblastoma U87 cells that expresses the tdTomato fluorescent protein (U87-tdTomato) were injected intracranially into NOD scid gamma (NSG) mice to generate glioblastoma tumors. (FIG. 7B) AdSyn-CO171 and AdSyn-CO176 were injected intravenously into NSG mice by tail vein injection 4 weeks after they received the intracranial injection of U87-tdTomato. Forty-eight hours after the injection of viruses, the tissues indicated in the panel were collected, incubated with luciferin for five minutes and then scanned for 1 minute using the IVIS™ imaging system. Only the AdSyn-CO176 injected mice showed luciferase signal in the brain, and this signal completely overlapped with tdTomato expression.



FIGS. 8A-8C. Administration of a synthetic adenovirus with a therapeutic transgene. (FIG. 8A) Schematic of the KPCL (KrasG12D; p53 knockout; Pdx1-Cre; firefly Luciferase) mouse model. KPCL mice are similar to homozygous “Kras; p53/p53; Cre” mice, which specifically express KrasG12D in the pancreas and have the p53 gene knocked out only in the pancreas. However, KPCL mice also specifically express firefly luciferase in the pancreas. The development of tumors in KPCL mice is also similar to the “Kras; p53/p53; Cre” mice. (FIG. 8B) Table showing average survival of KPCL mice for each treatment (at least 4 mice per treatment group). AdSyn-CO987 is a synthetic adenovirus based upon AdSyn-CO176. The herpes simplex virus-1 thymidine kinase (TK)/ganciclovir (GCV) suicide gene was cloned into AdSyn-CO176 to replace the firefly luciferase/GFP gene. A Renilla luciferase was also inserted just after TK in the genome of AdSyn-CO176. Control virus AdSyn-CO989 was generated by cloning TK-P2A-renilla luciferase into AdSyn-CO171 to replace the original firefly luciferase/GFP gene. KPCL mice were injected intravenously via the tail vein with 1×106 plaque forming units (PFU) of the indicated viruses at 5-6 weeks of age. Two days later, the mice were injected intraperitoneally (i.p.) or intravenously (i.v.) with GCV. Three control groups were used: AdSyn-CO989+GCV; AdSyn-CO987 followed with saline injection (AdSyn-CO987+saline); and GCV injection only (i.p. or i.v.). Treatment with AdSyn-CO987+GCV extended mouse survival compared with controls. (FIG. 8C) Images of representative mice showing firefly luciferase signal. The firefly luciferase signal (expressed by tumors) was analyzed during treatment to monitor tumor growth. The treatment for mouse Z619R was AdSyn-CO987+saline, which served as the control. Mice Z601R and Z607R were treated with AdSyn-CO987+GCV (i.p.). While the strength of the firefly luciferase signal increased in the control mouse Z619R (indicating an increase in tumor size), the signal decreased in mice Z601R and Z607R (indicating a reduction in tumor size).



FIGS. 9A-9B. Histology of tumors in mice treated with AdSyn-CO987. (FIG. 9A) Images of H&E staining of pancreatic tumors. Mice Z655, 1806, Z619 and Z621 were all control mice. Mouse Z655 was treated with i.p. injected GCV only; mouse 1806 was treated with i.v. injected GCV only; mouse Z619 was treated with AdSyn-CO987+saline; and mouse Z621 received no treatment. Mouse Z656 received treatment with AdSyn-CO987+GCV i.v. Compared to the controls, the tumor from Z656 had more regions of necrosis (as indicated by the arrowheads). (FIG. 9B) Representative regions of necrosis in the tumor of Z656 with magnification. The regions are also indicated in FIG. 9A.





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. 3, 2019, 215 KB, which is incorporated by reference herein. In the accompanying sequence listing:

    • SEQ ID NO: 1 is the nucleotide sequence of synthetic adenovirus AdSyn-CO171.
    • SEQ ID NO: 2 is the nucleotide sequence of synthetic adenovirus AdSyn-CO176.
    • SEQ ID NO: 3 is the amino acid sequence of Ad5 hexon.
    • SEQ ID NO: 4 is the amino acid sequence of Ad5 hexon E451Q.
    • SEQ ID NO: 5 is the nucleotide sequence of synthetic adenovirus AdSyn-CO987.
    • SEQ ID NO: 6 is the nucleotide sequence of synthetic adenovirus AdSyn-CO989.


DETAILED DESCRIPTION
I. Abbreviations





    • Ad adenovirus

    • CAR coxsackie adenovirus receptor

    • CEA carcinoembryonic antigen

    • EGF epidermal growth factor

    • EGFR epidermal growth factor receptor

    • FLT3 Fms-related tyrosine kinase 3

    • GCV ganciclovir

    • GFAP glial fibrillary acidic protein

    • GFP green fluorescent protein

    • GM-CSF granulocyte macrophage colony stimulating factor

    • H&E hematoxylin and eosin

    • HSV herpes simplex virus

    • ICAM intercellular adhesion molecule

    • IF immunofluorescence

    • IHC immunohistochemistry

    • IL interleukin

    • IRES internal ribosomal entry site

    • i.p. intraperitoneal

    • i.v. intravenous

    • KPCL KrasG12D; p53 knockout; Pdx1-Cre; firefly Luciferase

    • LFA lymphocyte function-associated antigen

    • miR microRNA

    • MUC1 mucin 1

    • NOD non-obese diabetic

    • NSG NOD scid gamma

    • PD-1 programmed cell death protein 1

    • PDGF platelet derived growth factor

    • PET positron emission tomography

    • PFU plaque forming unit

    • shRNA short hairpin RNA

    • SMA smooth muscle actin

    • TGF transforming growth factor

    • TK thymidine kinase

    • TNF tumor necrosis factor

    • UTR untranslated region

    • VEGF vascular endothelial growth factor

    • WT wild-type





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:


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).


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.


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.


Detargeted: In the context of the present disclosure, a “detargeted” adenovirus is a recombinant or synthetic adenovirus comprising one or more modifications that alter tropism of the virus such that is no longer infects, or no longer substantially infects, a particular cell or tissue type. In some embodiments, the recombinant or synthetic adenovirus comprises a capsid mutation, such as a mutation in the hexon protein (for example, E451Q). In some embodiments, the recombinant or synthetic adenovirus comprises a native capsid from an adenovirus that naturally does not infect, or does not substantially infect, a particular cell or tissue type. In some embodiments herein, the recombinant or synthetic adenovirus is liver detargeted and/or spleen detargeted.


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.


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.


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.


Glioblastoma: A fast-growing type of central nervous system tumor that forms from glial tissue of the brain and spinal cord. Glioblastoma usually occurs in adults and affects the brain more often than the spinal cord. Glioblastoma is the most common and most aggressive cancer that initiates in the brain. Glioblastoma is also known as glioblastoma multiforme (GBM) and grade IV astrocytoma.


