WO2009052426A2 - Oncolytic virus - Google Patents

Oncolytic virus Download PDF

Info

Publication number
WO2009052426A2
WO2009052426A2 PCT/US2008/080367 US2008080367W WO2009052426A2 WO 2009052426 A2 WO2009052426 A2 WO 2009052426A2 US 2008080367 W US2008080367 W US 2008080367W WO 2009052426 A2 WO2009052426 A2 WO 2009052426A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
virus
cells
seq
vasculostatin
Prior art date
Application number
PCT/US2008/080367
Other languages
French (fr)
Other versions
WO2009052426A3 (en
Inventor
Balveen Kaur
Antonio Chiocca
Yoshinaga Saeki
Original Assignee
The Ohio State University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Ohio State University filed Critical The Ohio State University
Publication of WO2009052426A2 publication Critical patent/WO2009052426A2/en
Publication of WO2009052426A3 publication Critical patent/WO2009052426A3/en
Priority to US12/697,891 priority Critical patent/US8450106B2/en
Priority to US13/903,695 priority patent/US20130316447A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/763Herpes virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/89Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microinjection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16632Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16641Use of virus, viral particle or viral elements as a vector
    • C12N2710/16643Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses

Definitions

  • the present invention is directed to the fields of virology, cancer biology, and medicine. More particularly, it concerns compositions and methods of treating cancer of the brain in a patient using oncolytic herpes simplex virus 1 (HSV-I ) armed with therapeutic transgenes.
  • HSV-I oncolytic herpes simplex virus 1
  • Malignant tumors that are intrinsically resistant to conventional therapies are significant therapeutic challenges.
  • Such malignant tumors include, but are not limited to malignant gliomas and recurrent systemic solid tumors such as lung cancer.
  • Malignant gliomas are the most abundant primary brain tumors having an annual incidence of 6.4 cases per 100,000 (CBTRUS, 2002-2003). These neurologically devastating tumors are the most common subtype of primary brain tumors and are one of the deadliest human cancers.
  • GBM glioblastoma multiforme
  • median survival duration for patients is 14 months, despite maximum treatment efforts (Stupp et al., 2005).
  • a prototypic disease, malignant glioma is inherently resistant to current treatment regimens (Shapiro and Shapiro, 1998). In fact, in approximately 1/3 of patients with GBM the tumor will continue to grow despite treatment with radiation and chemotherapy. Median survival even with aggressive treatment including surgery, radiation, and chemotherapy is less than 1 year (Schiffer, 1998). Because few good treatment options are available for many of these refractory tumors, the exploration of novel and innovative therapeutic approaches is essential.
  • Gene therapy is a promising treatment for tumors including gliomas because conventional therapies typically fail and are toxic.
  • identification of genetic abnormalities contributing to malignancies is providing crucial molecular genetic information to aid in the design of gene therapies.
  • Genetic abnormalities indicated in the progression of tumors include the inactivation of tumor suppressor genes and the overexpression of numerous growth factors and oncogenes.
  • Tumor treatment may be accomplished by supplying a polynucleotide encoding a therapeutic polypeptide or other therapeutic that target the mutations and resultant aberrant physiologies of tumors. It is these mutations and aberrant physiology that distinguishes tumor cells from normal cells.
  • a tumor-selective virus would be a promising tool for gene therapy.
  • HSV Oncolytic herpes simplex virus
  • HSV-1 based oncolytic viruses are particularly exciting because of: 1 ) their ability to infect a wide variety of tumors; 2) their inherent cytolytic nature; 3) their well characterized large genome (152 Kb) that provides ample opportunity for genetic manipulations wherein many of the non-essential genes (up to 30kb) can be replaced by therapeutic genes; 4) their ability to remain as episomes that avoid insertional mutagenesis in infected cells; and 5) the availability of anti-herpetic drugs to keep in check possible undesirable replication [38, 39].
  • the tumor microenvironment is also recognized as an important determinant for tumor progression.
  • Vasculature is a major component of the microenvironment of solid tumors such as malignant gliomas. Solid tumors depend on the development of a vasculature to provide them with nutrients. While tumor oncolysis is thought to set the stage for activating a systemic adaptive immune surveillance, innate defense mechanisms elicited upon OV infection are thought to be responsible for rapid viral clearance from tumor [55-59]. Thus, OV-induced inflammation and its attendant increased vasculature may be counterproductive to the goal of killing cancer cells.
  • the present embodiments address a long-felt need in the art by providing a potent oncolytic virus for therapy of undesirable cells, such as malignant cells.
  • a preferred embodiment provides an oncolytic virus capable of killing target cells, such as a tumor cells.
  • the conditionally replicating HSV comprises at least two mechanisms to rid a culture, tissue or organism of at least some undesirable cells, to inhibit proliferation of at least some undesirable cells, to prevent proliferation of at least some desirable cells, or a combination thereof.
  • a preferred embodiment may not only directly act on the cancer itself, but also may enhance tumor cell killing by influencing the microenvironment around the physical tumor.
  • the oncolytic virus is armed or encodes a therapeutic polypeptide.
  • Armed is a term that indicates that the virus contains a heterologous nucleic acid sequence encoding a polypepitde of interest or a nucleic acid comprising a polynucleotide of interest.
  • the nucleic acid encoding a therapeutic polypeptide may encode an angiostatic factor.
  • the nucleic acid encoding a therapeutic polypeptide encodes the polypeptide Vasculostatin.
  • Oncolytic viruses expressing angiostatic factors using a CMV promoter have demonstrated limited efficacy in rodent models of glioma (for example, US Pub. No. 2006/0147420 and US Pub. No. 2004/0009604, incorporated herein by reference). Given the rapid lytic cycle of HSV, the challenge has been to ensure robust expression of the therapeutic protein before lysis of the infected cell.
  • a preferred embodiment overcomes this challenge by utilizing the immediate early HSV promoter IE4/5 operably linked to a therapeutic transgene.
  • the transgene is angiostatic.
  • the transgene is a novel, brain specific angiostatic polypeptide, Vasculostatin.
  • robust expression of a therapeutic gene can be seen at least as early as 4 hours.
  • the oncolytic virus of an exemplary embodiment can be delivered by a number routes including, but not limited to intracranial (into the skull cavity) intra- tumoral or intravenous administration.
  • the tumor may be a primary tumor or it may be a tumor resulting from a metastasis to the skull or brain.
  • a recombinant oncolytic virus has been generated that can specifically replicate in cancer cells leading to their destruction and at the same time secrete robust amounts of an angiostatic factor to inhibit the regrowth of residual disease.
  • Such a dually armed OV destroys cancer cells through its tumor specific replication potential and also targets tumor vasculature to enhance therapeutic efficacy.
  • a dually armed OV has been generated and shown that it does express and secrete the therapeutic anti-angiogenic factor (Vasculostatin), a novel brain specific anti-angiogenic factor, even at early time points after infection.
  • At least one embodiment exploits the robust transgene expression from an early viral promoter to maximize the expression of Vasculostatin.
  • the expression profile of a preferred embodiment allows for maximal expression of therapeutic transgenes before the lytic phase.
  • At least one embodiment has shown efficacy in mice with established brain tumors. Compositions and methods disclosed herein have broad therapeutic applicability to most solid cancers.
  • Expression of Vasculostatin does not interfere with the virus's cytotoxicity to glioma cells.
  • the expressed Vasculostatin is functional. Additionally, the generated OV has therapeutic advantage over the control virus for the treatment of mice with established brain tumors.
  • FIGURE 1 shows the survival of rats implanted with U87 human glioma cells stably expressing Vasculostatin (clones U14 and U 18) with parental untransfected glioma cells (U87). Note that survival of rats implanted with cells expressing Vasculostatin was significantly greater than that of rats implanted with control parental U87MG cells (P ⁇ 0.05). (Kaur et al., unpublished results).
  • FIGURE 2 provides a schematic illustration of the steps utilized to first clone the cDNA encoding for Vasculostatin under IE4/5 promoter into a shuttle plasmid (ptransferlE4/5) in order to generate pVasculo-transfer
  • FIGURE 3 provides a schematic illustration of the steps utilized to generate rHSVQvasculo (also called "RAMBO”)
  • FIGURE 4 shows a western blot analysis confirming the expression of
  • Vasculostatin by the recombinant viral isolates vascular endothelial growth factor
  • FIGURE 5 shows a time course western analysis of Vasculostatin production by both OV isolates in LN229 cells
  • FIGURE 6 shows a cytotoxicity assay for both viral isolates with U87 ⁇ EGFR cells. Note that there is no significant difference in cytotoxicity between the control rHSVQ virus and the isolated rHSVQvasculol , and rHSVQvasculo2 expressing Vasculostatin. [0028] FIGURE 7 demonstrates the anti-angiogenic capabilities of rHSVQvasculo.
  • the experiment was performed using the Trevigen Direct In Vivo Angiogenesis Assay (DIVAATM) Inhibition Kit.
  • DIVAATM Trevigen Direct In Vivo Angiogenesis Assay
  • FIGURE 8 shows a Kaplan-Meier survival analysis of mice treated with rHSVQ control virus or rHSVQvasculo (the virus generated to express Vasculostatin).
  • mice were treated by direct intratumoral injection on day 5 after tumor implantation.
  • FIGURE 9 shows the results of an experiment to compare the cytotoxicity of rHSVQvasculo to rHSVQ (an OV equivalent to the G207 being tested in clinical trials) toward normal human astrocytes.
  • Normal human astrocytes NHA, CellSciences Canton, MA
  • MOh with rHSVQ and rHSVQvasculo were infected at different multiolicities of infection (MOh with rHSVQ and rHSVQvasculo to evaluate potential cytotoxicity of OV produced Vasculostatin towards
  • FIGURE 10 shows a comparison of the effect of Vasculostatin expression on ability of OV to be cytotoxic to glioma cells in the glioma cells LN229., U87 ⁇ EGFR, and
  • FIGURE 11 is a western blot demonstrating that cyclophosphamide (CPA) pretreatment enhances the anti-tumor ability of Vasculostatin.
  • CPA cyclophosphamide
  • Gliomas are the most common primary tumors of the central nervous system
  • Glioblastoma multiforme Glioblastoma multiforme (GBM), the most aggressive form (WHO grade IV) of malignant astrocytoma, is highly invasive and vascularized [40] and characterized by 1 ) rapidly proliferating endothelial cells that form tufted aggregates referred to as glomeruloid bodies and 2) multiple hypoxic-necrotic areas within the tumor that drive hypoxia-mediated activation of hypoxia inducible factor (HIF), which thereby leads to increased transcription of factors, such as vascular endothelial growth factor (VEGF), that heralds a phase of more malignant tumor growth [41].
  • HIF hypoxia inducible factor
  • VEGF vascular endothelial growth factor
  • Their very aggressive growth and highly vascular nature makes malignant gliomas an attractive target for testing the effects of anti-angiogenic gene therapy.
  • the increased vascularization essential for malignant progression is triggered by disruption of the normal homeostasis between angiogenic and angiostatic factors within the tumor microenvironment [42]. It has been shown that expression of angiogenesis inhibitors is reduced in GBMs but not in normal brain and benign gliomas [43, 44]. Although not to be limited by theory, physiologically occurring factors that inhibit angiogenesis which are lost during tumor progression should represent molecules of choice for restoration by gene therapy.
  • Vasculostatin a fragment of Brain Angiogenesis inhibitor 1 (BAM ) (GenBank
  • Vasculostatin 1 is expressed at high levels primarily in normal brain but not in most GBMs and 2) has potent anti-angiogenic, anti- tumorigenic, and anti-permeability properties and 3) the ability to target multiple receptors on endothelial cells ( ⁇ v ⁇ 3, ⁇ v ⁇ 5, and CD36), and 4) its over expression is well tolerated in brain tissue.
  • Vasculostatin may be a better candidate than other more popular and not so novel anti-angiogenic factors for therapy of GBMs.
  • HSV-1 -derived OVs that express endostatin and, more recently, oncolytic viruses that express platelet factor IV and dominant negative FGF receptor under the control of a CMV promoter have been described [47-49].
  • at least one embodiment disclosed herein is better for GBM therapy because of its combination with an IE4/5 promoter (SEQ ID NO:5) that drives the expression of the transgene to levels unseen with a CMV promoter.
  • Vasculostatin a novel angiostatic factor, has been successfully expressed as part of an oncolytic viral stategy and shown to successfully counter anti-therapeutic changes in residual disease after oncolysis.
  • Specific replication within tumor cells can be achieved by OVs genetically engineered for that purpose or by naturally occurring strains of some viruses that have such propensity [25].
  • Specific embodiments utilize one such mutant, designated G207, which comprises an F-strain derived HSV-1 with deletions in both copies of the ⁇ 34.5 gene (encoding for the viral ICP34.5 protein) and an inactivating insertion of Escherichia coli (E. coli) lacZ into the viral ICP6/RR gene (encoding for the large subunit of ribonucleotide reductase).
  • HSV-1 with gamma34.5 deletion does not lead to the reactivation of wild-type HSV-1 , produce toxicity from infection of neurons surrounding the glioma cavity, or lead to encephalitis or meningitis.
  • Angiogenesis is critical for the development and maintenance of glioblastomas, the most malignant and common form of primary brain tumors. Combining oncolysis with anti-angiogenesis may produce a synergistic effect since the anti-cancer mechanisms are different but complementary.
  • a preferred embodiment allows an anti- angiogenic nucleic acid or polypeptide, such as, but not limited to a Vasculostatin protein, to be produced, ultimately favoring delivery to the extracellular compartment. For that reason, the oncolytic HSV-1 is used as an improved HSV vector to deliver high and continuous levels of Vasculostatin to the tumor.
  • Angiogenesis refers to vessel formation by remodeling the primary vascular network or by sprouting from existing vessels (reviewed in Yancopoulos et al., 2000).
  • the "angiogenesis switch” is “off” when the effect of pro-angiogenic molecules is balanced by the activity of anti-angiogenic molecules, and is “on” when the net balance between the molecules is tipped in favor of angiogenesis (reviewed in Carmeliet and Jain, 2000).
  • Angiogenesis has an essential role in the development and maintenance of solid tumors, including malignant gliomas.
  • Embodiments of this invention may include other heterologous genes.
  • they may include therapeutic genes, pro-drug converting enzymes, cytosine deaminase (to convert 5-FC to 5-FU), a yeast cytosine deaminase, a humanized yeast cytosine deaminase, an image enhancing polypeptides, a sodium-iodide symporter, anti- sense or inhibitory VEGF, Bcl-2, Ang-2, or interferons alpha, beta or gamma.
  • cytosine deaminase to convert 5-FC to 5-FU
  • yeast cytosine deaminase to convert 5-FC to 5-FU
  • humanized yeast cytosine deaminase a humanized yeast cytosine deaminase
  • an image enhancing polypeptides a sodium-iodide symporter
  • anti- sense or inhibitory VEGF Bcl-2, Ang-2
  • recombinant HSV-1 vector as used herein defines a recombinant
  • HSV-1 vector comprising: (a) the DNA of, or corresponding to, at least a portion of the genome of an HSV-1 which portion is capable of transducing into a target cell at least one selected gene and is capable of promoting replication and packaging; and (b) at least one selected gene (or transgene) operatively linked to regulatory sequences directing its expression, the gene flanked by the DNA of (a) and capable of expression in the target cell in vivo or in vitro.
  • rHSV recombinant HSV
  • a "gene” or a “sequence which encodes” a particular protein is a nucleic acid molecule which is transcribed (in the case of DNA) and translated (in the case of mRNA) into a polypeptide in vitro or in vivo when placed under the control of appropriate regulatory sequences. The boundaries of the gene are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus.
  • a gene can include, but is not limited to, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic DNA, and even synthetic DNA sequences.
  • a transcription termination sequence will usually be located 3' to the gene sequence.
  • polyadenylation signal is provided to terminate transcription of genes inserted into a recombinant virus.
  • polypeptide or "protein” means a linear polymer of amino acids joined in a specific sequence by peptide bonds.
  • amino acid refers to either the D or L stereoisomer form of the amino acid, unless otherwise specifically designated.
  • transgene refers to a particular nucleic acid sequence encoding a polypeptide or a portion of a polypeptide to be expressed in a cell into which the nucleic acid sequence is inserted.
  • the term “transgene” is meant to include (1) a nucleic acid sequence that is not naturally found in the cell (i.e., a heterologous nucleic acid sequence); (2) a nucleic acid sequence that is a mutant form of a nucleic acid sequence naturally found in the cell into which it has been inserted; (3) a nucleic acid sequence that serves to add additional copies of the same (i.e., homologous) or a similar nucleic acid sequence naturally occurring in the cell into which it has been inserted; or (4) a silent naturally occurring or homologous nucleic acid sequence whose expression is induced in the cell into which it has been inserted.
  • mutant form is meant a nucleic acid sequence that contains one or more nucleotides that are different from the wild-type or naturally occurring sequence, i.e., the mutant nucleic acid sequence contains one or more nucleotide substitutions, deletions, and/or insertions.
  • the transgene may also include a sequence encoding a leader peptide or signal sequence such that the transgene product may be secreted from the cell.
  • a proteolytic fragment of BAM , Vasculostatin is utilized to as the anti-angiogenic transgene.
  • a preferred embodiment comprises a polypeptide having the angiostatic activity of Vasculostatin, including, but not limited to the polypeptide of SEQ ID NO:2 that is encoded by the nucleic acid sequence of SEQ ID NO:1.
  • the transgene may optionally include nucleotides encoding a his and/or myc tag as in SEQ ID NO:3.
  • the promoter is preferably the immediate early promoter IE4/5 (SEQ ID NO: 1
  • a preferred embodiment provides a method for treating a neoplastic disease in a subject, the subject being an animal or human, comprising administering to the subject a therapeutically effective amount of a recombinant tumor-specific conditional replication oncolytic activity, the vector comprising a DNA sequence encoding an anti-angiogenic agent, the DNA is operably linked to a promoter.
  • the anti-angiogenic agent is vasculostatin (which is a fragment of brain angiogenesis inhibitor 1 (BAM )) or a biologically active variant thereof.
  • operably linked refers to the arrangement of various nucleic acid molecule elements relative to each other such that the elements are functionally connected and are able to interact with each other.
  • Such elements may include, without limitation, a promoter, an enhancer, a polyadenylation sequence, one or more introns and/or exons, and a coding sequence of a gene of interest to be expressed (i.e., the transgene).
  • the nucleic acid sequence elements when operably linked, act together to modulate the activity of one another, and ultimately may affect the level of expression of the transgene. By modulate is meant increasing, decreasing, or maintaining the level of activity of a particular element.
  • transduction of the transgene of the invention increases the expression of the transgene, preferably that of the angiostatic polypeptide Vasculostatin.
  • the position of each element relative to other elements may be expressed in terms of the 5' terminus and the 3' terminus of each element.
  • transfection is used to refer to the uptake of foreign DNA by a mammalian cell.
  • a cell has been "transfected” when exogenous DNA has been introduced inside the cell membrane.
  • transfection techniques are known in the art. See, Graham et al. (1973) Virology, 52:456; and Sambrook et al. (1989) Molecular Cloning, a laboratory manual, Cold Spring Harbor Laboratories, New York. Such techniques can be used to introduce one or more exogenous DNA moieties, such as a viral vector and other nucleic acid molecules, into suitable host cells.
  • the term refers to both stable and transient uptake of the genetic material.
  • the vectors of the preferred embodiments may be useful for the introduction of additional genes in gene therapy.
  • the HSV vector of this invention can contain an additional exogenous gene for the expression of a protein effective in regulating the cell cycle, such as p53, Rb, or mitosin, or a biologically active variant thereof, or in inducing cell death, such as the conditional suicide gene thymidine kinase, the latter must be used in conjunction with a thymidine kinase metabolite in order to be effective, or any other anti-tumor gene, such as for example a toxin.
  • neoplasm and “neoplastic” refer to a tumor and/or to an abnormal tissue, including metastatic disease, that grows by cellular proliferation more rapidly than normal, continues to grow after the stimuli that initiated the new growth cease, shows partial or complete lack of structural organization and functional coordination with normal tissue, and usually forms a distinct mass of tissue which may be either benign or malignant.
  • Neoplastic diseases include, but are not limited to, benign solid tumors, malignant solid tumors, benign proliferative diseases of the blood, and malignant proliferative diseases of the blood.
  • Representative examples include colon carcinoma, prostate cancer, breast cancer, lung cancer, skin cancer, liver cancer, bone cancer, ovary cancer, pancreas cancer, brain cancer, head and neck cancer, and lymphoma.
  • animal is intended to be synonymous with mammal and is to include, but not be limited to, bovine, porcine, feline, simian, canine, equine, murine, rat or human.
  • Host cells include, but are not limited to, any neoplastic or tumor cell, such as osteosarcoma, ovarian carcinoma, breast carcinoma, melanoma, hepatocarcinoma, lung cancer, brain cancer, colorectal cancer, hematopoietic cell, prostate cancer, cervical carcinoma, retinoblastoma, esophageal carcinoma, bladder cancer, neuroblastoma, or renal cancer.
  • OVs embodiments discussed herein can be combined with various pharmaceutically acceptable carriers. Suitable pharmaceutically acceptable carriers are well known to those of skill in the art.
  • the compositions can then be administered therapeutically or prophylactically, in effective amounts, described in more detail below.
