WO2020014539A1 - Procédés et compositions pour cibler des cellules cancéreuses pour un traitement - Google Patents

Procédés et compositions pour cibler des cellules cancéreuses pour un traitement Download PDF

Info

Publication number
WO2020014539A1
WO2020014539A1 PCT/US2019/041484 US2019041484W WO2020014539A1 WO 2020014539 A1 WO2020014539 A1 WO 2020014539A1 US 2019041484 W US2019041484 W US 2019041484W WO 2020014539 A1 WO2020014539 A1 WO 2020014539A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
deletion
site
pharmaceutical composition
cancer
Prior art date
Application number
PCT/US2019/041484
Other languages
English (en)
Inventor
Michael J. Abrams
Christopher Larson
Tony R. REID
Bryan T. Oronsky
Original Assignee
Epicentrx, Inc.
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 Epicentrx, Inc. filed Critical Epicentrx, Inc.
Publication of WO2020014539A1 publication Critical patent/WO2020014539A1/fr

Links

Classifications

    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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
    • A61K35/761Adenovirus
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • A61K39/001195Prostate specific membrane antigen [PSMA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • 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
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0489Phosphates or phosphonates, e.g. bone-seeking phosphonates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0495Pretargeting
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • 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
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10332Use 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
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10371Demonstrated in vivo effect
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the field of the invention is methods and compositions for treating cancer.
  • viruses have shown promise as oncolytic agents that not only directly destroy malignant cells via an infection-to-reproduction-to-lysis chain reaction but also indirectly induce anti-tumor immunity. These immune stimulatory properties have been augmented with the insertion of therapeutic transgenes that are copied and expressed each time the virus replicates.
  • Previously developed oncolytic viruses include the oncolytic serotype 5 adenovirus (Ad5) referred to as TAV-255 that is transcriptionally attenuated in normal cells but transcriptionally active in cancer cells (see, PCT Publication No. W02010/101921). It is believed that the mechanism by which the TAV-255 vector achieves such tumor selectivity is through targeted deletion of three transcriptional factor (TF) binding sites for the transcription factors Pea3 and E2F, proteins that regulate adenovirus expression of Ela, the earliest gene to be transcribed after virus entry into the host cell, through binding to specific DNA sequences.
  • TF transcriptional factor
  • the invention is based, in part, upon the discovery that a delivery vehicle, e.g., a recombinant oncolytic virus, can be used to selectively express a protein defining a binding site or a protein that mediates the production of a biomolecule defining a binding site in a cancer cell, and thereby target the cancer cell for a treatment by a therapeutic agent that binds to the binding site.
  • a delivery vehicle e.g., a recombinant oncolytic virus described herein, can be used to selectively express a bone morphogenetic protein (BMP) in a cancer cell and mediate the production of bone tissue in a tumor.
  • BMP bone morphogenetic protein
  • a BMP in a cancer cell allows for treatment of a cancer by targeting a therapeutic agent that binds to bone tissue, e.g., a bone targeting radionuclide, to a tumor.
  • a therapeutic agent that binds to bone tissue
  • the delivery vehicle e.g., the recombinant oncolytic virus
  • the invention provides a method of treating a cancer in a subject in need thereof, the method comprising administering to the subject: (a) an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding: (i) a protein defining a binding site; or (ii) a protein that mediates the production of a biomolecule defining a binding site; and (b) an effective amount of a therapeutic agent that binds to the binding site.
  • the biomolecule defining a binding site is bone tissue, and/or the protein that mediates the production of the biomolecule defining a binding site is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR), osteocalcin, osteopontin, bone sialoprotein, osteonectin, osteoprotegerin (OPG) and alkaline phosphatase.
  • the protein defining the binding site is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR) and prostate specific membrane antigen (PSMA).
  • the therapeutic agent comprises a radionuclide or an antibody.