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


Hexon: A major adenovirus capsid protein. An exemplary hexon sequence from Ad5 is set forth herein as SEQ ID NO: 3. A mutant hexon sequence comprising an E451Q substitution is set forth herein as SEQ ID NO: 4.


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.


MicroRNA (miRNA or miR): A single-stranded RNA molecule that regulates gene expression in plants, animals and viruses. A gene encoding a microRNA is transcribed to form a primary transcript microRNA (pri-miRNA), which is processed to form a short stem-loop molecule, termed a precursor microRNA (pre-miRNA), followed by endonucleolytic cleavage to form the mature microRNA. Mature microRNAs are approximately 21-23 nucleotides in length and are partially complementary to the 3′UTR of one or more target messenger RNAs (mRNAs). MicroRNAs modulate gene expression by promoting cleavage of target mRNAs or by blocking translation of the cellular transcript. In the context of the present disclosure, a “liver-specific microRNA” is a microRNA that is preferentially expressed in the liver, such as a microRNA that is expressed only in the liver, or a microRNA that is expressed significantly more in the liver as compared to other organs or tissue types. In some embodiments, the microRNA is miR-122. In the context of the present disclosure, a “spleen-specific microRNA” is a microRNA that is preferentially expressed in the spleen, such as a microRNA that is expressed only in the spleen, or a microRNA that is expressed significantly more in the spleen as compared to other organs or tissue types. In some embodiments, the microRNA is miR-142-3p.


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.


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.


Pancreatic cancer: Cancer that begins in the tissues of the pancreas. Pancreatic cancer typically spreads rapidly and is seldom detected at early stages, leading to a poor prognosis for most diagnosed patients. The most common type of pancreatic cancer is pancreatic adenocarcinoma, which accounts for approximately 85% of pancreatic cancer cases.


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 synthetic 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 histadyl, 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.


Positron emission tomography (PET): An imaging technique that is used to observe metabolic processes in the body. PET detects pairs of gamma rays emitted indirectly by a positron-emitting radionuclide, which is introduced into the body on a biologically active molecule. PET reporter genes encode molecules (such as receptors or enzymes) that provide a target for PET probes, which can then be detected by imaging. PET reporter genes are generally classified into three different groups: (1) reporter genes encoding enzymes that phosphorylate specific PET reporter probes, leading to their intracellular entrapment; (2) reporter genes encoding protein receptors that can be bound by specific PET reporter probes; and (3) reporter genes encoding protein transporters that transport a radionuclide reporter probe into cells expressing the reporter gene (Yaghoubi et al., Theranostics 2(4):374-391, 2012). As used herein, a “PET reporter gene” includes any gene that encodes a protein capable of interacting with a PET reporter probe in a manner allowing for detection of the probe by molecular imaging. Exemplary PET reporter genes include, but are not limited to, herpes simplex virus (HSV) thymidine kinase (TK) and mutant forms thereof, varicella zoster virus (VSV) TK, human mitochondrial TK and mutants thereof, mutants of deoxycytidine kinase, dopamine 2 receptors mutants, human estrogen receptor α ligand binding domain (hERL), human somatostain receptor subtype 2 (hSSTr2), recombinant human carcinoembryonic antigen (CEA), engineered antibody fragments, humanized membrane anchored anti-polyethylene glycol (PEG), sodium iodide symporter (NIS), and human norepinephrine transporter (hNET) (see Yaghoubi et al. (2012) for a review of PET reporter genes and corresponding reporter probes).


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). A “tissue-specific promoter” is a promoter that is substantially active only in a particular tissue or tissues.


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.


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.


Stroma: The supportive tissues of an epithelial organ or tumor, consisting of connective tissues and blood vessels. Stromal cells are the cells that make up the stroma, primarily fibroblasts and pericytes. Tumor stroma is predominantly made up of fibroblasts, extracellular matrix, immune cells, vasculature and basement membrane (Bremnes et al., J Thorac Oncol 6:209-217, 2011). Tumor stromal cells are known to play a significant role in cancer growth and progression.


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: A chemical compound, small molecule, recombinant virus or other composition, such as an antisense compound, antibody, peptide or nucleic acid molecule capable of inducing a desired therapeutic or prophylactic effect when properly administered to a subject.


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.


Transgene: A gene that has been inserted into the genome of a different organism (such as a virus). Transgenes can also be referred to as heterologous genes. As used herein, a “diagnostic transgene” refers to any transgene encoding a detectable product, such as, but not limited to, a fluorescent protein, an enzyme or a PET reporter. As used herein a “therapeutic transgene” refers to any transgene encoding product with a therapeutic application. In the context of the present disclosure, a therapeutic transgene can be, for example, an anti-cancer agent or an agent that disrupts or kills cells of the tumor stroma.


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


Vector: A nucleic acid molecule allowing insertion of foreign nucleic acid without disrupting the ability of the vector to replicate and/or integrate in a host cell. A vector can include nucleic acid sequences that permit it to replicate in a host cell, such as an origin of replication. A vector can also include one or more selectable marker genes and other genetic elements. An expression vector is a vector that contains the necessary regulatory sequences to allow transcription and translation of inserted gene or genes.


III. Overview of Several Embodiments

It is disclosed herein that a liver-detargeted synthetic adenovirus expressing a fiber protein with an Ad34 knob domain is capable of homing to sites of tumors. The synthetic adenoviruses can be used, for example, to deliver and express diagnostic or therapeutic transgenes in tumor cells, including tumor stromal cells.


Provided herein is a method of expressing a transgene in tumor cells of a subject. In some embodiments, the method includes administering to the subject a synthetic adenovirus that includes the transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain.


In some embodiments, the transgene is a diagnostic transgene. In some examples, the diagnostic transgene encodes a fluorescent protein, such as, but not limited to a green fluorescent protein (GFP), a yellow fluorescent protein (YFP), a cyan fluorescent protein (CFP), a red fluorescent protein (RFP), a blue fluorescent protein (BFP), or an orange fluorescent protein (for example, mOrange). In other examples, the diagnostic transgene encodes an enzyme, such as a luciferase. In yet other examples, the diagnostic transgene comprises a PET reporter gene.