  • therapeutically effective amount is intended to mean the amount of vector or of transformed cells, which exerts oncolytic activity, causing attenuation or inhibition of tumor cell proliferation leading to tumor regression. An effective amount will vary on the pathology or condition to be treated, by the patient and his status, and other factors well known to those of skill in the art. Effective amounts are easily determined by those of skill in the art.
  • oncolytic activity refers to cytotoxic effects in vitro and/or in vivo exerted on tumor cells without any appreciable or significant deleterious effects to normal cells under the same conditions.
  • the cytotoxic effects under in vitro conditions are detected by various means as known in prior art, for example, by staining with a selective stain for dead cells, by inhibition of DNA synthesis, or by apoptosis. Detection of the cytotoxic effects under in vivo conditions is performed by methods known in the art.
  • Methods of treating a neoplastic disease may include administration of the compounds of exemplary embodiments as a single active agent, or in combination with additional methods of treatment including, but not limited to, irradiation therapy, therapy with immunosuppressive agents, chemotherapeutic or anti-proliferative agents, including cytokines.
  • additional methods of treatment including, but not limited to, irradiation therapy, therapy with immunosuppressive agents, chemotherapeutic or anti-proliferative agents, including cytokines.
  • the methods of treatment of the invention may be in parallel to, prior to, or following additional methods of treatment.
  • any of the vectors described herein are useful for the treatment of a neoplastic disease.
  • the vectors of the invention can be combined with one or more pharmaceutically acceptable carriers.
  • Pharmaceutically acceptable carriers are well known in the art and include aqueous solutions such as physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, vegetable oils (e.g., olive oil) or injectable organic esters.
  • a pharmaceutically acceptable carrier can be used to administer the compositions of the invention to a cell in vitro or to a subject in vivo.
  • a pharmaceutically acceptable carrier can contain a physiologically acceptable compound that acts, for example, to stabilize the composition or to increase the absorption of the agent.
  • a physiologically acceptable compound can include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives, which are particularly useful for preventing the growth or action of microorganisms.
  • Various preservatives are well known and include, for example, phenol and ascorbic acid.
  • a pharmaceutically acceptable carrier including a physiologically acceptable compound
  • a physiologically acceptable compound such as aluminum monosterate or gelatin is particularly useful as a delaying agent, which prolongs the rate of absorption of a pharmaceutical composition administered to a subject.
  • carriers, stabilizers or adjutants can be found in Martin, Remington's Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton, 1975), incorporated herein by reference.
  • composition refers to any of the compositions of matter described herein.
  • the compositions can then be administered therapeutically or prophylactically. They can be contacted with the host cell in vivo, ex vivo, or in vitro, in a therapeutically effective amount. In vitro and in vivo means of transfecting the vectors of the invention are provided below.
  • any suitable route of administration of the vectors may be adapted, including but not limited to, intravenous, oral, buccal, intranasal, inhalation, topical application to a mucosal membrane or injection, including intratumoral, intradermal, intrathecal, intracisternal, intralesional or any other type of injection. Administration can be effected continuously or intermittently and will vary with the subject and the condition to be treated.
  • An exemplary embodiment includes an oncolytic HSV, such as created by methods described herein, for example random mutagenesis, and further comprises a nucleic acid encoding an angiostatic polypeptide, such as Vasculostatin.
  • angiostatic polypeptide such as Vasculostatin.
  • An exemplary embodiment is directed to an HSV vector.
  • the vector comprises some or all of the following components.
  • the term "vector” is used to refers to a carrier nucleic acid molecule into which a nucleic acid sequence can be inserted for introduction into a cell where it can be replicated.
  • a nucleic acid sequence can be "exogenous,” which means that it is foreign to the cell into which the vector is being introduced or that the sequence is homologous to a sequence in the cell but in a position within the host cell nucleic acid in which the sequence is ordinarily not found.
  • Vectors include plasmids, cosmids, viruses (bacteriophage, animal viruses, and plant viruses), and artificial chromosomes (e.g., YACs).
  • viruses bacteria, animal viruses, and plant viruses
  • artificial chromosomes e.g., YACs.
  • One of skill in the art would be well equipped to construct a vector through standard recombinant techniques (see, for example, Maniatis et al., 1988 and Ausubel et al., 1994, both incorporated herein by reference).
  • expression vector refers to any type of genetic construct comprising a nucleic acid coding for a RNA capable of being transcribed. In some cases, RNA molecules are then translated into a protein, polypeptide, or peptide. In other cases, these sequences are not translated, for example, in the production of antisense molecules or ribozymes.
  • Expression vectors can contain a variety of "control sequences,” which refer to nucleic acid sequences necessary for the transcription and possibly translation of an operably linked coding sequence in a particular host cell. In addition to control sequences that govern transcription and translation, vectors and expression vectors may contain nucleic acid sequences that serve other functions as well and are described infra. [0067] Promoters and Enhancers
  • an HSV immediate early viral promoter is operably linked to the transgene in order to drive the expression of the heterologous transgene. More preferrably, the early viral promoter utilized is the HSV immediate early viral promoter IE4/5 (SEQ ID NO:5).
  • promoter refers to a nucleic acid sequence that regulates, either directly or indirectly, the transcription of a corresponding nucleic acid coding sequence to which it is operably linked.
  • the promoter may function alone to regulate transcription, or, in some cases, may act in concert with one or more other regulatory sequences such as an enhancer or silencer to regulate transcription of the transgene.
  • the promoter comprises a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene, which is capable of binding RNA polymerase and initiating transcription of a downstream (3'- direction) coding sequence.
  • a promoter generally comprises a sequence that functions to position the start site for RNA synthesis.
  • the best-known example of this is the TATA box, but in some promoters lacking a TATA box, such as, for example, the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation. Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well.
  • a promoter To bring a coding sequence "under the control of” a promoter, one positions the 5' end of the transcription initiation site of the transcriptional reading frame "downstream" of (i.e., 3' of) the chosen promoter.
  • the "upstream” promoter stimulates transcription of the DNA and promotes expression of the encoded RNA.
  • promoter The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.
  • a promoter may or may not be used in conjunction with an "enhancer,” which refers to a cis-acting regulatory sequence involved in the transcriptional activation of a nucleic acid sequence.
  • a promoter may be one naturally associated with a nucleic acid sequence, as may be obtained by isolating the 5' non-coding sequences located upstream of the coding segment and/or exon. Such a promoter can be referred to as "endogenous.”
  • an enhancer may be one naturally associated with a nucleic acid sequence, located either downstream or upstream of that sequence.
  • certain advantages may be gained by positioning the coding nucleic acid segment under the control of a recombinant or heterologous promoter, which refers to a promoter that is not normally associated with a nucleic acid sequence in its natural environment.
  • a recombinant or heterologous enhancer refers also to an enhancer not normally associated with a nucleic acid sequence in its natural environment.
  • Such promoters or enhancers may include promoters or enhancers of other genes, and promoters or enhancers isolated from any other virus, or prokaryotic or eukaryotic cell, and promoters or enhancers not "naturally occurring," i.e., containing different elements of different transcriptional regulatory regions, and/or mutations that alter expression.
  • promoters that are most commonly used in recombinant DNA construction include the .beta.-lactamase (penicillinase), lactose and tryptophan (trp) promoter systems.
  • sequences may be produced using recombinant cloning and/or nucleic acid amplification technology, in connection with the compositions disclosed herein (see U.S. Pat. Nos. 4,683,202 and 5,928,906, each incorporated herein by reference).
  • control sequences that direct transcription and/or expression of sequences within non-nuclear organelles such as mitochondria, chloroplasts, and the like, can be employed as well.
  • nucleic acid molecules of the invention are not limited strictly to molecules including the sequences set forth as SEQ ID NO:1 and SEQ ID NO:3. Rather, specific embodiments encompasses nucleic acid molecules carrying modifications such as substitutions, small deletions, insertions, or inversions, which nevertheless encode proteins having substantially the biochemical activity of the Vasculostatin polypeptide according to the specific embodiments, and/or which can serve as hybridization probes for identifying a nucleic acid with one of the disclosed sequences.
  • nucleic acid molecules the nucleotide sequence of which is at least 95% identical (e.g., at least 96%, 97%, 98%, or 99% identical) to the nucleotide sequence shown as SEQ ID NO:1 , 3 and 5 in the Sequence Listing.
  • Gapped BLAST is utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25: 3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST. See http://www.ncbi.nlm.nih.gov.
  • stringent hybridization conditions is known in the art from standard protocols (e.g., Current Protocols in Molecular Biology, editors F. Ausubel et al., John Wiley and Sons, Inc. 1994) and is to be understood as conditions as stringent as those defined by the following: hybridization to filter-bound DNA in 0.5 M NaHPO. sub.4 (pH 7.2), 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at +65. degree. C, and washing in 0.1.times.SSC/0.1% SDS at +68.degree. C.
  • SDS sodium dodecyl sulfate
  • nucleic acid molecule that has a nucleotide sequence which is a degenerate variant of a nucleic acid disclosed herein, e.g., SEQ ID NO: 1.
  • the invention also includes an isolated polypeptide encoded by a nucleic acid of the invention.
  • An "isolated" polypeptide is a polypeptide that is substantially free from the proteins and other naturally occurring organic molecules with which it is naturally associated. Purity can be measured by any art-known method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC.
  • An isolated polypeptide may be obtained, for example, by extraction from a natural source (e.g., a human cell); by expression of a recombinant nucleic acid encoding the polypeptide; or by chemical synthesis of the polypeptide.
  • an isolated polypeptide includes recombinant polypeptides synthesized, for example, in vivo, e.g., in the milk of transgenic animals, or in vitro, e.g., in a mammalian cell line, in E. coli or another single-celled microorganism, or in insect cells.
  • polypeptide of the invention include an amino acid sequence as set forth in SEQ ID NO:2 and SEQ ID NO:4.
  • polypeptides of the exemplary embodiments are not to limited to those having an amino acid sequence identical to one of SEQ ID NO:2 and SEQ ID NO:4 in the Sequence Listing. Rather, the invention also encompasses conservative variants of the disclosed sequences.
  • Constant variants include substitutions within the following groups: glycine and alanine; valine, alanine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine, glutamine, serine, and threonine; lysine, arginine, and histidine; and phenylalanine and tyrosine.
  • polypeptides carrying modifications such as substitutions, small deletions, insertions, or inversions which polypeptides nevertheless have substantially the biological activities of the Vasculostatin polypeptide. Consequently, included in the invention is a polypeptide, the amino acid sequence of which is at least 95% identical (e.g., at least 96%, 97%, 98%, or 99% identical) to an amino acid sequence set forth as SEQ ID NO:2 and SEQ ID NO:4 in the Sequence Listing. "Percent identity" is defined in accordance with the algorithm described above.
  • polypeptides of the invention that have been post-translationally modified, e.g., by cleavage of an N-terminal signal sequence, which can be, e.g., 1 to 25 amino acids long.
  • Vasculostatin is anti-tumohqenic:
  • Vasculostatin a fragment of brain angiogenesis inhibitor 1 (BAM), which inhibits angiogenesis, tumor growth, and vascular permeability.
  • BAM brain angiogenesis inhibitor 1
  • the protein's brain specific expression along with its absence in a majority of human GBM specimens implies that loss of BAM during tumor progression may give the tumors a growth advantage.
  • Vasculostatin is a novel and potent inhibitor of angiogenesis tumor growth and vascular permeability.
  • HSVQuik methodology is a novel BAC-based method that utilizes two different site-specific recombination systems to introduce a transgene of interest into the deleted UL39 locus.
  • the fHsvQuik-1 is the BAC DNA with the incorporation of the entire HSV-1 genome lacking a functional ICP6 gene and deleted in both copies of the ⁇ 34.5 gene incorporated in it.
  • ICP34.5 allows the virus to replicate in non-dividing cells and dephosphorylates the cellular translation initiation factor (elF-2 ⁇ ) that is phosphorylated in response to activation of double-stranded RNA activated protein kinase (PKR) [74, 75]. These modifications allow the virus to replicate selectively in cancer cells. Additionally, fHsvQuik-1 has an insertion of a red fluorescent protein (RFP) in the middle of the BAC (bacterial artificial chromosome) backbone and thereby allows for efficient monitoring of the presence of BAC sequences in the vector genome [15]. [0093] Using this methodology, one OV embodiment was created and named rHSVQvasculo (also named "RAMBO," Rapid Anti-angiogenesis Mediated By Oncolysis virus). In that OV, a Vasculostatin transgene is driven by the viral immediate early IE4/5 promoter.
  • rHSVQvasculo also named "RAMBO," Rapid Anti-angiogenesis Mediated By Oncolysis virus.
  • rHSVQvasculo we used standard molecular biology approaches to first clone the cDNA encoding for Vasculostatin under IE4/5 promoter into a shuttle plasmid (ptransferlE4/5 which is published in Yamamoto et al, Gene Therapy 13, 1731-1736 (2006), incorporated herein by reference) to generate pVasculo- transfer.
  • the generated pVasculo-transfer plasmid is a replication-conditional plasmid (cannot replicate at 43 s C) in which the Vasculostatin gene is flanked by one loxP site and an FRT site.
  • the early viral promoter IE4/5 was selected to drive Vasculostatin expression because it has an early and robust expression profile in the context of an oncolytic herpes simplex virus [1].
  • the generated plasmid was verified by restriction digest analysis and confirmed by sequencing (not shown).
  • the Vasculostatin cassette along with the entire shuttle plasmid is inserted by Flp-mediated recombination into the disrupted ICP6 locus of the mutant HSV (Deleted for both copies of the ⁇ 34.5 gene) genome in the fHSVQuik-1 BAC DNA.
  • Flp-mediated recombination into the disrupted ICP6 locus of the mutant HSV (Deleted for both copies of the ⁇ 34.5 gene) genome in the fHSVQuik-1 BAC DNA.
  • pVasculo-transfer ampicillin [Amp] resistant
  • fHSVQuik-1 Chloramphenicol [Cm] resistant
  • pFTP-T Flp-expressing plasmid
  • the pCMVvasculo-transfer and pFTP-T cannot replicate at this temperature, and 80% of the Cm- and Amp-resistant recombinants have the correct recombination to generate fHSVQI - Vasculo.
  • the harvested BACs are analyzed by PCR and restriction analysis for integration of pVasculo.
  • the selected recombinant fHSVQI -Vasculo BAC is then transfected into Vero cells with a Cre-expressing helper plasmid.
  • the Cre-mediated recombination results in the excision of the bacterial plasmid sequences flanked by LoxP sites.
  • HSV recombinants generated by this process are easily identified because they express GFP, but not the RFP (excised by Cre-mediated recombination) in infected Vero cells.
  • the isolated recombinants are purified through subsequent plaque purifications or serial dilutions, and confirmed by further southern blot analysis.
  • the generated rHSVQvasculo from at least 2 isolates may be confirmed for correct insertion of IE4/5- Vasculostatin by Southern blot analysis. Briefly, viral DNA isolated from infected Vero cells was digested with Xhol, resolved by agarose gel electrophoresis, and transferred to nylon membranes. Probes specific for IE4/5-Vasculostatin were used to confirm its correct size and insertion [15]. The selected virus was confirmed for correct insertion and recombination events by sequencing both of the sites of recombination. [0097] Example 3
  • fHSVQ1vasulo1 ((fHSVQ1vasulo1 , and fHSVQ1vasulo2) were selected for further analysis.
  • the resulting six viral isolates were used to infect two different glioma cell lines (LN229, and U87 ⁇ EGFR) to evaluate Vasculostatin and viral ICP4 expression ( Figure 1 ).
  • the indicated glioma cells were infected with the six viruses, 3 isolated from fHSVQ1vasculo1 (lanes 1-3) and with 3 isolated from fHSVQ1vasulo2 (lanes 4-6).
  • Example 4 Initial characterization of two selected viral isolates: [00103] Referring to Fig. 5, from this initial screen we selected two viruses rHSVQvasculo 1 (lane 3), and rHSVQvasculo 2 (lane 5). We have purified both of these viruses. Since Vasculostatin expression in this recombinant virus is under the control of
  • ICP4 promoter we checked the temporal pattern of expression of Vasculostatin and ICP4 in
  • LN229 cells transfected with these viruses (Fig. 5). Briefly, LN229 glioma cells were transfected with the indicated viral isolate at an MOI of 0.1. Cells were harvested at the indicated times after infection and analyzed for expression of Vasculostatin and ICP4 by western blot analysis. Note the expression of both Vasculostatin and ICP4 come up as early as 4 hours after infection.
  • Vasculostatin does not affect the cytotoxicity of the recombinant oncolytic virus:
  • DIVAA assay confirms the in vivo anti-anqioqenic capability of an embodiment:
  • Fig. 8 shows a Kaplan-Meier survival analysis of mice treated with rHSVQ control virus or rHSVQvasculo (the virus generated to express Vasculostatin).
  • Mice with intracranial tumors U87 ⁇ EGFR
  • rHSVQ or the rHSVQvasculo were treated with a single dosage (1 x 10 5 pfu) of the control rHSVQ or the rHSVQvasculo at day 5 after tumor implantation.
  • all of the rHSVQ mice died of tumor burden by day 55.
  • there were 20% survivors in rHSVQvasculo treated animals. Survival of mice treated with rHSVQvasculo was significantly greater than that of mice treated with the rHSVQ virus
  • rHSVQvasculo has potent anti-tumor efficacy compared to the parent control oncolytic virus.
  • NHA multiplicities of infection
  • MOI multiplicities of infection
  • rHSVQ multiplicities of infection
  • rHSVQvasculo to evaluate potential cytotoxicity of OV produced Vasculostatin towards NHA.
  • MOI multiplicities of infection
  • rHSVQ rHSVQvasculo to evaluate potential cytotoxicity of OV produced Vasculostatin towards NHA.
  • NHA cells were plated into 96 well plates (10,000 cells/well). The cells were infected with the indicated virus at MOI of 1 , 0.5, 0.1 , 0.01 , and 0.05. Forty-eight hours post infection the number of viable cells measured by a standard Colorimetric crystal violet assay. Note no significant difference in the cytotoxicity to NHA at any of the indicated multiplicity of infection between rHSVQVasculo and rHSVQ. This indicated that rHSVQvasculo was as cytotoxic to NHA cells as rHSVQ.
  • Viral Replication of rHSVQvasculo is similar to rHSVQ control virus: [00120] Referring to Table 1 below, glioma cell lines: LN229, and U87 ⁇ EGFR were infected with rHSVQ and rHSVQvasculo at an MOI 0.05. Seventy-two hours post infection the cells and supernatants were harvested and the number of infectious viral particles (pfu) in each cell line was assessed by a standard viral titration assay. Table 1 below shows the results of viral titration in each indicated cell line. Note: The results indicate no significant difference in the replication ability of rHSVQvasculo compared to rHSVQ.
  • Cyclophosphamide (CPA) pretreatment further enhances the anti-tumor ability of Vasculostatin:
  • mice were treated with PBS or CPA (200 mg/kg) by intraperitoneal injection.
  • CPA/PBS 200 mg/kg
  • Two days after CPA/PBS treatment the animals were anesthetized and tumors were injected with 1x10 6 pfu rHSVQvasculo, or control rHSVQ.
  • Animals were sacrificed 48 hrs after OV treatment and the tumors were explanted sectioned into small pieces, and snap frozen. The tumors were lysed and equal amounts of lysate was then assayed for the presence of Vasculostatin by western blot analysis.
  • Vasculostatin in subcutaneous tumors U87 ⁇ EGFR glioma injected with rHSVQvasculo or control rHSVQ OV.
  • Positive control is cell lysate from LN229 cells infected with rHSVQvasculo, (MOI 0.05) for 48 hours.
  • Aghi, M.;Rabkin, S. D.Martuza, R. L Angiogenic response caused by oncolytic herpes simplex virus-induced reduced thrombospondin expression can be prevented by specific viral mutations or by administering a thrombospondin-derived peptide. Cancer Res 2007, 67, 440-4.
  • Herpes simplex virus type 1 -induced ribonucleotide reductase activity is dispensable for virus growth and DNA synthesis: Isolation and characterization of an icp ⁇ lacz insertion mutant. J Virol 1988, 62, 196-205.
  • Cyr61 is overexpressed in gliomas and involved in integhn-linked kinase-mediated akt and beta-catenin-tcf/lef signaling pathways. Cancer Res 2004, 64, 1987-96. 63. Xie, D.;Miller, C. W.;O'kelly, J.;Nakachi, K.;Sakashita, A.;Said, J. W.;Gornbein, J.Koeffler, H. P. Breast cancer. Cyr61 is overexpressed, estrogen-inducible, and associated with more advanced disease. J Biol Chem 2001, 276, 14187-94.
  • Brain-specific angiogenesis inhibitor 1 expression is inversely correlated with vascularity and distant metastasis of colorectal cancer, lnt J Oncol 1998, 13, 967-70.
  • Vascularization is decreased in pulmonary adenocarcinoma expressing brain-specific angiogenesis inhibitor 1 (bail), lnt J MoI Med 2000, 5, 181 -3.
  • Vasculostatin a proteolytic fragment of brain angiogenesis inhibitor 1 , is an antiangiogenic and antitumorigenic factor. Oncogene 2005, 24, 3632-42.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Mycology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Malignant tumors that are intrinsically resistant to conventional therapies are significant therapeutic challenges. An embodiment of the present invention provides an oncolytic virus capable of killing target cells, such as a tumor cells. In various embodiments presented herein, the oncolytic virus is armed or encodes a therapeutic polypeptide. In at least one embodiment, a recombinant oncolytic virus has been generated that can specifically replicate in cancer cells leading to their destruction and at the same time secrete robust amounts of an angiostatic factor to inhibit the regrowth of residual disease. Compositions and methods disclosed herein have broad therapeutic applicability.