  • the radionuclide is a bone targeting radionuclide, e.g., 89 Sr (e.g., 89 Sr chloride), 223 Ra (e.g, 223 Ra dichloride), 153 Sm (e.g, 153 Sm-EDTMP), 177 Lu, 90 Y, 186 Re (e.g., 186 Re HEDP), 117m Sn (e.g, 117m Sn DTP A), and 32 P.
  • the antibody is an antibody drug conjugate or an antibody radionuclide conjugate.
  • the invention provides a method of treating bone loss and/or hypercalcemia in a subject in need thereof, the method comprising administering to the subject an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding a protein that mediates the production of bone tissue.
  • the subject has cancer.
  • the invention provides a method of generating bone tissue in a subject, the method comprising administering to the subject an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding a protein that mediates the production of bone tissue.
  • the protein that mediates the production of bone is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR), osteocalcin, osteopontin, bone sialoprotein, osteonectin, osteoprotegerin (OPG) and alkaline phosphatase.
  • BMP bone morphogenetic protein
  • BMPR bone morphogenetic protein receptor
  • osteocalcin osteopontin
  • bone sialoprotein osteonectin
  • osteoprotegerin OPG
  • alkaline phosphatase alkaline phosphatase
  • the BMP is selected from BMP -2, BMP-4, BMP-6, and BMP-7.
  • the BMP comprises SEQ ID NO: 1 or a functional fragment thereof, SEQ ID NO: 2 or a functional fragment thereof, SEQ ID NO: 3 or a functional fragment thereof, or SEQ ID NO: 4 or a functional fragment thereof, or an amino acid sequence having 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
  • the delivery vehicle is a recombinant oncolytic virus.
  • the recombinant oncolytic virus is selected from an adenovirus, fowlpox virus, lentivirus, herpes virus, vesicular stomatitis virus (VSV), maraba virus, polio virus, and alpha virus.
  • the recombinant virus is a recombinant oncolytic adenovirus, e.g., a type 5 adenovirus (Ad5).
  • the exogenous nucleotide sequence is inserted into an Elb-l9K insertion site located between the start site of Elb-l9K and the start site of Elb-55K.
  • the Elb-l9K insertion site is located between the start site of Elb-l9K and the stop site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion of from about 100 to about 305, about 100 to about 300, about 100 to about 250, about 100 to about 200, about 100 to about 150, about 150 to about 305, about 150 to about 300, about 150 to about 250, or about 150 to about 200 nucleotides adjacent the start site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion of about 200 nucleotides, e.g., 203 nucleotides adjacent the start site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion corresponding to nucleotides 1714-1916 of the Ad5 genome (SEQ ID NO: 5), or, the exogenous nucleotide sequence is inserted between nucleotides corresponding to 1714 and 1916 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between CTGACCTC (SEQ ID NO: 6) and TCACCAGG (SEQ ID NO: 7), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, CTGACCTC (SEQ ID NO: 6), the exogenous nucleotide sequence, and
  • the exogenous nucleotide sequence is inserted into an E3 insertion site located between the stop site of pVIII and the start site of Fiber.
  • the E3 insertion site comprises a deletion of from about 500 to about 3185, from about 500 to about 3000, from about 500 to about 2500, from about 500 to about 2000, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 3185, from about 1000 to about 3000, from about 1000 to about 2500, from about 1000 to about 2000, from about 1000 to about 1500, from about 1500 to about 3185, from about 1500 to about 3000, from about 1500 to about 2000, from about 2000 to about 3185, from about 2000 to about 3000, from about 2000 to about 2500, from about 2500 to about 3185, from about 2500 to about 3000, or from about 3000 to about 3185 nucleotides.
  • the E3 insertion site is located between the stop site of E3-10.5K and the stop site of E3-14.7K. In certain embodiments, the E3 insertion site comprises a deletion of from about 500 to about 1551, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 1551, from about 1000 to about 1500, or from about 1500 to about 1551 nucleotides adjacent the stop site of E3-10.5K. In certain embodiments, the E3 insertion site comprises a deletion of about 1050 nucleotides adjacent the stop site of E3-10.5K, e.g., the E3 insertion site comprises a deletion of 1063 or 1064 nucleotides adjacent the stop site of E3-10.5K.
  • the E3 insertion site comprises a deletion corresponding to the Ad5 dl309 E3 deletion. In certain embodiments, the E3 insertion site comprises a deletion corresponding to nucleotides 29773-30836 of the Ad5 genome (SEQ ID NO: 5), or, the exogenous nucleotide sequence is inserted between nucleotides corresponding to 29773 and 30836 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between CAGTATGA (SEQ ID NO: 8) and TAATAAAAAA (SEQ ID NO: 9), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, CAGTATGA (SEQ ID NO: 8), the exogenous nucleotide sequence, and TAATAAAAAA (SEQ ID NO: 9).