In other embodiments, the transgene is a therapeutic transgene. In some examples, the therapeutic transgene encodes an anti-cancer agent. In specific examples, the anti-cancer agent is a pro-inflammatory molecule or cytokine, such as granulocyte macrophage colony stimulating factor (GM-CSF), CD40 ligand (CD40L), Fms-related tyrosine kinase 3 (FLT3) ligand, interleukin (IL)-1b, IL-2, IL-4, IL-6, IL-12, tumor necrosis factor (TNF)-α, an interferon, a chemokine, B7-1, intercellular adhesion molecule (ICAM)-1, lymphocyte function-associated antigen (LFA)-3, transforming growth factor (TGF)-β, platelet derived growth factor (PDGF) or epidermal growth factor (EGF). In other specific examples, the anti-cancer agent is an anti-angiogenic factor, such as an inhibitor of vascular endothelial growth factor (VEGF). In other specific examples, the anti-cancer agent is an inhibitor (such as a siRNA or shRNA inhibitor) of KRas. In other specific examples, the anti-cancer agent is an inhibitor of cytotoxic T lymphocyte-associated molecule (CTLA)-4, programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), carcinoembryonic antigen (CEA) or mucin 1 (MUC1). In some examples, the therapeutic transgene encodes an agent that disrupts or kills tumor stromal cells. In specific examples, the agent is Rexin-G, herpes simplex virus (HSV) thymidine kinase (TK), p53, TNF-α, Fas/FasL, or diphtheria toxin A.


Also provided herein is a method of diagnosing a subject as having a tumor. In some embodiments, the method includes administering to the subject a synthetic adenovirus that includes a diagnostic transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain.


In some embodiments, the diagnostic transgene is a PET reporter gene. In some examples, the PET reporter gene is a viral or human thymidine kinase (or mutant form thereof), a mutant of deoxycytidine kinase, a dopamine 2 receptor mutant, a human estrogen receptor α ligand binding domain (hERL), a human somatostain receptor subtype 2 (hSSTr2), a recombinant human CEA, an engineered antibody fragment, a humanized membrane anchored anti-polyethylene glycol (PEG), a sodium iodide symporter (NIS), or a human norepinephrine transporter (hNET).


In other embodiments, the diagnostic transgene encodes a fluorescent protein. In some examples, the fluorescent protein comprises a GFP, YFP, CFP, RFP, BFP, or orange fluorescent protein.


In other embodiments, the diagnostic transgene encodes an enzyme. In one example, the enzyme is a luciferase.


Further provided herein is a method of treating a tumor in a subject. In some embodiments, the method includes administering to the subject a synthetic adenovirus that includes a therapeutic transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain.


In some embodiments, the therapeutic transgene encodes an anti-cancer agent. In some examples, the anti-cancer agent is a pro-inflammatory molecule or cytokine, such as GM-CSF, CD40L, FLT3, IL-1b, IL-2, IL-4, IL-6, IL-12, TNF-α, an interferon, a chemokine, B7-1, ICAM-1, LFA-3, TGF-β, PDGF or EGF. In other examples, the anti-cancer agent is an anti-angiogenic factor, such as an inhibitor of VEGF. In other examples, the anti-cancer agent is an inhibitor (such as a siRNA or shRNA inhibitor) of KRas. In other examples, the anti-cancer agent is an inhibitor of CTLA-4, PD-1, CEA or MUC1. In other embodiments, the therapeutic transgene encodes an agent that disrupts or kills tumor stromal cells. In some examples, the agent is Rexin-G, HSV-TK, p53, TNF-α, Fas/FasL, or diphtheria toxin A.


In some embodiments of the methods disclosed herein, the synthetic adenovirus includes a modified capsid that detargets the virus from the liver. In some examples, the synthetic adenovirus comprises a modified hexon protein, such as an E451Q mutation (set forth herein as SEQ ID NO: 4). In other embodiments, the synthetic adenovirus has a native (unmodified) capsid that detargets the synthetic adenovirus from the liver (for example, a capsid from an adenovirus serotype that naturally does not infect the liver).


In some embodiments of the methods disclosed herein, the synthetic adenovirus further includes one or more binding sites, such as two or three binding sites, for a liver-specific microRNA. In some examples, the liver-specific microRNA is miR-122. In some examples, the one or more binding sites are in the 3′UTR of the transgene.


In some embodiments of the methods disclosed herein, the synthetic adenovirus further includes one or more binding sites, such as two or three binding sites, for a spleen-specific microRNA. In some examples, the spleen-specific microRNA is miR142-3p. In some examples, the one or more binding sites are in the 3′UTR of the transgene.


In some embodiments of the methods disclosed herein, the transgene is regulated by a tissue-specific promoter, such as a promoter active in the pancreas or the cells of the central nervous system. In other embodiments, the transgene is regulated by a tumor-specific promoter.


In some embodiments of the methods disclosed herein, the synthetic adenovirus is generated from an Ad5 vector genome. In some examples, the synthetic adenovirus comprises Ad5 capsid proteins and a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain.


In some embodiments of the methods disclosed herein, the tumor is a pancreatic tumor. In other embodiments, the tumor is a glioblastoma. In other embodiments, the tumor is a breast cancer, prostate cancer, gastrointestinal cancer, bone cancer or melanoma tumor.


In some embodiments of the methods disclosed herein, the tumor is characterized by a loss of p53 tumor suppressor activity. In some examples, the tumor exhibits mutations in p53. In some examples, the tumor exhibits loss of a wild-type p53 allele.


In some embodiments of the methods disclosed herein, the tumor is characterized by mutations in a Ras gene, such as KRas, HRas or NRas. In some embodiments of the methods disclosed herein, the tumor is characterized by alterations or mutations in neurofibromatosis type 1 (NF1), epidermal growth factor receptor (EGFR), BRCA1, BRCA2 or HER2.


In some embodiments of the methods disclosed herein, the genome of the synthetic adenovirus comprises a nucleotide sequence 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 SEQ ID NO: 2. In some examples, the genome of the synthetic adenovirus comprises or consists of the nucleotide sequence of SEQ ID NO: 2.


In other embodiments of the methods disclosed herein, the genome of the synthetic adenovirus comprises a nucleotide sequence 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 SEQ ID NO: 5. In some examples, the genome of the synthetic adenovirus comprises or consists of the nucleotide sequence of SEQ ID NO: 5.


Further provided herein are synthetic adenovirus genomes having 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 SEQ ID NO: 2 or SEQ ID NO: 5. In some examples, the genome of the synthetic adenovirus comprises or consists of the nucleotide sequence of SEQ ID NO: 2 or SEQ ID NO: 5.


IV. Synthetic Adenoviruses

The Adsembly, AdSLICr and RapAD technologies enable the modular design and production of adenoviruses with unique capabilities (see PCT Publication Nos. WO2012/024351 and WO2013/138505, which are herein incorporated by reference in their entirety). The ability to design custom viruses with novel functions and properties opens up the potential to expand the utility of adenovirus as a vehicle to deliver therapeutic proteins by persuading the host to produce proteins in situ. This provides the unique capability to use human proteins that are difficult to manufacture for therapeutic purposes, and enable flexible delivery of almost any protein to diseased tissues.


The specific modifications disclosed herein are described with reference to the adenovirus 5 (Ad5) genome sequence, but may be used with any adenovirus serotype. Adenovirus is a natural multi-gene expression vehicle. The E1, E3, and E4 regions are either not necessary for replication in culture or can be complemented with available cell lines. Each of these regions has independent promoter elements that can be replaced with cellular promoters if necessary to drive the expression of multiple gene products via alternative splicing.