Description

ONCOLYTIC VIRUS
CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the priority benefit of U.S. Patent Application No.
60/980,664, filed October 17, 2007, which is hereby incorporated by reference in its entirety.
TECHNICAL FIELD
[0002] The present invention is directed to the fields of virology, cancer biology, and medicine. More particularly, it concerns compositions and methods of treating cancer of the brain in a patient using oncolytic herpes simplex virus 1 (HSV-I ) armed with therapeutic transgenes.
BACKGROUND OF THE ART
[0003] Malignant tumors that are intrinsically resistant to conventional therapies are significant therapeutic challenges. Such malignant tumors include, but are not limited to malignant gliomas and recurrent systemic solid tumors such as lung cancer. Malignant gliomas are the most abundant primary brain tumors having an annual incidence of 6.4 cases per 100,000 (CBTRUS, 2002-2003). These neurologically devastating tumors are the most common subtype of primary brain tumors and are one of the deadliest human cancers. In the most aggressive cancer, manifestation glioblastoma multiforme (GBM), median survival duration for patients is 14 months, despite maximum treatment efforts (Stupp et al., 2005). A prototypic disease, malignant glioma is inherently resistant to current treatment regimens (Shapiro and Shapiro, 1998). In fact, in approximately 1/3 of patients with GBM the tumor will continue to grow despite treatment with radiation and chemotherapy. Median survival even with aggressive treatment including surgery, radiation, and chemotherapy is less than 1 year (Schiffer, 1998). Because few good treatment options are available for many of these refractory tumors, the exploration of novel and innovative therapeutic approaches is essential.
[0004] Gene therapy is a promising treatment for tumors including gliomas because conventional therapies typically fail and are toxic. In addition, the identification of genetic abnormalities contributing to malignancies is providing crucial molecular genetic information to aid in the design of gene therapies. Genetic abnormalities indicated in the progression of tumors include the inactivation of tumor suppressor genes and the overexpression of numerous growth factors and oncogenes. Tumor treatment may be accomplished by supplying a polynucleotide encoding a therapeutic polypeptide or other therapeutic that target the mutations and resultant aberrant physiologies of tumors. It is these mutations and aberrant physiology that distinguishes tumor cells from normal cells. A tumor-selective virus would be a promising tool for gene therapy. Recent advances in the knowledge of how viruses replicate have been used to design tumor-selective oncolytic viruses. [0005] In gliomas, several kinds of conditionally replication competent viruses have been shown to be useful in animal models for example: reoviruses that can replicate selectively in tumors with an activated ras pathway (Coffey et al., 1998); genetically altered herpes simplex viruses (Martuza et al., 1991 ; Mineta et al., 1995; Andreanski et al., 1997), including those that can be activated by the different expression of proteins in normal and cancer cells (Chase et al., 1998); and mutant adenoviruses that are unable to express the E1 B55 kDa protein and are used to treat p53-mutant tumors (Bischof et al., 1996; Heise et al., 1997; Freytag et al., 1998; Kim et al., 1998). Taken together, these reports confirm the relevance of oncolytic viruses as anti-cancer agents. In all three systems, the goal is the intratumoral spread of the virus and the ability to selectively kill cancer cells. Along with directly killing the cancers cells, agents that can also influence the microenvironment surrounding the tumor may enhance the therapeutic effect of the OV. [0006] Replication selective oncolytic viruses have shown great promise as antitumor agents for solid tumors. The viruses have been constructed genetically so that they are able to preferentially replicate within tumor cells, while being restricted in their ability to replicate in normal cells. The principal anti-tumor mechanism of oncolytic viruses is through a direct cytopathic effect as they propagate and spread from initially infected tumor cells to surrounding tumor cells, achieving a larger volume of distribution and anticancer effects. Oncolytic herpes simplex virus (HSV) were initially designed and constructed for the treatment of brain tumors (Martuza et al., 2007; Boviastsis et al., 1994). Subsequently, they have been found to be effective in a variety of other human solid tumors, including breast (Toda et al., 1998), prostate (Walker et al., 1999), lung (Toyoizumi et al., 1999), ovarian (Coukos et al., 1999), colon and liver cancers (Carroll et al., 1996; Pawlik et al., 2000). The safety of oncolytic HSVs has also been extensively tested in mice (Sundaresan et al., 2000) and primates (Aotus), which are extremely sensitive to HSV.
[0007] HSV-1 based oncolytic viruses are particularly exciting because of: 1 ) their ability to infect a wide variety of tumors; 2) their inherent cytolytic nature; 3) their well characterized large genome (152 Kb) that provides ample opportunity for genetic manipulations wherein many of the non-essential genes (up to 30kb) can be replaced by therapeutic genes; 4) their ability to remain as episomes that avoid insertional mutagenesis in infected cells; and 5) the availability of anti-herpetic drugs to keep in check possible undesirable replication [38, 39].
[0008] Despite these encouraging preclinical studies, results from early clinical trials have suggested that the current versions of oncolytic viruses, although safe, may only have limited anti-tumor activity on their own (Markert et al., 2000; Rampling et al., 2000; Nemunaitis et al., 2001 ). One of the main reasons for the sub-optimal oncolytic efficacy is probably because viral gene deletions that confer tumor selectivity also result in reduced potency of the virus in tumors. For example, the complete elimination of endogenous gamma.34.5 function from HSV, one of the common approaches for the construction of oncolytic HSV, significantly reduces viral replication potential and therefore may compromise the ability of the virus to spread within the targeted tumors (Kramm et al., 1997). Therefore, strategies designed to further enhance the potency of oncolytic viruses will likely increase their chance of clinical success.
[0009] The tumor microenvironment is also recognized as an important determinant for tumor progression. Vasculature is a major component of the microenvironment of solid tumors such as malignant gliomas. Solid tumors depend on the development of a vasculature to provide them with nutrients. While tumor oncolysis is thought to set the stage for activating a systemic adaptive immune surveillance, innate defense mechanisms elicited upon OV infection are thought to be responsible for rapid viral clearance from tumor [55-59]. Thus, OV-induced inflammation and its attendant increased vasculature may be counterproductive to the goal of killing cancer cells.
[0010] Therefore, there is an unmet need for an OV therapy that would reduce the area of vascularity and would thus limit OV induced inflammation and enhance the efficacy of the OV therapy.
SUMMARY OF THE INVENTION
[0011] The present embodiments address a long-felt need in the art by providing a potent oncolytic virus for therapy of undesirable cells, such as malignant cells. [0012] A preferred embodiment provides an oncolytic virus capable of killing target cells, such as a tumor cells. In preferred embodiments, the conditionally replicating HSV comprises at least two mechanisms to rid a culture, tissue or organism of at least some undesirable cells, to inhibit proliferation of at least some undesirable cells, to prevent proliferation of at least some desirable cells, or a combination thereof. A preferred embodiment may not only directly act on the cancer itself, but also may enhance tumor cell killing by influencing the microenvironment around the physical tumor. [0013] In various embodiments presented herein, the oncolytic virus is armed or encodes a therapeutic polypeptide. "Armed" is a term that indicates that the virus contains a heterologous nucleic acid sequence encoding a polypepitde of interest or a nucleic acid comprising a polynucleotide of interest. In certain embodiments, the nucleic acid encoding a therapeutic polypeptide may encode an angiostatic factor. Preferably, the nucleic acid encoding a therapeutic polypeptide encodes the polypeptide Vasculostatin. [0014] Oncolytic viruses expressing angiostatic factors using a CMV promoter have demonstrated limited efficacy in rodent models of glioma (for example, US Pub. No. 2006/0147420 and US Pub. No. 2004/0009604, incorporated herein by reference). Given the rapid lytic cycle of HSV, the challenge has been to ensure robust expression of the therapeutic protein before lysis of the infected cell.
[0015] Disclosed herein, a preferred embodiment overcomes this challenge by utilizing the immediate early HSV promoter IE4/5 operably linked to a therapeutic transgene. In at least one embodiment, the transgene is angiostatic. In a preferred embodiment, the transgene is a novel, brain specific angiostatic polypeptide, Vasculostatin. In a preferred embodiment, robust expression of a therapeutic gene can be seen at least as early as 4 hours.
[0016] The oncolytic virus of an exemplary embodiment can be delivered by a number routes including, but not limited to intracranial (into the skull cavity) intra- tumoral or intravenous administration. The tumor may be a primary tumor or it may be a tumor resulting from a metastasis to the skull or brain.
[0017] In an exemplary embodiment, a recombinant oncolytic virus has been generated that can specifically replicate in cancer cells leading to their destruction and at the same time secrete robust amounts of an angiostatic factor to inhibit the regrowth of residual disease. Such a dually armed OV destroys cancer cells through its tumor specific replication potential and also targets tumor vasculature to enhance therapeutic efficacy. [0018] In a preferred embodiment, a dually armed OV has been generated and shown that it does express and secrete the therapeutic anti-angiogenic factor (Vasculostatin), a novel brain specific anti-angiogenic factor, even at early time points after infection. In order to maximize the expression of Vasculostatin, at least one embodiment exploits the robust transgene expression from an early viral promoter to maximize the expression of Vasculostatin. The expression profile of a preferred embodiment allows for maximal expression of therapeutic transgenes before the lytic phase. At least one embodiment has shown efficacy in mice with established brain tumors. Compositions and methods disclosed herein have broad therapeutic applicability to most solid cancers. [0019] Expression of Vasculostatin does not interfere with the virus's cytotoxicity to glioma cells. The expressed Vasculostatin is functional. Additionally, the generated OV has therapeutic advantage over the control virus for the treatment of mice with established brain tumors.
BRIEF DESCRIPTION OF THE DRAWINGS
[0020] The following drawings form part of the present specification and are included to further demonstrate certain aspects of a preferred embodiment. [0021] The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein:
[0022] FIGURE 1 shows the survival of rats implanted with U87 human glioma cells stably expressing Vasculostatin (clones U14 and U 18) with parental untransfected glioma cells (U87). Note that survival of rats implanted with cells expressing Vasculostatin was significantly greater than that of rats implanted with control parental U87MG cells (P < 0.05). (Kaur et al., unpublished results).
[0023] FIGURE 2 provides a schematic illustration of the steps utilized to first clone the cDNA encoding for Vasculostatin under IE4/5 promoter into a shuttle plasmid (ptransferlE4/5) in order to generate pVasculo-transfer
[0024] FIGURE 3 provides a schematic illustration of the steps utilized to generate rHSVQvasculo (also called "RAMBO")
[0025] FIGURE 4 shows a western blot analysis confirming the expression of
Vasculostatin by the recombinant viral isolates.
[0026] FIGURE 5 shows a time course western analysis of Vasculostatin production by both OV isolates in LN229 cells
[0027] FIGURE 6 shows a cytotoxicity assay for both viral isolates with U87ΔEGFR cells. Note that there is no significant difference in cytotoxicity between the control rHSVQ virus and the isolated rHSVQvasculol , and rHSVQvasculo2 expressing Vasculostatin. [0028] FIGURE 7 demonstrates the anti-angiogenic capabilities of rHSVQvasculo.
The experiment was performed using the Trevigen Direct In Vivo Angiogenesis Assay (DIVAA™) Inhibition Kit.
[0029] FIGURE 8 shows a Kaplan-Meier survival analysis of mice treated with rHSVQ control virus or rHSVQvasculo (the virus generated to express Vasculostatin). In this experiment mice were treated by direct intratumoral injection on day 5 after tumor implantation.
[0030] FIGURE 9 shows the results of an experiment to compare the cytotoxicity of rHSVQvasculo to rHSVQ (an OV equivalent to the G207 being tested in clinical trials) toward normal human astrocytes. Normal human astrocytes (NHA, CellSciences Canton, MA) were infected at different multiolicities of infection (MOh with rHSVQ and rHSVQvasculo to evaluate potential cytotoxicity of OV produced Vasculostatin towards
NHA.
[0031] FIGURE 10 shows a comparison of the effect of Vasculostatin expression on ability of OV to be cytotoxic to glioma cells in the glioma cells LN229., U87ΔEGFR, and
U343, using a standard colorimetric assay. Note that there are no significant differences in the cytotoxicity for the two viruses.
[0032] FIGURE 11 is a western blot demonstrating that cyclophosphamide (CPA) pretreatment enhances the anti-tumor ability of Vasculostatin.
DETAILED DESCRIPTION OF A PREFERRED EMBODIMENT
[0033] Gliomas are the most common primary tumors of the central nervous system
(CNS). Glioblastoma multiforme (GBM), the most aggressive form (WHO grade IV) of malignant astrocytoma, is highly invasive and vascularized [40] and characterized by 1 ) rapidly proliferating endothelial cells that form tufted aggregates referred to as glomeruloid bodies and 2) multiple hypoxic-necrotic areas within the tumor that drive hypoxia-mediated activation of hypoxia inducible factor (HIF), which thereby leads to increased transcription of factors, such as vascular endothelial growth factor (VEGF), that heralds a phase of more malignant tumor growth [41]. Their very aggressive growth and highly vascular nature makes malignant gliomas an attractive target for testing the effects of anti-angiogenic gene therapy. The increased vascularization essential for malignant progression is triggered by disruption of the normal homeostasis between angiogenic and angiostatic factors within the tumor microenvironment [42]. It has been shown that expression of angiogenesis inhibitors is reduced in GBMs but not in normal brain and benign gliomas [43, 44]. Although not to be limited by theory, physiologically occurring factors that inhibit angiogenesis which are lost during tumor progression should represent molecules of choice for restoration by gene therapy.
[0034] Vasculostatin, a fragment of Brain Angiogenesis inhibitor 1 (BAM ) (GenBank
Accession No. AB005297), can inhibit angiogenesis, permeability, and subcutaneous as well as intracerebral tumor growth [45, 46]. Vasculostatin 1) is expressed at high levels primarily in normal brain but not in most GBMs and 2) has potent anti-angiogenic, anti- tumorigenic, and anti-permeability properties and 3) the ability to target multiple receptors on endothelial cells (αvβ3, αvβ5, and CD36), and 4) its over expression is well tolerated in brain tissue. Hence, Vasculostatin may be a better candidate than other more popular and not so novel anti-angiogenic factors for therapy of GBMs.
[0035] HSV-1 -derived OVs that express endostatin and, more recently, oncolytic viruses that express platelet factor IV and dominant negative FGF receptor under the control of a CMV promoter have been described [47-49]. However, at least one embodiment disclosed herein is better for GBM therapy because of its combination with an IE4/5 promoter (SEQ ID NO:5) that drives the expression of the transgene to levels unseen with a CMV promoter. More particularly, in certain embodiments disclosed herein Vasculostatin, a novel angiostatic factor, has been successfully expressed as part of an oncolytic viral stategy and shown to successfully counter anti-therapeutic changes in residual disease after oncolysis.
[0036] Specific replication within tumor cells can be achieved by OVs genetically engineered for that purpose or by naturally occurring strains of some viruses that have such propensity [25]. Specific embodiments utilize one such mutant, designated G207, which comprises an F-strain derived HSV-1 with deletions in both copies of the ^34.5 gene (encoding for the viral ICP34.5 protein) and an inactivating insertion of Escherichia coli (E. coli) lacZ into the viral ICP6/RR gene (encoding for the large subunit of ribonucleotide reductase). The available human evidence shows that the injection of HSV-1 with gamma34.5 deletion (and intact vhs) does not lead to the reactivation of wild-type HSV-1 , produce toxicity from infection of neurons surrounding the glioma cavity, or lead to encephalitis or meningitis.
[0037] Angiogenesis is critical for the development and maintenance of glioblastomas, the most malignant and common form of primary brain tumors. Combining oncolysis with anti-angiogenesis may produce a synergistic effect since the anti-cancer mechanisms are different but complementary. A preferred embodiment allows an anti- angiogenic nucleic acid or polypeptide, such as, but not limited to a Vasculostatin protein, to be produced, ultimately favoring delivery to the extracellular compartment. For that reason, the oncolytic HSV-1 is used as an improved HSV vector to deliver high and continuous levels of Vasculostatin to the tumor.
[0038] Angiogenesis refers to vessel formation by remodeling the primary vascular network or by sprouting from existing vessels (reviewed in Yancopoulos et al., 2000). The "angiogenesis switch" is "off" when the effect of pro-angiogenic molecules is balanced by the activity of anti-angiogenic molecules, and is "on" when the net balance between the molecules is tipped in favor of angiogenesis (reviewed in Carmeliet and Jain, 2000). Angiogenesis has an essential role in the development and maintenance of solid tumors, including malignant gliomas.
[0039] Embodiments of this invention may include other heterologous genes. For example, they may include therapeutic genes, pro-drug converting enzymes, cytosine deaminase (to convert 5-FC to 5-FU), a yeast cytosine deaminase, a humanized yeast cytosine deaminase, an image enhancing polypeptides, a sodium-iodide symporter, anti- sense or inhibitory VEGF, Bcl-2, Ang-2, or interferons alpha, beta or gamma. [0040] In describing the exemplary embodiments, the following terms will be employed, and are intended to be defined as indicated below.
[0041] The term "recombinant HSV-1 vector" as used herein defines a recombinant
HSV-1 vector comprising: (a) the DNA of, or corresponding to, at least a portion of the genome of an HSV-1 which portion is capable of transducing into a target cell at least one selected gene and is capable of promoting replication and packaging; and (b) at least one selected gene (or transgene) operatively linked to regulatory sequences directing its expression, the gene flanked by the DNA of (a) and capable of expression in the target cell in vivo or in vitro. Thus, when referring to a "recombinant HSV" (rHSV) it is meant the HSV that has been genetically altered, e.g., by the addition or insertion of a selected gene. [0042] A "gene" or a "sequence which encodes" a particular protein, is a nucleic acid molecule which is transcribed (in the case of DNA) and translated (in the case of mRNA) into a polypeptide in vitro or in vivo when placed under the control of appropriate regulatory sequences. The boundaries of the gene are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus. A gene can include, but is not limited to, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic DNA, and even synthetic DNA sequences. A transcription termination sequence will usually be located 3' to the gene sequence. Typically, polyadenylation signal is provided to terminate transcription of genes inserted into a recombinant virus.
[0043] As is known to those of skill in the art, the term "polypeptide" or "protein" means a linear polymer of amino acids joined in a specific sequence by peptide bonds. As used herein, the term "amino acid" refers to either the D or L stereoisomer form of the amino acid, unless otherwise specifically designated.
[0044] The term "transgene" refers to a particular nucleic acid sequence encoding a polypeptide or a portion of a polypeptide to be expressed in a cell into which the nucleic acid sequence is inserted. The term "transgene" is meant to include (1) a nucleic acid sequence that is not naturally found in the cell (i.e., a heterologous nucleic acid sequence); (2) a nucleic acid sequence that is a mutant form of a nucleic acid sequence naturally found in the cell into which it has been inserted; (3) a nucleic acid sequence that serves to add additional copies of the same (i.e., homologous) or a similar nucleic acid sequence naturally occurring in the cell into which it has been inserted; or (4) a silent naturally occurring or homologous nucleic acid sequence whose expression is induced in the cell into which it has been inserted. By "mutant form" is meant a nucleic acid sequence that contains one or more nucleotides that are different from the wild-type or naturally occurring sequence, i.e., the mutant nucleic acid sequence contains one or more nucleotide substitutions, deletions, and/or insertions. In some cases, the transgene may also include a sequence encoding a leader peptide or signal sequence such that the transgene product may be secreted from the cell.
[0045] In a preferred embodiment, a proteolytic fragment of BAM , Vasculostatin is utilized to as the anti-angiogenic transgene. A preferred embodiment comprises a polypeptide having the angiostatic activity of Vasculostatin, including, but not limited to the polypeptide of SEQ ID NO:2 that is encoded by the nucleic acid sequence of SEQ ID NO:1. Additionally, the transgene may optionally include nucleotides encoding a his and/or myc tag as in SEQ ID NO:3.
[0046] The promoter is preferably the immediate early promoter IE4/5 (SEQ ID
NO:5). The use of other viral or mammalian cellular or bacterial phage promoters which are well-known in the art to achieve expression of a coding sequence of interest is contemplated as well, provided that the levels of expression are sufficient for a given purpose.
[0047] A preferred embodiment provides a method for treating a neoplastic disease in a subject, the subject being an animal or human, comprising administering to the subject a therapeutically effective amount of a recombinant tumor-specific conditional replication oncolytic activity, the vector comprising a DNA sequence encoding an anti-angiogenic agent, the DNA is operably linked to a promoter. Preferably, the anti-angiogenic agent is vasculostatin (which is a fragment of brain angiogenesis inhibitor 1 (BAM )) or a biologically active variant thereof.
[0048] The term "operably linked" refers to the arrangement of various nucleic acid molecule elements relative to each other such that the elements are functionally connected and are able to interact with each other. Such elements may include, without limitation, a promoter, an enhancer, a polyadenylation sequence, one or more introns and/or exons, and a coding sequence of a gene of interest to be expressed (i.e., the transgene). The nucleic acid sequence elements, when operably linked, act together to modulate the activity of one another, and ultimately may affect the level of expression of the transgene. By modulate is meant increasing, decreasing, or maintaining the level of activity of a particular element. Typically, transduction of the transgene of the invention increases the expression of the transgene, preferably that of the angiostatic polypeptide Vasculostatin. The position of each element relative to other elements may be expressed in terms of the 5' terminus and the 3' terminus of each element.
[0049] The term "transfection" is used to refer to the uptake of foreign DNA by a mammalian cell. A cell has been "transfected" when exogenous DNA has been introduced inside the cell membrane. A number of transfection techniques are known in the art. See, Graham et al. (1973) Virology, 52:456; and Sambrook et al. (1989) Molecular Cloning, a laboratory manual, Cold Spring Harbor Laboratories, New York. Such techniques can be used to introduce one or more exogenous DNA moieties, such as a viral vector and other nucleic acid molecules, into suitable host cells. The term refers to both stable and transient uptake of the genetic material.
[0050] The vectors of the preferred embodiments may be useful for the introduction of additional genes in gene therapy. Thus, for example, the HSV vector of this invention can contain an additional exogenous gene for the expression of a protein effective in regulating the cell cycle, such as p53, Rb, or mitosin, or a biologically active variant thereof, or in inducing cell death, such as the conditional suicide gene thymidine kinase, the latter must be used in conjunction with a thymidine kinase metabolite in order to be effective, or any other anti-tumor gene, such as for example a toxin.
[0051] As used hereafter, the terms "neoplasm" and "neoplastic" refer to a tumor and/or to an abnormal tissue, including metastatic disease, that grows by cellular proliferation more rapidly than normal, continues to grow after the stimuli that initiated the new growth cease, shows partial or complete lack of structural organization and functional coordination with normal tissue, and usually forms a distinct mass of tissue which may be either benign or malignant.
[0052] A wide variety of neoplastic diseases can be treated by the same therapeutic strategy of exemplary embodiments. Neoplastic diseases include, but are not limited to, benign solid tumors, malignant solid tumors, benign proliferative diseases of the blood, and malignant proliferative diseases of the blood. Representative examples include colon carcinoma, prostate cancer, breast cancer, lung cancer, skin cancer, liver cancer, bone cancer, ovary cancer, pancreas cancer, brain cancer, head and neck cancer, and lymphoma.
[0053] As used throughout this application, the term animal is intended to be synonymous with mammal and is to include, but not be limited to, bovine, porcine, feline, simian, canine, equine, murine, rat or human. Host cells include, but are not limited to, any neoplastic or tumor cell, such as osteosarcoma, ovarian carcinoma, breast carcinoma, melanoma, hepatocarcinoma, lung cancer, brain cancer, colorectal cancer, hematopoietic cell, prostate cancer, cervical carcinoma, retinoblastoma, esophageal carcinoma, bladder cancer, neuroblastoma, or renal cancer.
[0054] When used pharmaceutically, OVs embodiments discussed herein can be combined with various pharmaceutically acceptable carriers. Suitable pharmaceutically acceptable carriers are well known to those of skill in the art. The compositions can then be administered therapeutically or prophylactically, in effective amounts, described in more detail below. [0055] As used herein, the term "therapeutically effective amount" is intended to mean the amount of vector or of transformed cells, which exerts oncolytic activity, causing attenuation or inhibition of tumor cell proliferation leading to tumor regression. An effective amount will vary on the pathology or condition to be treated, by the patient and his status, and other factors well known to those of skill in the art. Effective amounts are easily determined by those of skill in the art.
[0056] The term "oncolytic activity" as used herein refers to cytotoxic effects in vitro and/or in vivo exerted on tumor cells without any appreciable or significant deleterious effects to normal cells under the same conditions. The cytotoxic effects under in vitro conditions are detected by various means as known in prior art, for example, by staining with a selective stain for dead cells, by inhibition of DNA synthesis, or by apoptosis. Detection of the cytotoxic effects under in vivo conditions is performed by methods known in the art.
[0057] Methods of treating a neoplastic disease may include administration of the compounds of exemplary embodiments as a single active agent, or in combination with additional methods of treatment including, but not limited to, irradiation therapy, therapy with immunosuppressive agents, chemotherapeutic or anti-proliferative agents, including cytokines. The methods of treatment of the invention may be in parallel to, prior to, or following additional methods of treatment.
[0058] Any of the vectors described herein are useful for the treatment of a neoplastic disease. When used pharmaceutically, the vectors of the invention can be combined with one or more pharmaceutically acceptable carriers. Pharmaceutically acceptable carriers are well known in the art and include aqueous solutions such as physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, vegetable oils (e.g., olive oil) or injectable organic esters. A pharmaceutically acceptable carrier can be used to administer the compositions of the invention to a cell in vitro or to a subject in vivo.
[0059] A pharmaceutically acceptable carrier can contain a physiologically acceptable compound that acts, for example, to stabilize the composition or to increase the absorption of the agent. A physiologically acceptable compound can include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients. Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives, which are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid. One skilled in the art would know that the choice of a pharmaceutically acceptable carrier, including a physiologically acceptable compound, depends, for example, on the route of administration of the polypeptide. For example, a physiologically acceptable compound such as aluminum monosterate or gelatin is particularly useful as a delaying agent, which prolongs the rate of absorption of a pharmaceutical composition administered to a subject. Further examples of carriers, stabilizers or adjutants can be found in Martin, Remington's Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton, 1975), incorporated herein by reference.
[0060] As used herein, "pharmaceutical composition" or "composition" refers to any of the compositions of matter described herein. The compositions can then be administered therapeutically or prophylactically. They can be contacted with the host cell in vivo, ex vivo, or in vitro, in a therapeutically effective amount. In vitro and in vivo means of transfecting the vectors of the invention are provided below.