As disclosed herein, to create Ad5 expression vectors for in vivo use and transgene delivery, the E1A/E1B genes were deleted and replaced with at least one transgene. In some embodiments, the transgene is an EF1α driven luciferase-GFP fusion.


The synthetic adenoviruses disclosed herein may further include modifications that detarget the virus from the liver. Ad5 hexon can bind to Factor X in the blood, which can lead to its absorption by Kuppfer cells in the liver that prevent systemic dissemination and limiting inflammation. To overcome this, synthetic adenoviruses were engineered to include additional genomic modifications in the E1 and core modules that prevent adenovirus uptake and transgene expression in the liver, as described further below.


A. Ad34 Fiber and Chimeric Fiber Proteins for Retargeting


While the fiber proteins of Ad5 and many other serotypes have been shown to bind to the coxsackie adenovirus receptor (CAR) for cellular attachment, other serotypes have been shown to use CD46 (Gaggar et al., Nat Med 9:1408-1412, 2003), desmoglein 2 (Wang et al., Nat Med 17:96-104, 2011), sialic acid (Nilsson et al., Nat Med 17:105-109, 2011), or others (Arnberg, Trends Pharmacol Sci 33:442-448, 2012). The receptor usage of many serotypes has not been thoroughly examined and CD46 is not thought to be expressed in mature mice. Since the globular knob at the C-terminus of the fiber protein is typically responsible for receptor binding, a chimera was created by replacing the Ad5 fiber knob with that from Ad34 (see Example 1 below). The synthetic virus included an E1 module containing an E1A/E1B deletion and a luciferase-GFP fusion driven by an EF1α promoter. The synthetic adenovirus also included a liver detargeting modification in the hexon protein (E451Q) and binding sites in the 3′UTR of the transgene for a microRNA that is specifically expressed in the liver (miR-122) to prevent off-target expression of the transgene.


The data disclosed herein demonstrate the ability to combine modified parts from other serotypes in order to improve Ad5-based vectors. In this case allowing for rapid assembly of viruses that are optimized for entry into tumor cells.


B. Liver Detargeting Modifications


Natural Ad5 vectors will only infect the lungs (via inhalation) or liver (via intravenous administration). Ad5 hexon binds to Factor X in the blood, which leads its absorption by Kuppfer cells in the liver, preventing systemic dissemination and inducing virus-limiting inflammation. To overcome this and enable intravenous delivery of viruses that could travel to sites of tumors systemically, synthetic adenoviruses were engineered to include additional genomic modifications in the E1 and core modules that prevent uptake and expression in the liver.


To prevent virus uptake and sequestration in the liver through Ad5 hexon binding to Factor X, viruses were engineered with an additional mutation in hexon (E451Q) that prevents liver uptake. For example, AdSyn-CO171 does not accumulate in the liver and instead is able to target other organs, such as the spleen and lymph nodes. Thus, in some embodiments herein, the synthetic adenovirus comprises a modified hexon protein with an E451Q substitution.


To prevent off-target transgene expression in the liver, viruses were engineered to include binding sites in the 3′ untranslated region (UTR) of the transgene for microRNAs that are specifically expressed in the liver. In particular embodiments, miR122 was selected as the liver-specific microRNA as its expression and binding sites are conserved in both human and mouse liver cells. In some examples, two micro-RNA binding sites for liver-specific miR122 were inserted in the 3′UTR of the transgene to prevent any residual transgene expression in the liver.


It is disclosed herein that a synthetic adenovirus with miR-122 binding sites and hexon mutation does not accumulate in the liver and instead is able to target tumors. In some embodiments, the one or more binding sites for the liver-specific microRNA are located in the 3′-UTR of the transgene. In some examples, the liver-specific microRNA is miR-122, miR-30 or miR-192.


Other mutations to the adenovirus hexon gene are contemplated herein to prevent adenovirus accumulation in the liver. For example, a synthetic adenovirus could be detargeted from the liver by replacing the nine hypervariable regions of hexon with those from different serotypes.


In some examples, the recombinant adenovirus comprises a hexon protein comprising or consisting of the amino acid sequence of SEQ ID NO: 3 or SEQ ID NO: 4.


C. Capsid Swaps for Evading Neutralizing Antibodies


The majority of the human population already has antibodies that recognize Ad5, the serotype most frequently used in research and therapeutic applications. Moreover, once a particular adenovirus serotype is used in a patient, new antibodies that recognize the viral capsid will be generated, making repeated administration of the same vector problematic. Therefore, the present disclosure further contemplates exploiting natural adenovirus modularity to create chimeric viruses capable of evading existing neutralizing antibodies. For example, the recombinant adenoviruses disclosed herein may further have complete ‘capsid’ module swaps (almost 60% of genome), which render them ‘invisible’ to pre-existing antibodies and enables repeated inoculations.


In some embodiments, the E1, E3 and E4 regions of the genome are derived from a first adenovirus serotype and the E2B, L1, L2, L3, E2A and L4 regions of the genome are derived from a second adenovirus serotype, such as Ad34. In some examples, the E1 region of the first adenovirus serotype is modified to encode a pIX protein from the second adenovirus serotype; and/or the E3 region of the first adenovirus serotype is modified to encode Uexon and fiber proteins from the second adenovirus serotype. In particular examples, the first adenovirus serotype is Ad5 and the second adenovirus serotype is Ad34.


D. Expression of Transgenes for Diagnostic and Therapeutic Applications


It is disclosed herein that recombinant adenoviruses comprising a chimeric fiber protein having an Ad34 knob domain and liver detargeting mutations are capable of targeting tumors. It is further disclosed that the recombinant adenoviruses are capable of expressing transgenes in tumor tissue, such as in tumor stromal cells. In one example, the transgene is a reporter, such as a luciferase-GFP reporter that enables detection of virus-transduced cells. In another example, the transgene is a therapeutic transgene, such as an anti-cancer molecule. The present disclosure provides synthetic adenoviruses that include diagnostic or therapeutic transgenes for the diagnosis and treatment of tumors.


Provided herein is a method of diagnosing a subject as having a tumor by administering to the subject a synthetic adenovirus that includes a diagnostic transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain. The diagnostic transgene can be, for example, a PET reporter gene, a fluorescent protein or an enzyme.


Also provided herein is a method of treating a tumor in a subject by administering to the subject a synthetic adenovirus that includes a therapeutic transgene, a native or modified capsid that detargets the synthetic adenovirus from the liver, and an Ad34 fiber protein or a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain. The therapeutic transgene can encode, for example, an anti-cancer agent or an agent that disrupts or kills tumor stromal cells.


In some embodiments, the transgene is inserted into the E1 or E3 region. Appropriate transgene insertion sites are well known in the art (see, for example, PCT Publication No. WO2012/024351).