[0061] According to the invention, any suitable route of administration of the vectors may be adapted, including but not limited to, intravenous, oral, buccal, intranasal, inhalation, topical application to a mucosal membrane or injection, including intratumoral, intradermal, intrathecal, intracisternal, intralesional or any other type of injection. Administration can be effected continuously or intermittently and will vary with the subject and the condition to be treated.
[0062] An exemplary embodiment includes an oncolytic HSV, such as created by methods described herein, for example random mutagenesis, and further comprises a nucleic acid encoding an angiostatic polypeptide, such as Vasculostatin. [0063] Nucleic Acid-Based Expression Systems
[0064] An exemplary embodiment is directed to an HSV vector. In specific embodiments, the vector comprises some or all of the following components. [0065] The term "vector" is used to refers to a carrier nucleic acid molecule into which a nucleic acid sequence can be inserted for introduction into a cell where it can be replicated. A nucleic acid sequence can be "exogenous," which means that it is foreign to the cell into which the vector is being introduced or that the sequence is homologous to a sequence in the cell but in a position within the host cell nucleic acid in which the sequence is ordinarily not found. Vectors include plasmids, cosmids, viruses (bacteriophage, animal viruses, and plant viruses), and artificial chromosomes (e.g., YACs). One of skill in the art would be well equipped to construct a vector through standard recombinant techniques (see, for example, Maniatis et al., 1988 and Ausubel et al., 1994, both incorporated herein by reference).
[0066] The term "expression vector" refers to any type of genetic construct comprising a nucleic acid coding for a RNA capable of being transcribed. In some cases, RNA molecules are then translated into a protein, polypeptide, or peptide. In other cases, these sequences are not translated, for example, in the production of antisense molecules or ribozymes. Expression vectors can contain a variety of "control sequences," which refer to nucleic acid sequences necessary for the transcription and possibly translation of an operably linked coding sequence in a particular host cell. In addition to control sequences that govern transcription and translation, vectors and expression vectors may contain nucleic acid sequences that serve other functions as well and are described infra. [0067] Promoters and Enhancers
[0068] In exemplary embodiments, an HSV immediate early viral promoter is operably linked to the transgene in order to drive the expression of the heterologous transgene. More preferrably, the early viral promoter utilized is the HSV immediate early viral promoter IE4/5 (SEQ ID NO:5).
[0069] The term "promoter" refers to a nucleic acid sequence that regulates, either directly or indirectly, the transcription of a corresponding nucleic acid coding sequence to which it is operably linked. The promoter may function alone to regulate transcription, or, in some cases, may act in concert with one or more other regulatory sequences such as an enhancer or silencer to regulate transcription of the transgene. The promoter comprises a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene, which is capable of binding RNA polymerase and initiating transcription of a downstream (3'- direction) coding sequence.
[0070] A promoter generally comprises a sequence that functions to position the start site for RNA synthesis. The best-known example of this is the TATA box, but in some promoters lacking a TATA box, such as, for example, the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation. Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well. To bring a coding sequence "under the control of" a promoter, one positions the 5' end of the transcription initiation site of the transcriptional reading frame "downstream" of (i.e., 3' of) the chosen promoter. The "upstream" promoter stimulates transcription of the DNA and promotes expression of the encoded RNA.
[0071] The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription. A promoter may or may not be used in conjunction with an "enhancer," which refers to a cis-acting regulatory sequence involved in the transcriptional activation of a nucleic acid sequence.
[0072] A promoter may be one naturally associated with a nucleic acid sequence, as may be obtained by isolating the 5' non-coding sequences located upstream of the coding segment and/or exon. Such a promoter can be referred to as "endogenous." Similarly, an enhancer may be one naturally associated with a nucleic acid sequence, located either downstream or upstream of that sequence. Alternatively, certain advantages may be gained by positioning the coding nucleic acid segment under the control of a recombinant or heterologous promoter, which refers to a promoter that is not normally associated with a nucleic acid sequence in its natural environment. A recombinant or heterologous enhancer refers also to an enhancer not normally associated with a nucleic acid sequence in its natural environment. Such promoters or enhancers may include promoters or enhancers of other genes, and promoters or enhancers isolated from any other virus, or prokaryotic or eukaryotic cell, and promoters or enhancers not "naturally occurring," i.e., containing different elements of different transcriptional regulatory regions, and/or mutations that alter expression. For example, promoters that are most commonly used in recombinant DNA construction include the .beta.-lactamase (penicillinase), lactose and tryptophan (trp) promoter systems. In addition to producing nucleic acid sequences of promoters and enhancers synthetically, sequences may be produced using recombinant cloning and/or nucleic acid amplification technology, in connection with the compositions disclosed herein (see U.S. Pat. Nos. 4,683,202 and 5,928,906, each incorporated herein by reference). Furthermore, it is contemplated the control sequences that direct transcription and/or expression of sequences within non-nuclear organelles such as mitochondria, chloroplasts, and the like, can be employed as well.
[0073] The nucleic acid molecules of the invention are not limited strictly to molecules including the sequences set forth as SEQ ID NO:1 and SEQ ID NO:3. Rather, specific embodiments encompasses nucleic acid molecules carrying modifications such as substitutions, small deletions, insertions, or inversions, which nevertheless encode proteins having substantially the biochemical activity of the Vasculostatin polypeptide according to the specific embodiments, and/or which can serve as hybridization probes for identifying a nucleic acid with one of the disclosed sequences. Included in the invention are nucleic acid molecules, the nucleotide sequence of which is at least 95% identical (e.g., at least 96%, 97%, 98%, or 99% identical) to the nucleotide sequence shown as SEQ ID NO:1 , 3 and 5 in the Sequence Listing.
[0074] The determination of percent identity or homology between two sequences is accomplished using the algorithm of Karlin and Altschul (1990) Proc. Nat'l Acad. Sci. USA 87: 2264-2268, modified as in Karlin and Altschul (1993) Proc. Nat'l Acad. Sci. USA 90:5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al. (1990) J. MoI. Biol. 215:403-410. BLAST nucleotide searches are performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to the nucleic acid molecules of the invention. BLAST protein searches are performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to the protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST is utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25: 3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) are used. See http://www.ncbi.nlm.nih.gov.
[0075] The term "stringent hybridization conditions" is known in the art from standard protocols (e.g., Current Protocols in Molecular Biology, editors F. Ausubel et al., John Wiley and Sons, Inc. 1994) and is to be understood as conditions as stringent as those defined by the following: hybridization to filter-bound DNA in 0.5 M NaHPO. sub.4 (pH 7.2), 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at +65. degree. C, and washing in 0.1.times.SSC/0.1% SDS at +68.degree. C.
[0076] Also included in the invention is a nucleic acid molecule that has a nucleotide sequence which is a degenerate variant of a nucleic acid disclosed herein, e.g., SEQ ID NO: 1. A sequential grouping of three nucleotides, a "codon," encodes one amino acid. Since there are 64 possible codons, but only 20 natural amino acids, most amino acids are encoded by more than one codon. This natural "degeneracy" or "redundancy" of the genetic code is well known in the art. It will thus be appreciated that the nucleic acid sequences shown in the Sequence Listing provide only an example within a large but definite group of nucleic acid sequences that will encode the polypeptides as described above. [0077] The invention also includes an isolated polypeptide encoded by a nucleic acid of the invention. An "isolated" polypeptide is a polypeptide that is substantially free from the proteins and other naturally occurring organic molecules with which it is naturally associated. Purity can be measured by any art-known method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC. An isolated polypeptide may be obtained, for example, by extraction from a natural source (e.g., a human cell); by expression of a recombinant nucleic acid encoding the polypeptide; or by chemical synthesis of the polypeptide. In the context of a polypeptide obtained by extraction from a natural source, "substantially free" means that the polypeptide constitutes at least 60% (e.g., at least 75%, 90%, or 99%) of the dry weight of the preparation. A protein that is chemically synthesized, or produced from a source different from the source from which the protein naturally originates, is by definition substantially free from its naturally associated components. Thus, an isolated polypeptide includes recombinant polypeptides synthesized, for example, in vivo, e.g., in the milk of transgenic animals, or in vitro, e.g., in a mammalian cell line, in E. coli or another single-celled microorganism, or in insect cells. [0078] In various embodiments, the polypeptide of the invention include an amino acid sequence as set forth in SEQ ID NO:2 and SEQ ID NO:4. However, polypeptides of the exemplary embodiments are not to limited to those having an amino acid sequence identical to one of SEQ ID NO:2 and SEQ ID NO:4 in the Sequence Listing. Rather, the invention also encompasses conservative variants of the disclosed sequences. "Conservative variants" include substitutions within the following groups: glycine and alanine; valine, alanine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine, glutamine, serine, and threonine; lysine, arginine, and histidine; and phenylalanine and tyrosine.
[0079] Also included in the invention are polypeptides carrying modifications such as substitutions, small deletions, insertions, or inversions, which polypeptides nevertheless have substantially the biological activities of the Vasculostatin polypeptide. Consequently, included in the invention is a polypeptide, the amino acid sequence of which is at least 95% identical (e.g., at least 96%, 97%, 98%, or 99% identical) to an amino acid sequence set forth as SEQ ID NO:2 and SEQ ID NO:4 in the Sequence Listing. "Percent identity" is defined in accordance with the algorithm described above.
[0080] Also included in the invention are polypeptides of the invention that have been post-translationally modified, e.g., by cleavage of an N-terminal signal sequence, which can be, e.g., 1 to 25 amino acids long.
EXAMPLES
[0081] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those skilled in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventors to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents that are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims. [0082] Oncolytic HSV
[0083] Recent developments in the use of gene therapy vectors have utilized viral and nonviral vectors to transduce cancer or stem cells [18-20]. Oncolytic viral treatment exploits tumor-specific conditional replication of viruses to lyse tumor cells [21-23]. Genetic modifications of viral proteins to infect tumor cells specifically has been exploited to enhance tumor-specific viral tropism [24]. Specific replication within tumor cells can be achieved by OVs genetically engineered for that purpose or by naturally occurring strains of some viruses that have such propensity [25]. In a preferred embodiment, an HSV-1 derived OV deleted for both copies of the γ34.5 gene and additionally disrupted for the ICP6/RR genes that expresses Vasculostatin under the control of an early viral promoter, IE4/5, was constructed. [0084] Example 1
[0085] Vasculostatin is anti-tumohqenic:
[0086] We have recently discovered Vasculostatin, a fragment of brain angiogenesis inhibitor 1 (BAM), which inhibits angiogenesis, tumor growth, and vascular permeability. We have found that BAM is differentially expressed in normal and neoplastic brain. This is consistent with its reduced expression in pulmonary adenocarcinoma and pancreatic and gastric cancers, we and others have found it to be absent from brain tumors, but present in normal brain and benign gliomas (Kaur et al 2003, See appendix) [43, 44, 67-69]. The protein's brain specific expression along with its absence in a majority of human GBM specimens implies that loss of BAM during tumor progression may give the tumors a growth advantage.
[0087] Previous work has demonstrated that BAM is processed at a conserved GPS site through proteolysis, and this processing leads to the secretion of the extracellular domain of the protein. The cleaved extracellular domain yields a 120-kDa secreted fragment called Vasculostatin.
[0088] To investigate if Vasculostatin could inhibit the growth of intracranial tumors, we compared the survival of rats implanted with U87 human glioma cells stably expressing Vasculostatin (clones U14 and U 18) with parental untransfected glioma cells. Referring to FIG. 1 , 1 x 106 cells, derived from parental U87MG and Vasculo-expressing clones U14 and U18 were implanted stereotactically in the brains of athymic nude rats as described. Animals were carefully monitored for signs of necropsy and were sacrified according to our IACUC guidelines. Survival of rats implanted with cells expressing Vasulostatin was significantly greater than that of rats implanted with control parental U87MG cells (P < 0.05). [0089] Therefore, Vasculostatin is a novel and potent inhibitor of angiogenesis tumor growth and vascular permeability. However, because of the many variables involved with oncolytic viral expression, it was unknown whether or not it would make an effective therapeutic factor in the context of an oncolytic virus. Furthermore, it was unknown whether or not the presence of vasculostatin would interfere with the cytoxic ability of the virus.
[0090] Example 2
[0091] Creation of rHSVQvasculo:
[0092] In order to produce an oncolytic virus expressing vasculostatin HSVQuick methodology was employed[1]. The HSVQuik methodology is a novel BAC-based method that utilizes two different site-specific recombination systems to introduce a transgene of interest into the deleted UL39 locus. The fHsvQuik-1 is the BAC DNA with the incorporation of the entire HSV-1 genome lacking a functional ICP6 gene and deleted in both copies of the γ34.5 gene incorporated in it. ICP34.5 allows the virus to replicate in non-dividing cells and dephosphorylates the cellular translation initiation factor (elF-2α) that is phosphorylated in response to activation of double-stranded RNA activated protein kinase (PKR) [74, 75]. These modifications allow the virus to replicate selectively in cancer cells. Additionally, fHsvQuik-1 has an insertion of a red fluorescent protein (RFP) in the middle of the BAC (bacterial artificial chromosome) backbone and thereby allows for efficient monitoring of the presence of BAC sequences in the vector genome [15]. [0093] Using this methodology, one OV embodiment was created and named rHSVQvasculo (also named "RAMBO," Rapid Anti-angiogenesis Mediated By Oncolysis virus). In that OV, a Vasculostatin transgene is driven by the viral immediate early IE4/5 promoter.