The transgene, such as a gene encoding a fluorescent protein, is operably linked to a promoter. In some embodiments, the promoter is a heterologous promoter. In some examples, the promoter is the EF1α promoter. The selection of promoter is within the capabilities of one of skill in the art. In some cases, the promoter is an inducible promoter or a tissue-specific promoter. An exemplary tissue-specific promoter for expression in pancreatic tissue is Pdx1.


In some cases a single promoter is used to regulate expression of multiple genes, which can be achieved by use of an internal ribosomal entry site (IRES) or 2A peptide.


V. Pharmaceutical Compositions and Administration Thereof

Provided herein are compositions comprising a synthetic adenovirus (or one or more nucleic acids or vectors encoding the recombinant adenovirus). The compositions are, optionally, suitable for formulation and administration in vitro or in vivo. Optionally, the compositions comprise one or more of the recombinant adenovirus 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. The pharmaceutical preparation is preferably 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.


In some embodiments, the compositions include at least two different recombinant adenoviruses, such as recombinant adenoviruses that encode different transgenes. In some examples, the composition includes two, three, four, five or six different recombinant adenoviruses.


In therapeutic applications, recombinant adenoviruses or compositions thereof are administered to a subject in an effective amount or dose. Single or multiple administrations of the compositions may be administered as needed. A “patient” or “subject” includes both humans and other animals, particularly mammals. Thus, the methods are applicable to both human therapy and veterinary applications.


An effective amount of a recombinant adenovirus 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 general health. For example, for administration to 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 therapeutic agents, such as one or more agents for the treatment of cancer, such as pancreatic cancer or glioblastoma.


Administration of the synthetic adenoviruses disclosed herein that harbor a therapeutic transgene can be accompanied by administration of other anti-cancer agents or therapeutic treatments (such as surgical resection of a tumor). Any suitable anti-cancer agent can be administered in combination with the recombinant viruses disclosed herein. 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), CDK inhibitors and anti-angiogenesis agents. Other anti-cancer treatments include radiation therapy and other antibodies that specifically target cancer cells (e.g., biologics).


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).


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.


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, CIN K4, 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).


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.


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
A Synthetic Adenovirus Expressing an Ad5/Ad34 Chimeric Fiber Protein and Liver Detargeting Modifications

While the fiber proteins of Ad5 and many other serotypes have been shown to bind to CAR for cellular attachment, other serotypes have been shown to use CD46 (Gaggar et al., Nat Med 9:1408-1412, 2003), desmoglein 2 (Wang et al., Nat Med 17:96-104, 2011), sialic acid (Nilsson et al., Nat Med 17:105-109, 2011), or others (Arnberg, Trends Pharmacol Sci 33:442-448, 2012). The receptor usage of many serotypes has not been thoroughly examined and CD46 is not thought to be expressed in mature mice.


Adsembly/AdSLIC (see PCT Publication No. WO 2012/024351, incorporated herein by reference) was used to generate a synthetic adenovirus having a chimeric fiber protein. Since the globular knob at the C-terminus of the fiber protein is typically responsible for receptor binding, a virus with a chimeric fiber protein was created by replacing the Ad5 fiber knob with fiber knob from Ad34 (AdSyn-CO176). The control virus (AdSyn-CO171) contains an Ad5 fiber protein (i.e. both the shaft and knob domains are from Ad5). Both viruses were created with the same E1 module containing an E1A/E1B deletion and a luciferase-GFP fusion driven by an EF1α promoter (Table 1). The recombinant viruses also include liver detargeting modifications. Natural Ad5 vectors will only infect the lungs (via inhalation) or liver (via intravenous administration). Ad5 hexon binds to Factor X in the blood, which leads to its absorption by Kuppfer cells in the liver, preventing systemic dissemination and inducing limited inflammation. To overcome this and allow for systemic administration to alternative cell types, the synthetic adenoviruses were engineered to include additional genomic modifications in the E1 and core regions that prevent uptake and expression in the liver. Specifically, both viruses include binding sites in the 3′UTR of the transgene for a microRNA that is specifically expressed in the liver (miR-122) and an E451Q mutation in hexon.









TABLE 1







Adenoviruses with Chimeric Fiber Proteins and Liver


Detargeting Modifications













SEQ







ID


Virus Name
NO:
E1
L3
L5
E4





AdSyn-CO171
1
ΔE1 + EF1α-
hexon
wt
wt




luciferase-miR122
E451Q


AdSyn-CO176
2
ΔE1 + EF1α-
hexon
Ad34 knob
wt




luciferase-miR122
E451Q
Ad5 shaft






fiber chimera









Example 2
A Synthetic Adenovirus Expressing the Ad34 Knob Domain Exhibits Tropism to Tumor Stroma

This example describes the finding that AdSyn-CO176, expressing a chimeric fiber protein with an Ad34 knob domain, specifically traffics to tumor stroma.


Pancreatic Tumor Models


Shown in FIG. 1A is a schematic overview of the Cre-LoxP KrasG12D/p53 pancreatic tumor model. Mice designated as “Kras; p53/p53” encode the KrasG12D oncogene downstream of the sequence encoding LoxP-stop codon-LoxP. The stop codon blocks the expression of mutant Kras (KrasG12D) in the absence of Cre recombinase. However, in the presence of Cre recombinase, the stop codon is removed and allows for expression of the KrasG12D oncogene. In these same mice, both alleles of the p53 gene are flanked by LoxP sites (LoxP-p53-LoxP). Mice designated “p53/p53; Cre” also have both alleles of the p53 gene flanked by LoxP (LoxP-p53-LoxP), and they express the Cre recombinase transgene driven by the pancreatic and duodenal homeobox 1 (Pdx1) promoter. Pdx1 is a gene that is expressed specifically in the pancreatic cells, and thus both copies of p53 are deleted in the pancreatic cells. Breeding between the strains gives rise to offspring in which the Pdx1 promoter-driven Cre mediates the deletion of both alleles of the tumor suppressor p53 and activation of the mutant KrasG12D in pancreatic cells. Homozygous mice designated “Kras; p53/p53; Cre” develop pancreatic tumors in 5-7 weeks. AdSyn-CO176 was injected intravenously into Kras; p53/p53 and Kras; p53/p53; Cre mice. Seventy-two hours after the injection of virus, tissues were collected, incubated for 5 minutes with luciferin, and then scanned for 5 minutes using the IVIS™ imaging system. As shown in FIG. 1B, the Kras; p53/p53 mouse had a normal pancreas, and the luciferase signal was mainly from the spleen. In contrast, the Kras; p53/p53; Cre mouse had pancreatic tumors, and the signal was mainly from the pancreatic tumor.


Another study was performed in a Cre-mediated genetic manipulation heterozygous model (FIG. 2A). Mice designated “p53/+; Cre” have one wild type p53 allele and one p53 allele flanked by LoxP sites (LoxP-p53-LoxP). Breeding between the Kras; p53/p53 and p53/+; Cre strains gives rise to offspring in which the Pdx1 promoter-driven Cre recombinase mediates the deletion of a single allele of the tumor suppressor p53 and activation of the mutant KrasG12D in pancreatic cells. These heterozygous mice, designated “Kras; p53/+; Cre,” develop tumors later in life (at 4-9 months of age) due to the fact that they have one wild type allele of p53. This wild type allele must be lost or mutated in order for pancreatic tumors to develop. AdSyn-CO176 was injected intravenously into 4-month old p53/+; Cre and Kras; p53/+; Cre mice. Seventy-two hours after the injection of virus, tissues were collected, incubated with luciferin for 5 minutes, and then scanned for 1 minute using the IVIS™ imaging system. The p53/+; Cre mouse had a normal pancreas, and the signal was mainly from spleen. In contrast, the Kras; p53/+; Cre mouse at 4 months of age had a pancreatic tumor, and the signal was mainly from the tumor and liver.


The heterozygous Kras; p53/+; Cre mice develop pancreatic tumors in 4-9 months. To test whether AdSyn-CO176 can infect pancreatic tumors at a very early stage of tumor development (before tumors are visible), AdSyn-CO176 was injected intravenously into Kras; p53/+; Cre mice at 2 months of age. Seventy-two hours after the injection of virus, tissues were collected, incubated with luciferin for 5 minutes, and scanned for 4 minutes using the IVIS™ imaging system. The pancreas of Kras; p53/+; Cre mouse at 2 months of age looked normal, but luciferase signal was found in this tissue (FIG. 3A). H&E staining was performed to evaluate histology of the pancreas following infection with AdSyn-CO176. For comparison, FIG. 3B shows the typical histology of normal pancreas tissue and pancreatic tumor tissue. H&E staining of the pancreas from a Kras; p53/+; Cre mouse at 2 months of age showed that a small part of the pancreas was developing the tumor (FIG. 3C, indicated by the polygon), while most of the pancreas tissue appeared normal. This result indicated that AdSyn-CO176 can infect pancreatic tumors at a very early stage.


In pancreatic tumors, only 10% of the cells are cancer cells; the remaining 90% are stromal cells. To determine which cell type was targeted by AdSyn-CO176, IHC and IF staining were performed. CK19 is a marker of tumor cells, while smooth muscle actin (SMA) is a marker of stromal cells. IHC staining of a pancreatic tumor infected with AdSyn-CO176 showed that GFP, which was expressed from AdSyn-CO176, overlapped with SMA staining, indicating that AdSyn-CO176 targeted stromal cells (FIG. 4A). IF staining of a pancreatic tumor infected by AdSyn-CO176 also demonstrated that GFP overlapped with SMA staining (FIG. 4B), confirming that AdSyn-CO176 infects stromal cells.


Glioblastoma Model


Shown in FIG. 5A is a schematic of a Cre-mediated genetic manipulation glioblastoma model. Lentiviruses were injected directly into the brain of GFAP-Cre mice. Glial fibrillary acidic protein (GFAP) promoter-driven Cre recombinase cleaves out RFP from the lentivirus-encoded gene and induces the expression of HRasV12 and GFP primarily in astrocytes. Expression of lentivirus-encoded U6-p53 shRNA knocks down the expression of p53 in the brain cells that take up the virus. The expression of HRasV12 and the knock down of p53 induces tumorigenesis in the brain from 1 week after the injection. GFP signal is used to indicate the formation of glioblastoma. Saline, AdSyn-CO171, or AdSyn-CO176 were injected via intravenous (IV) administration into GFAP-Cre mice that had received the tumor-inducing lentiviruses 4 weeks earlier. Forty-eight hours after the injection of virus, mice were scanned for 1 minute using the IVIS™ imaging system 5 minutes after the intraperitoneal injection of luciferin (FIG. 5B). A luciferase signal was detected in mice injected with AdSyn-CO176, but not in mice injected with saline or AdSyn-CO171.


Wild type mice (normal brain) and GFAP-Cre mice that had received injection of tumor-inducing lentiviruses were injected with AdSyn-CO171 or AdSyn-CO176. Brain tissues were collected 72 hours after the injection of synthetic adenoviruses, incubated with luciferin for 5 minutes, and scanned for 5 minutes using the IVIS™ imaging system (FIG. 5C). Only the GFAP-Cre mouse injected with the tumor-inducing lentiviruses showed a luciferase signal from AdSyn-CO176. This demonstrates that AdSyn-CO176 traffics to the brain tissue only when a tumor is present. Brain tissues were also scanned for the GFP signal (FIG. 5D). The GFP signal is used to identify the glioblastoma. Both of the GFAP-Cre mice that received the tumor-inducing lentiviruses had the GFP signal in the brain, while no GFP was detected in wild type mice. The GFP signal overlapped with the luciferase signal perfectly in the GFAP-Cre mouse that received the tumor-inducing lentiviruses and AdSyn-CO176.


The injection of the tumor-inducing lentiviruses causes temporary injury to the brain at the injection site. Although the synthetic adenoviruses (AdSyn-CO171 and AdSyn-CO176) were injected 4 weeks after the initial injection of lentiviruses, it was still unclear whether trafficking of AdSyn-CO176 to the glioblastoma was driven by the tumor or by the injection site injury. To answer this question, GFAP-Cre mice were injected with synthetic adenovirus 4 weeks after either no injection, sham-injection or injection with tumor-inducing lentivirus. GFAP-Cre mice were injected with either Hanks' balanced salt solution (HBSS) or tumor-inducing lentiviruses. After 4 weeks, AdSyn-CO171 was injected intravenously. As shown in FIG. 6A, there was no luciferase signal from AdSyn-CO171 in the brain in either group of mice. GFAP-Cre mice were injected with HBSS or tumor-inducing lentiviruses, or received no injection. After 4 weeks, mice were injected intravenously with AdSyn-CO176. As shown in FIG. 6B, the luciferase signal was detected only in the brain of the mouse that received the tumor-inducing lentiviruses, while the mouse that received HBSS or no injection produced no signal. These results demonstrate that the specificity of AdSyn-CO176 is driven by the tumor and not injection site injury.


Example 3
AdSyn-CO176 Traffics to Human Glioblastoma Tumors in a Xenograft Model

This example describes the finding that a synthetic adenovirus expressing a chimeric fiber protein with an Ad34 knob domain is capable of targeting human glioblastoma tumors.