[0094] Referring to FIG. 2, to construct rHSVQvasculo, we used standard molecular biology approaches to first clone the cDNA encoding for Vasculostatin under IE4/5 promoter into a shuttle plasmid (ptransferlE4/5 which is published in Yamamoto et al, Gene Therapy 13, 1731-1736 (2006), incorporated herein by reference) to generate pVasculo- transfer. The generated pVasculo-transfer plasmid is a replication-conditional plasmid (cannot replicate at 43s C) in which the Vasculostatin gene is flanked by one loxP site and an FRT site. The early viral promoter IE4/5 was selected to drive Vasculostatin expression because it has an early and robust expression profile in the context of an oncolytic herpes simplex virus [1]. The generated plasmid was verified by restriction digest analysis and confirmed by sequencing (not shown).
[0095] Referring to Fig. 3, the Vasculostatin cassette along with the entire shuttle plasmid is inserted by Flp-mediated recombination into the disrupted ICP6 locus of the mutant HSV (Deleted for both copies of the γ34.5 gene) genome in the fHSVQuik-1 BAC DNA. To accomplish this pVasculo-transfer (ampicillin [Amp] resistant), along with fHSVQuik-1 (Chloramphenicol [Cm] resistant) and an Flp-expressing plasmid (pFTP-T) is electroporated into bacteria carrying fHSVQuik-1 DNA and grown at 43s C. The pCMVvasculo-transfer and pFTP-T cannot replicate at this temperature, and 80% of the Cm- and Amp-resistant recombinants have the correct recombination to generate fHSVQI - Vasculo. The harvested BACs are analyzed by PCR and restriction analysis for integration of pVasculo.
[0096] Again referring to Fig. 3, the selected recombinant fHSVQI -Vasculo BAC is then transfected into Vero cells with a Cre-expressing helper plasmid. The Cre-mediated recombination results in the excision of the bacterial plasmid sequences flanked by LoxP sites. HSV recombinants generated by this process are easily identified because they express GFP, but not the RFP (excised by Cre-mediated recombination) in infected Vero cells. The isolated recombinants are purified through subsequent plaque purifications or serial dilutions, and confirmed by further southern blot analysis. The generated rHSVQvasculo from at least 2 isolates may be confirmed for correct insertion of IE4/5- Vasculostatin by Southern blot analysis. Briefly, viral DNA isolated from infected Vero cells was digested with Xhol, resolved by agarose gel electrophoresis, and transferred to nylon membranes. Probes specific for IE4/5-Vasculostatin were used to confirm its correct size and insertion [15]. The selected virus was confirmed for correct insertion and recombination events by sequencing both of the sites of recombination. [0097] Example 3
[0098] Confirmation of expression of Vasculostatin by the recombinant viral isolates:
[0099] Referring to Fig. 4, three viral isolates from each recombinant BAC
((fHSVQ1vasulo1 , and fHSVQ1vasulo2) were selected for further analysis. The resulting six viral isolates were used to infect two different glioma cell lines (LN229, and U87ΔEGFR) to evaluate Vasculostatin and viral ICP4 expression (Figure 1 ). The indicated glioma cells were infected with the six viruses, 3 isolated from fHSVQ1vasculo1 (lanes 1-3) and with 3 isolated from fHSVQ1vasulo2 (lanes 4-6).
[00100] The infected cells and conditioned media were harvested 10 hours post infection and the cell lysate and TCA precipitated conditioned media was analyzed for Vasculostatin protein expression by western blot analysis. Note the presence of Vasculostatin in infected cell lysate and harvested conditioned media from infected cells but not in the control rHSVQ infected cells (bottom panel). [00101] Example 4 [00102] Initial characterization of two selected viral isolates: [00103] Referring to Fig. 5, from this initial screen we selected two viruses rHSVQvasculo 1 (lane 3), and rHSVQvasculo 2 (lane 5). We have purified both of these viruses. Since Vasculostatin expression in this recombinant virus is under the control of
ICP4 promoter we checked the temporal pattern of expression of Vasculostatin and ICP4 in
LN229 cells transfected with these viruses (Fig. 5). Briefly, LN229 glioma cells were transfected with the indicated viral isolate at an MOI of 0.1. Cells were harvested at the indicated times after infection and analyzed for expression of Vasculostatin and ICP4 by western blot analysis. Note the expression of both Vasculostatin and ICP4 come up as early as 4 hours after infection.
[00104] These results confirm that a recombinant oncolytic HSV which also expresses vasculostatin was generated.
[00105] Example 5
[00106] Vasculostatin does not affect the cytotoxicity of the recombinant oncolytic virus:
[00107] Referring to Fig. 6, to compare the effect of Vasculostatin expression on oncolytic virus replication we compared the cytotoxixcity of the control rHSVQ virus with the
2 selected viral isolates rHSVQvasculol , and rHSVQvasculo2. Six thousand U87ΔEGFR glioma cells were infected with the indicated virus at MOI of 1 , 0.5, 0.1 , 0.01 , and 0.05, on day zero. The number of viable cells was measured by a standard crystal violet assay on day 1 , day, 2, day, 3 and day 5. Briefly, the cells at the indicated time point were fixed with
1% glutaraldehyde for 15 minutes and then stained with 5% crystal violet (dissolved in
4.75% ethanol) for 15 minutes. The plates were washed to remove unbound stain and the crystal violet crystals were dissolved in Sorensen's buffer prior to reading absorbance read at 590nm. Note that there is no significant difference in cytotoxicity between the control rHSVQ virus and the isolated rHSVQvasculol , and rHSVQvasculo2 expressing
Vasculostatin.
[00108] Example 6
[00109] DIVAA assay confirms the in vivo anti-anqioqenic capability of an embodiment:
[00110] Referring to Fig. 7, the anti-angiogenic capabilities of rHSVQvasculo was tested using the Trevigen Direct In Vivo Angiogenesis Assay (DIVAA™) Inhibition Kit (Cat
#: 3450-048-IK). Briefly, 2.5x105 U87ΔEGFR cells treated with PBS or infected with rHSVQ, rHSVQvasculo was mixed with basement membrane. The samples were then pipette into angioreactors and allowed to polymerize for 1 hour at 370C. The tubes were then implanted into the rear flanks of nu/nu mice and 12 days later the mice were sacrifice and angioreactors removed. The amount of angiogenesis that occurred in the angioreactors was quantified using the Wako Hemoglobin B kit. Angiogenesis is initiated into the tubes from the one open end in the tubes. Note both visually and graphically the significant reduction in angiogenesis for samples infected with RAMBO compared to rHSVQ
(n=10/group, and p=0.07) and PBS control (Figure 6).
[00111] Example 7
[00112] The Oncolytic Virus rHSVQvasculo is therapeutically effective:
[00113] We have tested the therapeutic potential of rHSVQvasculo in human glioma cells grown in a mouse brain tumor model. U87ΔEGFR human glioma cells were implanted intracranial^ in mice. Later, Mice were treated with direct intratumoral injection of rHSVQ or rHSVQvasculo. Animals were carefully monitored for any signs of morbidity and were sacrificed in accordance with our IACUC guidelines.
[00114] Fig. 8 shows a Kaplan-Meier survival analysis of mice treated with rHSVQ control virus or rHSVQvasculo (the virus generated to express Vasculostatin). Briefly, Mice with intracranial tumors (U87ΔEGFR) were treated with a single dosage (1 x 105 pfu) of the control rHSVQ or the rHSVQvasculo at day 5 after tumor implantation. As can be observed from the figure, all of the rHSVQ mice died of tumor burden by day 55. However, there were 20% survivors in rHSVQvasculo treated animals. Survival of mice treated with rHSVQvasculo was significantly greater than that of mice treated with the rHSVQ virus
(P=O.0246). Hence, rHSVQvasculo has potent anti-tumor efficacy compared to the parent control oncolytic virus.
[00115] Example 8
[00116] Comparison of rHSVQvasculo and rHSVQ mediated cytotoxicity to normal human astrocytes (NHA):
[00117] Referring to Fig. 9, to evaluate the cytotoxicity of rHSVQvasculo produced by the recombinant OV, we infected normal human astrocytes (NHA, CellSciences Canton,
MA) at different multiplicities of infection (MOI) with rHSVQ and rHSVQvasculo to evaluate potential cytotoxicity of OV produced Vasculostatin towards NHA. Briefly NHA: cells were plated into 96 well plates (10,000 cells/well). The cells were infected with the indicated virus at MOI of 1 , 0.5, 0.1 , 0.01 , and 0.05. Forty-eight hours post infection the number of viable cells measured by a standard Colorimetric crystal violet assay. Note no significant difference in the cytotoxicity to NHA at any of the indicated multiplicity of infection between rHSVQVasculo and rHSVQ. This indicated that rHSVQvasculo was as cytotoxic to NHA cells as rHSVQ.
[00118] Example 9
[00119] Viral Replication of rHSVQvasculo is similar to rHSVQ control virus: [00120] Referring to Table 1 below, glioma cell lines: LN229, and U87ΔEGFR were infected with rHSVQ and rHSVQvasculo at an MOI 0.05. Seventy-two hours post infection the cells and supernatants were harvested and the number of infectious viral particles (pfu) in each cell line was assessed by a standard viral titration assay. Table 1 below shows the results of viral titration in each indicated cell line. Note: The results indicate no significant difference in the replication ability of rHSVQvasculo compared to rHSVQ.
TABLE 1
Figure imgf000024_0001
[00121] Example 10
[00122] Cellular cytotoxicity results in multiple glioma cell lines demonstrating the ability of the rHSVQvasculo to be cytotoxic to multiple glioma cells in vitro:
[00123] Referring to Fig. 10, the effect of Vasculostatin expression on ability of OV to be cytotoxic to glioma cells was co mpared in the glioma cells LN229., U87ΔEGFR, and
U343, using a standard colorimetric assay. All cell lines were infected with control rHSVQ virus or rHSVQvasculo at indicated MOIs (1 , 0.5, 0.1 , 0.01 , or 0.05). The number of viable cells was measured by a standard Colorimetric crystal violet assay on day 1 , day, 2, day, 3 and day5. Note that there are no significant differences in the cytotoxicity for the two viruses.
[00124] Example 11
[00125] Cyclophosphamide (CPA) pretreatment further enhances the anti-tumor ability of Vasculostatin:
[00126] Referring to Fig. 11 , Athymic nude mice were injected with 2.5x106
U87ΔEGFR cells. Seventeen days later, when the tumors were of sufficient size
(>750mm3), the mice were treated with PBS or CPA (200 mg/kg) by intraperitoneal injection. Two days after CPA/PBS treatment the animals were anesthetized and tumors were injected with 1x106 pfu rHSVQvasculo, or control rHSVQ. Animals were sacrificed 48 hrs after OV treatment and the tumors were explanted sectioned into small pieces, and snap frozen. The tumors were lysed and equal amounts of lysate was then assayed for the presence of Vasculostatin by western blot analysis. Western blot analysis for expression of
Vasculostatin in subcutaneous tumors (U87ΔEGFR glioma) injected with rHSVQvasculo or control rHSVQ OV. Positive control is cell lysate from LN229 cells infected with rHSVQvasculo, (MOI 0.05) for 48 hours. Note the presence of vasculostatin in the rHSVQvasculo treated tumors indicating the ability of rHSVQvasculo to express Vasculostatin in vivo. Note also the increase in Vasculostatin expression in the CPA treated animals.
[00127] Publications
[00128] The following references and others cited herein but not listed here, to the extent that they provide exemplary procedural and other details supplementary to those set forth herein, are specifically incorporated herein by reference.
1. Terada, K.;Wakimoto, H.;Tyminski, E.;Chiocca, E. A.Saeki, Y. Development of a rapid method to generate multiple oncolytic hsv vectors and their in vivo evaluation using syngeneic mouse tumor models. Gene Ther 2006, 13, 705-14.
2. Yang, G. P.Lau, L. F. Cyr61 , product of a growth factor-inducible immediate early gene, is associated with the extracellular matrix and the cell surface. Cell Growth Differ 1991, 2, 351-7.
3. Pendurthi, U. R.;Tran, T. T.;Post, M.Rao, L. V. Proteolysis of ccn1 by plasmin: Functional implications. Cancer Res 2005, 65, 9705-11.
4. Jackson, J. R.;Seed, M. P.;Kircher, C. H.;Willoughby, D. A.Winkler, J. D. The codependence of angiogenesis and chronic inflammation. Faseb J 1997, 11, 457- 65.
5. Kunstfeld, R.;Hirakawa, S.;Hong, Y. K.;Schacht, V.;l_ange-Asschenfeldt, B.;Velasco, P.;ϋn, C.;Fiebiger, E.;Wei, X.;Wu, Y.;Hicklin, D.;Bohlen, P.Detmar, M. Induction of cutaneous delayed-type hypersensitivity reactions in vegf-a transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia. Blood 2004, 104, 1048-57.
6. Lange-Asschenfeldt, B.;Weninger, W.;Velasco, P.;Kyriakides, T. R.;Von Andrian, U. H.;Bornstein, P.Detmar, M. Increased and prolonged inflammation and angiogenesis in delayed-type hypersensitivity reactions elicited in the skin of thrombospondin-2-- deficient mice. Blood 2002, 99, 538-45.
7. Pike, S. E.;Yao, L.;Jones, K. D.;Cherney, B.;Appella, E.;Sakaguchi, K.;Nakhasi, H.;Teruya-Feldstein, J.;Wirth, P.;Gupta, G.Tosato, G. Vasostatin, a calreticulin fragment, inhibits angiogenesis and suppresses tumor growth. J Exp Med 1998, 188, 2349-56.
8. Park, Y. W.;Kang, Y. M.;Butterfield, J.;Detmar, M.;Goronzy, J. J.Weyand, C. M. Thrombospondin 2 functions as an endogenous regulator of angiogenesis and inflammation in rheumatoid arthritis. Am J Pathol 2004, 165, 2087-98.
9. Babic, A. M.;Kireeva, M. L;Kolesnikova, T. V.Lau, L. F. Cyr61 , a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc Natl Acad Sci U S A 1998, 95, 6355-60. 10. Koh, J. T.;Kook, H.;Kee, H. J.;Seo, Y. W.;Jeong, B. C.;Lee, J. H.;Kim, M. Y.;Yoon, K. C.;Jung, S.Kim, K. K. Extracellular fragment of brain-specific angiogenesis inhibitor 1 suppresses endothelial cell proliferation by blocking alphavbetaδ integhn. Exp Cell Res 2004, 294, 172-84.
11. Heliums, E. K.;Markert, J. M.;Parker, J. N.;He, B.;Perbal, B.;Roizman, B.;Whitley, R. J.;Langford, C. P.;Bharara, S.Gillespie, G. Y. Increased efficacy of an interleukin-12- secreting herpes simplex virus in a syngeneic intracranial murine glioma model. Neuro-oncol 2005, 7, 213-24.
12. Chiocca, E. A. Oncolytic viruses. Nat Rev Cancer 2002, 2, 938-50.
13. Wakimoto, H.;Fulci, G.;Tyminski, E. Chiocca, E. A. Altered expression of antiviral cytokine mrnas associated with cyclophosphamide's enhancement of viral oncolysis. Gene Ther2004, 11, 214-23.
14. Aghi, M.Martuza, R. L. Oncolytic viral therapies - the clinical experience. Oncogene 2005, 24, 7802^6.
15. Kambara, H.;Okano, H.;Chiocca, E. A.Saeki, Y. An oncolytic hsv-1 mutant expressing icp34.5 under control of a nestin promoter increases survival of animals even when symptomatic from a brain tumor. Cancer Res 2005, 65, 2832-9.
16. Ikeda, K.;lchikawa, T.;Wakimoto, H.;Silver, J. S.;Deisboeck, T. S.;Finkelstein, D.;Harsh, G. R. T.;Louis, D. N.;Bartus, R. T.;Hochberg, F. H. Chiocca, E. A. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med 1999, 5, 881 -7.
17. Ikeda, K.;Wakimoto, H.;lchikawa, T.;Jhung, S.;Hochberg, F. H.;Louis, D. N. Chiocca, E. A. Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant. J Virol 2000, 74, 4765-75.