The U87-tdTomato cell line is a human glioblastoma cell line that expresses the tdTomato fluorescent protein as a reporter to enable monitoring of tumor growth. When U87-tdTomato cells are injected intracranially into NSG mice to generate glioblastoma tumors, it typically takes 4-8 weeks for tumors to develop (FIG. 7A). This glioblastoma xenograft model was used to determine whether AdSyn-CO176 could traffic to a human glioblastoma tumor. AdSyn-CO171 (SEQ ID NO: 1) and AdSyn-CO176 (SEQ ID NO: 2) were injected intravenously into NSG mice by tail vein injection four weeks after the mice had received an intracranial injection of U87-tdTomato cells. Forty-eight hours after the injection of viruses, liver, spleen and brain tissue were collected, incubated with luciferin for five minutes and then scanned for 1 minute using the IVIS™ imaging system. As shown in FIG. 7B, only the AdSyn-CO176 injected mice showed a luciferase signal in the brain, and this signal completely overlapped with tdTomato expression. Thus, these results demonstrate that AdSyn-CO176 can traffic to human glioblastoma tumors, while AdSyn-CO171 cannot.


Example 4
A Synthetic Adenovirus Targeting Tumor Stroma and Expressing a Therapeutic Transgene Reduces Tumor Size in an Animal Model

This example demonstrates that a synthetic adenovirus expressing a chimeric fiber protein with an Ad34 knob domain and a therapeutic payload is capable of tracking to tumor stroma and reducing tumor size.


Studies were performed to determine whether a therapeutic transgene could be incorporated into AdSyn-CO176 (SEQ ID NO: 2) to enable treatment of tumors. To conduct this study, the KPCL (KrasG12D; p53 knockout; Pdx1-Cre; firefly Luciferase) mouse model was used (FIG. 8A). KPCL mice are similar to homozygous “Kras; p53/p53; Cre” mice, which specifically express KrasG12D in the pancreas and have the p53 gene knocked out only in the pancreas. However, KPCL mice also specifically express firefly luciferase in the pancreas. The development of tumors in KPCL mice is also similar to the “Kras; p53/p53; Cre” mice.


Two additional synthetic adenoviruses were generated—AdSyn-CO987 (SEQ ID NO: 5) and AdSyn-CO989 (SEQ ID NO: 6). AdSyn-CO987 is a synthetic adenovirus based upon AdSyn-CO176. The herpes simplex virus-1 thymidine kinase (TK)/ganciclovir (GCV) suicide gene was cloned into AdSyn-CO176 to replace the firefly luciferase/GFP gene. A Renilla luciferase was also inserted just after TK in the genome of AdSyn-CO176. Control virus AdSyn-CO989 was generated by cloning TK-P2A-renilla luciferase into AdSyn-CO171 to replace the original firefly luciferase/GFP gene.


KPCL mice were injected intravenously via the tail vein with 1×106 PFU of AdSyn-CO987 or AdSyn-CO989 at 5-6 weeks of age. Two days later, the mice were i.p. or i.v. injected with GCV. Three control groups were used: AdSyn-CO989+GCV; AdSyn-CO987 followed with saline injection (AdSyn-CO987+saline); and GCV injection only (i.p. or i.v.). FIG. 8B provides a table showing the average survival of KPCL mice for each treatment group. Treatment with AdSyn-CO987+GCV extended mouse survival compared with controls.


The firefly luciferase signal expressed by tumors was analyzed during treatment to monitor tumor growth. The results are shown in FIG. 8C. The treatment for mouse Z619R was AdSyn-CO987+saline, which served as the control. Mice Z601R and Z607R were treated with AdSyn-CO987+GCV (i.p.). While the strength of the firefly luciferase signal increased in the control mouse Z619R (indicating an increase in tumor size), the signal decreased in mice Z601R and Z607R (indicating a reduction in tumor size).


Histology of the pancreatic tumors was also evaluated by H&E staining (FIG. 9A). Mice Z655, 1806, Z619 and Z621 were all control mice. Mouse Z655 was treated with i.p. injected GCV only; mouse 1806 was treated with i.v. injected GCV only; mouse Z619 was treated with AdSyn-CO987+saline; and mouse Z621 received no treatment. Mouse Z656 received treatment with AdSyn-CO987+GCV i.v. Compared to the controls, the tumor from Z656 had more regions of necrosis (FIG. 9B).


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 method of expressing a transgene in stromal cells in a tumor, comprising infecting the stromal cells in the tumor with a synthetic adenovirus comprising: the transgene inserted into the E1 region, wherein the transgene encodes an anti-cancer agent;a native or modified capsid that detargets the synthetic adenovirus from the liver; and an adenovirus type 34 (Ad34) fiber protein or a chimeric fiber protein comprising an adenovirus type 5 (Ad5) shaft domain and an Ad34 knob domain,wherein the stromal cells in the tumor express the transgene.
  • 2. The method of claim 1, wherein the synthetic adenovirus comprises a modified capsid that detargets the synthetic adenovirus from the liver.
  • 3. The method of claim 1, wherein the synthetic adenovirus further comprises one or more binding sites for a liver-specific microRNA.
  • 4. The method of claim 3, wherein the liver-specific microRNA is miR-122.
  • 5. The method of claim 1, wherein the synthetic adenovirus further comprises one or more binding sites for a spleen-specific microRNA.
  • 6. The method of claim 5, wherein the spleen-specific microRNA is miR142-3p.
  • 7. The method of claim 1, wherein expression of the transgene is regulated by a tissue-specific promoter.
  • 8. The method of claim 1, wherein the synthetic adenovirus comprises Ad5 capsid proteins and a chimeric fiber protein comprising an Ad5 shaft domain and an Ad34 knob domain.
  • 9. The method of claim 1, wherein the tumor is a pancreatic tumor or a glioblastoma.
  • 10. The method of claim 1, wherein the transgene encodes an anti-cancer agent selected from the group consisting of a pro-inflammatory molecule, a cytokine, an anti-angiogenic factor, an inhibitor of KRas, and an inhibitor of cytotoxic T lymphocyte-associated molecule (CTLA)-4, programmed cell death protein 1 (PD-1), carcinoembryonic antigen (CEA) and mucin 1 (MUC1).
  • 11. The method of claim 1, wherein the anti-cancer agent disrupts or kills the stromal cells in the tumor.
  • 12. The method of claim 1, wherein the transgene is operably linked to the EF1α promoter.
  • 13. The method of claim 1, wherein the transgene encodes a pro-inflammatory molecule or cytokine selected from the group consisting of granulocyte macrophage colony stimulating factor (GM-CSF), CD40 ligand (CD40L), Fms-related tyrosine kinase 3 (FLT3) ligand, interleukin (IL)-1b, IL-2, IL-4, IL-6, IL-12, tumor necrosis factor (TNF)-α, an interferon, a chemokine, B7-1, intercellular adhesion molecule (ICAM)-1, lymphocyte function-associated antigen (LFA)-3, transforming growth factor (TGF)-β, platelet derived growth factor (PDGF), and epidermal growth factor (EGF).
  • 14. The method of claim 13, wherein the transgene is operably linked to the EF1α promoter.
  • 15. The method of claim 1, wherein the transgene encodes an inhibitor of vascular endothelial growth factor (VEGF).
  • 16. The method of claim 15, wherein the transgene is operably linked to the EF1α promoter.
  • 17. A method of reducing the size of a tumor in a subject, comprising infecting stromal cells in the tumor of the subject with a synthetic adenovirus comprising: a therapeutic transgene inserted into the E1 region, wherein the therapeutic transgene encodes an anti-cancer agent;a native or modified capsid that detargets the synthetic adenovirus from the liver; and an adenovirus type 34 (Ad34) fiber protein or a chimeric fiber protein comprising an adenovirus type 5 (Ad5) shaft domain and an Ad34 knob domain,wherein the stromal cells in the tumor express the transgene, andwherein expression of the transgene results in a reduction in the size of the tumor.
  • 18. The method of claim 17, wherein the therapeutic transgene encodes an anti-cancer agent selected from the group consisting of a pro-inflammatory molecule, a cytokine, an anti-angiogenic factor, an inhibitor of KRas, and an inhibitor of cytotoxic T lymphocyte-associated molecule (CTLA)-4, programmed cell death protein 1 (PD-1), carcinoembryonic antigen (CEA), and mucin 1 (MUC1).
  • 19. The method of claim 17, wherein the anti-cancer agent disrupts or kills the stromal cells in the tumor.
  • 20. The method of claim 17, wherein the transgene is operably linked to the EF1α promoter.
  • 21. The method of claim 17, wherein the tumor is a pancreatic tumor.
  • 22. The method of claim 17, wherein the tumor is a glioblastoma.
  • 23. A synthetic adenovirus genome, comprising a nucleotide sequence at least 95% identical to SEQ ID NO: 5.
  • 24. The synthetic adenovirus genome of claim 23, comprising SEQ ID NO: 5.
CROSS REFERENCE TO RELATED APPLICATIONS