18. Li, S. Electroporation gene therapy: New developments in vivo and in vitro. Current gene therapy 2004, 4, 309-316.
19. Kurre, P.;Anandakumar, P.;Harkey, M. A.;Thomasson, B.Kiem, H. P. Efficient marking of murine long-term repopulating stem cells targeting unseparated marrow cells at low lentiviral vector particle concentration. MoI Ther 2004, 9, 914-22.
20. Seth, P. Vector-mediated cancer gene therapy: An overview. Cancer Biology Therapy 2005, 4, 512-517.
21. Liu, T. C. Kirn, D. Viruses with deletions in antiapoptotic genes as potential oncolytic agents. Oncogene 2005, 24, 6069-79. 22. Liu, T. C.;Wang, Y.;Hallden, G.;Brooks, G.;Francis, J.;Lemoine, N. R.Kirn, D. Functional interactions of antiapoptotic proteins and tumor necrosis factor in the context of a replication-competent adenovirus. Gene Ther2005, 12, 1333-46.
23. Conrad, C.;Miller, C. R.;Ji, Y.;Gomez-Manzano, C.;Bharara, S.;Mcmurray, J. S.;l_ang, F. F.;Wong, F.;Sawaya, R.;Yung, W. K.Fueyo, J. Delta24-hycd adenovirus suppresses glioma growth in vivo by combining oncolysis and chemosensitization. Cancer Gene Ther 2005, 12, 284-94.
24. Mathis, J. M.;Stoff-Khalili, M. A.Curiel, D. T. Oncolytic adenoviruses - selective retargeting to tumor cells. Oncogene 2005, 24, 7775-91.
25. Mohr, I. To replicate or not to replicate: Achieving selective oncolytic virus replication in cancer cells through translational control. Oncogene 2005, 24, 7697-709.
26. Markert, J. M.;Medlock, M. D.;Rabkin, S. D.;Gillespie, G. Y.;Todo, T.;Hunter, W. D.;Palmer, C. A.;Feigenbaum, F.;Tornatore, C.;Tufaro, F.Martuza, R. L. Conditionally replicating herpes simplex virus mutant, g207 for the treatment of malignant glioma: Results of a phase i trial. Gene Ther 2000, 7, 867-74.
27. Rampling, R.;Cruickshank, G.;Papanastassiou, V.;Nicoll, J.;Hadley, D.;Brennan, D.;Petty, R.; Maclean, A.;Harland, J.;Mckie, E.;Mabbs, R. Brown, M. Toxicity evaluation of replication-competent herpes simplex virus (icp 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther 2000, 7, 859-66.
28. Harrow, S.;Papanastassiou, V.;Harland, J.;Mabbs, R.;Petty, R.;Fraser, M.;Hadley, D.;Patterson, J.;Brown, S. M. Rampling, R. Hsv1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: Safety data and long- term survival. Gene Ther 2004, 11, 1648-58.
29. Chiocca, E. A.;Abbed, K. M.;Tatter, S.;Louis, D. N.;Hochberg, F. H.;Barker, F.;Kracher, J.;Grossman, S. A.;Fisher, J. D.;Carson, K.;Rosenblum, M.;Mikkelsen, T.;Olson, J.;Markert, J.;Rosenfeld, S.;Nabors, L. B.;Brem, S.;Phuphanich, S.;Freeman, S.;Kaplan, R.Zwiebel, J. A phase i open-label, dose-escalation, multi- institutional trial of injection with an e1 b-attenuated adenovirus, onyx-015, into the peritumoral region of recurrent malignant gliomas, in the adjuvant setting. MoI Ther 2004, 10, 958-66.
30. Hallak, L. K.;Merchan, J. R.;Storgard, C. M.;Loftus, J. C.Russell, S. J. Targeted measles virus vector displaying echistatin infects endothelial cells via alpha(v)beta3 and leads to tumor regression. Cancer Res 2005, 65, 5292-300.
31. Hedley, S. J.;Auf Der Maur, A.;Hohn, S.;Escher, D.;Barbehs, A.;Glasgow, J. N.;Douglas, J. T.;Korokhov, N.Curiel, D. T. An adenovirus vector with a chimeric fiber incorporating stabilized single chain antibody achieves targeted gene delivery. Gene Ther 2005.
32. Borovjagin, A. V.;Krendelchtchikov, A.;Ramesh, N.;Yu, D. C.;Douglas, J. T.Curiel, D. T. Complex mosaicism is a novel approach to infectivity enhancement of adenovirus type 5-based vectors. Cancer Gene Ther 2005, 12, 475-86.
33. Nakano, K.;Asano, R.;Tsumoto, K.;Kwon, H.;Goins, W. F.;Kumagai, l.;Cohen, J. B.GIorioso, J. C. Herpes simplex virus targeting to the egf receptor by a gd-specific soluble bridging molecule. MoI Ther 2005, 11, 617-26.
34. Biglari, A.;Bataille, D.;Naumann, U.;Weller, M.;Zirger, J.;Castro, M. G.Lowenstein, P. R. Effects of ectopic decorin in modulating intracranial glioma progression in vivo, in a rat syngeneic model. Cancer Gene Ther 2004, 11, 721-32.
35. Mahendra, G.;Kumar, S.;lsayeva, T.;Mahasreshti, P. J.;Curiel, D. T.;Stockardt, C. R.;Gιϊzzle, W. E.;Alapati, V.;Singh, R.;Siegal, G. P.;Meleth, S.Ponnazhagan, S. Antiangiogenic cancer gene therapy by adeno-associated virus 2-mediated stable expression of the soluble fms-like tyrosine kinase-1 receptor. Cancer Gene Ther 2005, 12, 26-34.
36. Kikuchi, E.;Menendez, S.;Ohori, M.;Cordon-Cardo, C.;Kasahara, N.Bochner, B. H. Inhibition of orthotopic human bladder tumor growth by lentiviral gene transfer of endostatin. Clin Cancer Res 2004, 10, 1835-42.
37. Finger, C.;Sun, Y.;Sanz, L;Alvarez-Vallina, L;Buchholz, C. J.Cichutek, K. Replicating retroviral vectors mediating continuous production and secretion of therapeutic gene products from cancer cells. Cancer Gene Ther 2005, 12, 464-74.
38. Lawler, S. E.;Peruzzi, P. P.Chiocca, E. A. Genetic strategies for brain tumor therapy. Cancer Gene Ther 2005.
39. Varghese, S.Rabkin, S. D. Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther 2002, 9, 967-78.
40. Gillespie, G. Y.;Soroceanu, L;Manning, T. J., Jr.;Gladson, C. L.Rosenfeld, S. S. Glioma migration can be blocked by nontoxic inhibitors of myosin ii. Cancer Res 1999, 59, 2076-82.
41. Brat, D. J.;Kaur, B. Van Meir, E. G. Genetic modulation of hypoxia induced gene expression and angiogenesis: Relevance to brain tumors. Front Biosci 2003, 8, D100-16.
42. Nyberg, P.;Xie, L.Kalluh, R. Endogenous inhibitors of angiogenesis. Cancer Res 2005, 65, 3967-79. 43. Kaur, B.;Brat, D. J.;Calkins, C. C. Van Meir, E. G. Brain angiogenesis inhibitor 1 is differentially expressed in normal brain and glioblastoma independently of p53 expression. Am J Pathol 2003, 162, 19-27.
44. Nam, D. H.;Park, K.;Suh, Y. L.Kim, J. H. Expression of vegf and brain specific angiogenesis inhibitor-1 in glioblastoma: Prognostic significance. Oncol Rep 2004, 11, 863-9.
45. Kaur, B.;Brat, D. J.;Devi, N. S. Van Meir, E. G. Vasculostatin, a proteolytic fragment of brain angiogenesis inhibitor 1 , is an antiangiogenic and antitumorigenic factor. Oncogene 2005.
46. Kaur, B.;Sandberg, E.;Khwaja, F.;Brat, D. J.;Devi, N. S.;Olson, J. J.;Zhang, Z. Van Meir, E. G. Vasculostatin, a 120kda bail fragment can efficiently inhibit intracranial angiogenesis and tumorigenesis, despite a proangiogenic stimulus. In preparation unpublished results.
47. Mullen, J. T.;Donahue, J. M.;Chandrasekhar, S.;Yoon, S. S.;Liu, W.;Ellis, L. M.;Nakamura, H.;Kasuya, H.;Pawlik, T. M.Tanabe, K. K. Oncolysis by viral replication and inhibition of angiogenesis by a replication-conditional herpes simplex virus that expresses mouse endostatin. Cancer 2004, 101, 869-77.
48. Liu, T. C.;Zhang, T.;Fukuhara, H.;Kuroda, T.;Todo, T.;Canron, X.;Bikfalvi, A.;Martuza, R. L.;Kurtz, A.Rabkin, S. D. Dominant-negative fibroblast growth factor receptor expression enhances antitumoral potency of oncolytic herpes simplex virus in neural tumors. Clin Cancer Res 2006, 12, 6791-9.
49. Liu, T. C.;Zhang, T.;Fukuhara, H.;Kuroda, T.;Todo, T.;Martuza, R. L.;Rabkin, S. D.Kurtz, A. Oncolytic hsv armed with platelet factor 4, an antiangiogenic agent, shows enhanced efficacy. MoI Ther 2006, 14, 789-97.
50. Aghi, M.;Rabkin, S. D.Martuza, R. L Angiogenic response caused by oncolytic herpes simplex virus-induced reduced thrombospondin expression can be prevented by specific viral mutations or by administering a thrombospondin-derived peptide. Cancer Res 2007, 67, 440-4.
51. Nishimoh, H.;Shiratsuchi, T.;Urano, T.;Kimura, Y.;Kiyono, K.;Tatsumi, K.;Yoshida, S.;Ono, M.;Kuwano, M.;Nakamura, Y.Tokino, T. A novel brain-specific p53-target gene, bail , containing thrombospondin type 1 repeats inhibits experimental angiogenesis. Oncogene 1997, 15, 2145-50.
52. Simpson, S. A.;Manchak, M. D.;Hager, E. J.;Krummenacher, C.;Whitbeck, J. C.;Levin, M. J.;Freed, C. R.;Wilcox, C. L.;Cohen, G. H.;Eisenberg, R. J.Pizer, L I. Nectin-1/hvec mediates herpes simplex virus type 1 entry into primary human sensory neurons and fibroblasts. J Neurovirol 2005, 11, 208-18. 53. Goldstein, D. J.Weller, S. K. Herpes simplex virus type 1 -induced ribonucleotide reductase activity is dispensable for virus growth and DNA synthesis: Isolation and characterization of an icpθ lacz insertion mutant. J Virol 1988, 62, 196-205.
54. Oura, H.;Bertoncini, J.;Velasco, P.;Brown, L. F.;Carmeliet, P.Detmar, M. A critical role of placental growth factor in the induction of inflammation and edema formation. Blood 2003, 101, 560-7.
55. Abordo-Adesida, E.;Follenzi, A.;Barcia, C.;Sciascia, S.;Castro, M. G.;Naldini, L.Lowenstein, P. R. Stability of lentiviral vector-mediated transgene expression in the brain in the presence of systemic antivector immune responses. Hum Gene Ther
2005, 76, 741-51.
56. Balachandran, S. Barber, G. N. Defective translational control facilitates vesicular stomatitis virus oncolysis. Cancer Cell 2004, 5, 51 -65.
57. Andreansky, S.;He, B.;Van Cott, J.;Mcghee, J.;Markert, J. M.;Gillespie, G. Y.;Roizman, B.Whitley, R. J. Treatment of intracranial gliomas in immunocompetent mice using herpes simplex viruses that express murine interleukins. Gene Ther 1998, 5, 121-30.
58. Bennett, J. J.;Malhotra, S.;Wong, R. J.;Delman, K.;Zager, J.;St-Louis, M.;Johnson, P.Fong, Y. lnterleukin 12 secretion enhances antitumor efficacy of oncolytic herpes simplex viral therapy for colorectal cancer. Ann Surg 2001, 233, 819-26.
59. Todo, T.;Martuza, R. L;Dallman, M. J.Rabkin, S. D. In situ expression of soluble b7- 1 in the context of oncolytic herpes simplex virus induces potent antitumor immunity. Cancer Res 2001, 61, 153-61.
60. Lamfers, M. L;Fulci, G.;Gianni, D.;Tang, Y.;Kurozumi, K.;Kaur, B.;Moeniralm, S.;Saeki, Y.;Carette, J. E.;Weissleder, R.;Vandertop, W. P.;Van Beusechem, V. W.;Dirven, C. M.Chiocca, E. A. Cyclophosphamide increases transgene expression mediated by an oncolytic adenovirus in glioma-bearing mice monitored by bioluminescence imaging. MoI Ther 2006, 14, 779-88.
61. Fulci, G.;Breymann, L;Gianni, D.;Kurozomi, K.;Rhee, S. S.;Yu, J.;Kaur, B.;Louis, D. N.;Weissleder, R.;Caligiuh, M. A.Chiocca, E. A. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc Natl Acad ScI U S A
2006, 103, 12873-8.
62. Xie, D.;Yin, D.fTong, X.;O'kelly, J.;Moh, A.;Miller, C.;Black, K.;Gui, D.;Said, J. W.Koeffler, H. P. Cyr61 is overexpressed in gliomas and involved in integhn-linked kinase-mediated akt and beta-catenin-tcf/lef signaling pathways. Cancer Res 2004, 64, 1987-96. 63. Xie, D.;Miller, C. W.;O'kelly, J.;Nakachi, K.;Sakashita, A.;Said, J. W.;Gornbein, J.Koeffler, H. P. Breast cancer. Cyr61 is overexpressed, estrogen-inducible, and associated with more advanced disease. J Biol Chem 2001, 276, 14187-94.
64. Xie, D.;Yin, D.;Wang, H. J.;Liu, G. T.;Elashoff, R.;Black, K.Koeffler, H. P. Levels of expression of cyr61 and ctgf are prognostic for tumor progression and survival of individuals with gliomas. Clin Cancer Res 2004, 10, 2072-81.
65. Mo, F. E.;Muntean, A. G.;Chen, C. C.;Stolz, D. B.;Watkins, S. C.Lau, L. F. Cyr61 (ccn1) is essential for placental development and vascular integrity. MoI Cell Biol 2002, 22, 8709-20.
66. Menendez, J. A.;Mehmi, l.;Griggs, D. W.Lupu, R. The angiogenic factor cyr61 in breast cancer: Molecular pathology and therapeutic perspectives. Endocr Relat Cancer 2003, 10, 141-52.
67. Fukushima, Y.;Oshika, Y.;Tsuchida, T.;Tokunaga, T.;Hatanaka, H.;Kijima, H.;Yamazaki, H.;Ueyama, Y.;Tamaoki, N.Nakamura, M. Brain-specific angiogenesis inhibitor 1 expression is inversely correlated with vascularity and distant metastasis of colorectal cancer, lnt J Oncol 1998, 13, 967-70.
68. Hatanaka, H.;Oshika, Y.;Abe, Y.;Yoshida, Y.;Hashimoto, T.;Handa, A.;Kijima, H.;Yamazaki, H.;lnoue, H.;Ueyama, Y.Nakamura, M. Vascularization is decreased in pulmonary adenocarcinoma expressing brain-specific angiogenesis inhibitor 1 (bail), lnt J MoI Med 2000, 5, 181 -3.
69. Lee, J. H.;Koh, J. T.;Shin, B. A.;Ahn, K. Y.;Roh, J. H.;Kim, Y. J.Kim, K. K. Comparative study of angiostatic and anti-invasive gene expressions as prognostic factors in gastric cancer, lnt J Oncol 2001 , 18, 355-61.
70. Kaur, B.;Brat, D. J.;Devi, N. S. Van Meir, E. G. Vasculostatin, a proteolytic fragment of brain angiogenesis inhibitor 1 , is an antiangiogenic and antitumorigenic factor. Oncogene 2005, 24, 3632-42.
71. Volpert, O. V.;Zaichuk, T.;Zhou, W.;Reiher, F.;Ferguson, T. A.;Stuart, P. M.;Amin, M.Bouck, N. P. Inducer-stimulated fas targets activated endothelium for destruction by anti-angiogenic thrombospondin-1 and pigment epithelium-derived factor. Nat Med 2002, 8, 349-57.
72. Sun, X.;Skorstengaard, K.Mosher, D. F. Disulfides modulate rgd-inhibitable cell adhesive activity of thrombospondin. J Cell Biol 1992, 118, 693-701.
73. Tyminski, E.;Leroy, S.fTerada, K.;Finkelstein, D. M.; Hyatt, J. L;Danks, M. K.;Potter, P. M.;Saeki, Y.Chiocca, E. A. Brain tumor oncolysis with replication-conditional herpes simplex virus type 1 expressing the prodrug-activating genes, cyp2b1 and secreted human intestinal carboxylesterase, in combination with cyclophosphamide and irinotecan. Cancer Res 2005, 65, 6850-7.
74. Barber, G. N. The dsrna-dependent protein kinase, pkr and cell death. Cell Death Differ 2005, 12, 563-70.
75. Jacobs, A.;Breakefield, X. O.Fraefel, C. Hsv-1 -based vectors for gene therapy of neurological diseases and brain tumors: Part i. Hsv-1 structure, replication and pathogenesis. Neoplasia 1999, 1, 387-401.
76. GaIIi, R.;Binda, E.;Orfanelli, U.;Cipelletti, B.;Ghtti, A.;De Vitis, S.;Fiocco, R.;Foroni, C.;Dimeco, F.Vescovi, A. Isolation and characterization of tumohgenic, stem-like neural precursors from human glioblastoma. Cancer Res 2004, 64, 7011 -21.
77. Singh, S. K.;Hawkins, C.;Clarke, I. D.;Squire, J. A.;Bayani, J.;Hide, T.;Henkelman, R. M.;Cusimano, M. D. Dirks, P. B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396-401.
78. Kambara, H.;Saeki, Y.Chiocca, E. A. Cyclophosphamide allows for in vivo dose reduction of a potent oncolytic virus. Cancer Res 2005, 65, 11255-8.
79. Ribatti, D.;Nico, B.;Vacca, A.Presta, M. The gelatin sponge-chohoallantoic membrane assay. Nature Protocols 2006, 1, 85-91.
80. AIi, S.;King, G. D.;Curtin, J. F.;Candolfi, M.;Xiong, W.;Liu, C.;Puntel, M.;Cheng, Q.;Pheto, J.;Ribas, A.;Kupiec-Weglinski, J.;Van Rooijen, N.;l_assmann, H.;Lowenstein, P. R.Castro, M. G. Combined immunostimulation and conditional cytotoxic gene therapy provide long-term survival in a large glioma model. Cancer Res 2005, 65, 7194-204.
81. Kaur, B.;Tan, C.;Brat, D. J.;Post, D. E.Van Meir, E. G. Genetic and hypoxic regulation of angiogenesis in gliomas. J Neurooncol 2004, 70, 229-43.
82. Jain, R. K. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science 2005, 307, 58-62.
83. Kaur, B.;Khwaja, F. W.;Severson, E. A.;Matheny, S. L;Brat, D. J.Van Meir, E. G. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro-oncol 2005, 7, 134-53.
84. Boviatsis, E. J.;Scharf, J. M.;Chase, M.; Harrington, K.;Kowall, N. W.;Breakefield, X. O.Chiocca, E. A. Antitumor activity and reporter gene transfer into rat brain neoplasms inoculated with herpes simplex virus vectors defective in thymidine kinase or ribonucleotide reductase. Gene Ther 1994, 1, 323-31.
85. Vredenburgh, J. J.;Desjardins, A.;Herndon, J. E., 2nd;Dowell, J. M.;Reardon, D. A.;Quinn, J. A.;Rich, J. N.;Sathornsumetee, S.;Gururangan, S.;Wagner, M.;Bigner, D. D.;Friedman, A. H.Friedman, H. S. Phase ii trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res 2007, 13, 1253-9. 86. Lamfers, M. L;Fulci, G.;Gianni, D.;Tang, Y.;Kurozumi, K.;Kaur, B.;Moeniralm, S.;Saeki, Y.;Carette, J. E.;Weissleder, R.;Vandertop, W. P.;Van Beusechem, V. W.;Dirven, C. M.Chiocca, E. A. Cyclophosphamide increases transgene expression mediated by an oncolytic adenovirus in glioma-bearing mice monitored by bioluminescence imaging. MoI Ther 2006.
[00129] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the exemplary embodiments, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

Claims

CLAIMS WE CLAIM:
1. A recombinant expression vector comprising an immediate early HSV promoter IE4/5 operably linked to a nucleic acid sequence encoding an angiostatic polypeptide.
2. The recombinant expression vector of claim 1 , further comprising: a nucleic acid sequence encoding a modified herpes simplex virus.
3. The recombinant expression vector of claim 2, wherein: the modified herpes simplex virus is a mutant herpes simplex virus type 1.
4. The recombinant viral expression vector of claim 1 , wherein: the nucleic acid sequence encoding an angiostatic polypeptide comprises a nucleotide sequence encoding a proteolytic polypeptide fragment of BAM .
5. The recombinant expression vector of claim 1 or 3, wherein: the nucleic acid sequence encoding an angiostatic polypeptide comprises SEQ ID NO: 1 or of a degenerate variant of SEQ ID NO: 1.
6. The recombinant expression vector of claim 1 or 3, wherein: the nucleic acid sequence encoding an angiostatic polypeptide comprises SEQ ID NO: 3 or of a degenerate variant of SEQ ID NO: 3.
7. The recombinant expression vector of claim 1 or 3, further comprising: a further nucleic acid sequence encoding a therapeutic polypeptide.
8. A recombinant oncolytic virus comprising: a nucleic acid sequence encoding a modified herpes virus; and a nucleotide sequence of SEQ ID NO: 1 or of a degenerate variant of SEQ ID NO: 1 operably linked to an expression control sequence.
9. The recombinant oncolytic virus of claim 8, wherein: the modified herpes simplex virus is a mutant herpes simplex virus type 1.
10. The oncolytic virus of claim 9, wherein: the expression control sequence comprises a viral promoter.
11. The oncolytic virus of claim 8 or 10, wherein: the expression control sequence comprises an immediate early HSV promoter IE4/5.
12. The oncolytic virus of claim 8, wherein: the expression control sequence comprises the promoter of SEQ ID NO: 5.
13. A method of killing intracranial tumor cells in a mammal comprising introducing into the vicinity of the tumor cells an expression vector, comprising: a modified herpes virus nucleic acid sequence; and a nucleotide sequence of SEQ ID NO: 1 or of a degenerate variant of SEQ ID NO: 1 operably linked to an expression control sequence.
14. The method of claim 13, wherein: the expression control sequence comprises an immediate early HSV promoter IE4/5.
15. The method of claim 13, further comprising the step of: mixing a pharmacologically acceptable carrier with the expression vector prior to the introducing step.
PCT/US2008/080367 2007-10-17 2008-10-17 Oncolytic virus WO2009052426A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/697,891 US8450106B2 (en) 2007-10-17 2010-02-01 Oncolytic virus
US13/903,695 US20130316447A1 (en) 2007-10-17 2013-05-28 Oncolytic virus

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US98066407P 2007-10-17 2007-10-17
US60/980,664 2007-10-17

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/697,891 Continuation US8450106B2 (en) 2007-10-17 2010-02-01 Oncolytic virus

Publications (2)

Publication Number Publication Date
WO2009052426A2 true WO2009052426A2 (en) 2009-04-23
WO2009052426A3 WO2009052426A3 (en) 2009-07-30

Family

ID=40568089

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/080367 WO2009052426A2 (en) 2007-10-17 2008-10-17 Oncolytic virus

Country Status (1)

Country Link
WO (1) WO2009052426A2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6379674B1 (en) * 1997-08-12 2002-04-30 Georgetown University Use of herpes vectors for tumor therapy
US20060099224A1 (en) * 2002-08-12 2006-05-11 David Kirn Methods and compositions concerning poxviruses and cancer
US20060147420A1 (en) * 2004-03-10 2006-07-06 Juan Fueyo Oncolytic adenovirus armed with therapeutic genes

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6379674B1 (en) * 1997-08-12 2002-04-30 Georgetown University Use of herpes vectors for tumor therapy
US20060099224A1 (en) * 2002-08-12 2006-05-11 David Kirn Methods and compositions concerning poxviruses and cancer
US20060147420A1 (en) * 2004-03-10 2006-07-06 Juan Fueyo Oncolytic adenovirus armed with therapeutic genes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
S. YAMAMOTO ET AL.: 'Imaging innediate-early and strict-late promoter activity during oncolytic herpes simplex virus type 1 infection and replication in tumors.' GENE THERAPY. vol. 13, 2006, pages 1731 - 1736 *

Also Published As

Publication number Publication date
WO2009052426A3 (en) 2009-07-30

Similar Documents

Publication Publication Date Title
US8450106B2 (en) Oncolytic virus
JP7262134B2 (en) Oncolytic HSV vectors
JP6644378B2 (en) Virus strain
Burton et al. Gene delivery using herpes simplex virus vectors
Biederer et al. Replication-selective viruses for cancer therapy
Zhao et al. Potent antitumor activity of oncolytic adenovirus expressing mda-7/IL-24 for colorectal cancer
AU2018230046B9 (en) Recombinant Herpes simplex virus and use thereof
EP1648481B1 (en) Viral vectors
Parker et al. Oncolytic viral therapy of malignant glioma
US6723316B2 (en) Herpes simplex virus-1 Glycoprotein C mutants for treating unwanted hyperproliferative cell growth
WO2013038066A1 (en) Modified oncolytic vaccinia virus
JP2005521398A (en) A powerful oncolytic herpes simplex virus for cancer treatment
Lachmann Herpes simplex virus‐based vectors
Wildner Oncolytic viruses as therapeutic agents
Fulci et al. ONCOLYTIC VIRUSES FOR THE THERAPY OF BRAIN TUMORS AND OTHER SOLID MALIGNANCIES: A
US20230026342A1 (en) New generation regulatable fusogenic oncolytic herpes simplex virus type 1 virus and methods of use
JP5070583B2 (en) Recombinant HSV useful for human glioma treatment
Marconi et al. HSV as a vector in vaccine development and gene therapy
US10610554B2 (en) Oncolytic expression vector comprising a nucleic acid comprising a nucleotide sequence encoding GADD34
US20220002680A1 (en) Regulatable fusogenic oncolytic herpes simplex virus type 1 virus and methods of use
Stanziale et al. Novel approaches to cancer therapy using oncolytic viruses
WO2009052426A2 (en) Oncolytic virus
US20150246086A1 (en) Use of Mutant Herpes Simplex Virus-2 for Cancer Therapy
Hu et al. Oncolytic herpes simplex virus for tumor therapy
Latchman Herpes simplex virus vectors for Parkinson's disease

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08839446

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08839446

Country of ref document: EP

Kind code of ref document: A2