This application a continuation of International Application No. PCT/US2017/065604, filed Dec. 11, 2017, published in English under PCT Articles 21(2), which claims the benefit of U.S. Provisional Application No. 62/433,140, filed Dec. 12, 2016, which are both herein incorporated by reference in their entireties.

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'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 Carrióet 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 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 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 et al. 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. Mar 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 Carrióet 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 (172)
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 2003092579 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 2012038606 Mar 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 (97)
Entry
Fukazawa et al. Adenovirus-mediated Cancer Gene Therapy and Virotherapy (Review). International Journal of Molecular Medicine , 2010. 25:3-10.
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.
Bremnes et al., “The Role of Tumor Stroma in Cancer Progression and Prognosis,” J. Thorac. Oncol., vol. 6:209-217, 2011.
Yaghoubi et al., “Positron Emission Tomography Reporter Genes and Reporter Probes: Gene and Cell Therapy Applications,” Theranostics, vol. 2:374-391, 2012.
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 d1309,” 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.
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.
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.
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.
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. Viral., vol. 78(22): 12333-12343, 2004.
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.
Funston et al., “Expression of heterologous genes in oncol ytic adenoviruses using picornaviral 2A sequences that trigger ribosome skipping,” J Gen Viral 89:389-396, 2008.
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 EIA 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.
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.
Kirn, “Clinical research results with d11520 (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 E1A 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.
Liu et al., “Oncolytic Adenoviral Vector Carrying the Cytosine Deaminase Gene for Melanoma Gene Therapy,” Cancer Gene Ther., vol. 13:845-855, 2006.
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.
Minskaia et al., “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.
Murakami et al., “Chimeric Adenoviral Vectors Incorporating a Fiber of Human Adenovims 3 Efficiently Mediate Gene Transfer into Prostrate Cancer Cells,” The Prostate, vol. 70:362-376, 2009.
NCBI Accession No. CV1 10986, Jan. 11, 2011, 3 pages.
Nevels et al., “The Adenovirus E4orf6 can Promote E1A/E1B-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. 2013 80014047.7, dated Aug. 5, 2015 (English translation).
Ono et al., “Noninvasive Visualization of Adenovirus Replication with a Fluorescent Reporter in the E3 Region,” Cancer Res., vol. 65: 10154-10158, 2005.
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.
Pelka et al., “Adenovirus EIA Directly Targets the E2F/DP-1 Complex,” J Viral 85(17):8841-8851, 2011.
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).
Szymczak et al., “Correction of Multi-Gene Deficiency in vivo using a Single ‘selfcleaving’ 2A Peptide-Based Retroviral Vector,” Nature Biotech., vol. 22:589-594, 2004.
Tan et al., “Coexpression of double or triple 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.
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 E1A-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 E1A Proteins Bind to the Retinoblastoma Gene Product,” Nature 334:124-129, 1988.
Yount et al., “Strategy for Systematic Assembly of Large RNA and DNA Genomes: Transmissible Gastroenteritis Virus Model,” J. Viral., vol. 74: 10600-10611, 2000.
Fueyo et al., “A mutant Oncolytic Adenovirus Targeting the Rb Pathway Produces Anti-Glioma Effect In Vivo,” Oncogene, vol. 19:2-12, 2000.
Ji et al., “Oncolytic Adenoviruses Delivering Herpes Simplex Virus Thymidine Kinase Suicide Gene Reduces the Growth of Human Retinoblastoma in an in vivo Mouse Model,” Experimental Eye Res., vol. 89:193-199, 2009.
Kubo et al., “Complete Regression of Human Malignant Mesothelioma Xenografts Following Local Injection of Midkine Promoter-Driven Oncolytic Adenovirus,” J. Gene Med., vol. 12:681-692, 2010.
Li, “Self-Cleaving Fusion Tags for Recombinant Protein Production,” Biotechnol. Lett., vol. 33:869-881, 2011 .
Loskog, “Immunostimulatory Gene Therapy Using Oncolytic Viruses as Vehicles,” Viruses, vol. 7:5780-5791. 2015.
Wu et al., “The Application of Intein in the Research of Membrane Protein,” Chemistry of Life, vol. 35:200-205, 2015 (with English language abstract).
Hibma et al., “Increased apoptosis and reduced replication efficiency of the E3 region-modified dl309 adenovirus in cancer cells,” Virus Res 145:112-120, 2009.
Dias et al., “Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4,” Gene Therapy, vol. 19:988-998, 2012.
Kurihara et al., “Selectivity of a replication-competent adenovirus for human breast carcinoma cells expressing the MUCI antigen,” J. Clin. Invest., vol. 106:763-771, 2000.
Related Publications (1)
Number Date Country
20190314523 A1 Oct 2019 US
Provisional Applications (1)
Number Date Country
62433140 Dec 2016 US
Continuations (1)
Number Date Country
Parent PCT/US2017/065604 Dec 2017 US
Child 16434714 US