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one Pea3 binding site, or a functional portion thereof, e.g., the virus may comprise a deletion of nucleotides
  • the recombinant adenovirus may comprise a deletion of nucleotides corresponding to 195-244 of the Ad5 genome (SEQ ID NO: 5), and/or the recombinant adenovirus may comprise the sequence GGTGTTTTGG (SEQ ID NO: 10).
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one Pea3 binding site, or a functional portion thereof, and not comprise a deletion of an E2F binding site.
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one E2F binding site, or a functional portion thereof. In certain embodiments, the recombinant oncolytic adenovirus may comprise a deletion of at least one E2F binding site, or a functional portion thereof, and not comprise a deletion of a Pea3 binding site.
  • the recombinant oncolytic adenovirus may comprise an Ela promoter having a deletion of a functional TATA box, e.g., the deletion of an entire TATA box.
  • the virus may comprise a deletion of nucleotides corresponding to -29 to -26, -33 to -26, -44 to +52, or -148 to +52 of the Ela promoter.
  • the deletion comprises a deletion of nucleotides corresponding to 353-552 of the Ad5 genome (SEQ ID NO: 5), and/or the Ela promoter comprises the sequence CTAGGACTG (SEQ ID NO: 11).
  • the exogenous nucleotide sequence is not operably linked to an exogenous promoter sequence.
  • the recombinant oncolytic adenovirus may selectively replicate in a hyperproliferative cell.
  • the recombinant oncolytic adenovirus may selectively express the protein defining a binding site or the protein that mediates the production of a biomolecule defining a binding site in a hyperproliferative cell.
  • the hyperproliferative cell may be a cancer cell, e.g., a lung cancer cell, a colon cancer cell, and a pancreatic cancer cell.
  • the recombinant oncolytic adenovirus comprises a nucleotide sequence of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, or SEQ ID NO: 15, or a nucleotide sequence having 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, or SEQ ID NO: 15.
  • the recombinant oncolytic virus further comprises an exogenous nucleotide sequence encoding a
  • costimulatory molecule an antiangiogenic and/or antivascular molecule, an anti-fibrotic molecule, an anti-inflammatory or pro-inflammatory molecule, a monoclonal antibody or fragment thereof, a pro-apoptotic molecule, an enzyme, an immunomodulatory molecule, a cytokine, or a cytokine inhibitor (e.g., a cytokine trap).
  • the delivery vehicle e.g. , a recombinant oncolytic virus
  • the delivery vehicle e.g., a recombinant oncolytic virus
  • the therapeutic agent are administered in combination with one or more therapies selected from surgery, radiation, chemotherapy, immunotherapy, hormone therapy, and virotherapy.
  • the cancer is selected from melanoma, squamous cell carcinoma of the skin, basal cell carcinoma, head and neck cancer, breast cancer, anal cancer, cervical cancer, non-small cell lung cancer, mesothelioma, small cell lung cancer, renal cell carcinoma, prostate cancer, gastroesophageal cancer, colorectal cancer, testicular cancer, bladder cancer, ovarian cancer, liver cancer, hepatocellular carcinoma, cholangiocarcinoma, brain and central nervous system cancer, thyroid cancer, parathyroid cancer (e.g., parathyroid carcinoma), endometrial cancer, neuroendocrine cancer, lymphoma (e.g., Hodgkin and non- Hodgkin), leukemia, merkel cell carcinoma, gastrointestinal stromal tumors, multiple myeloma, uterine cancer, a sarcoma, kidney cancer, ocular cancer, pancreatic cancer, and a germ cell cancer (e.g., ovarian germ cell cancer).
  • melanoma
  • the delivery vehicle and/or the therapeutic agent are administered in combination with a bisphosphonate, hyaluronic acid, vitamin D, calcium, testosterone, estrogen, progesterone, a selective estrogen modulator (SERM), calcitonin, a RANK or RANKL inhibitor (e.g., denosumab), pulsed ultrasound (e.g., low intensity pulsed ultrasound (LIPU)), electrical stimulation,
  • a bisphosphonate, hyaluronic acid, vitamin D, calcium, testosterone, estrogen, progesterone a selective estrogen modulator (SERM), calcitonin, a RANK or RANKL inhibitor (e.g., denosumab), pulsed ultrasound (e.g., low intensity pulsed ultrasound (LIPU)), electrical stimulation
  • SERM selective estrogen modulator
  • ESWT extracorporeal shockwave therapy
  • PEMF pulsed electromagnetic field
  • the effective amount of the recombinant oncolytic virus is 10 2 -10 15 plaque forming units (pfiis).
  • the subject can, e.g., be a human, e.g., a pediatric human, or an animal.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: (a) an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding: (i) a protein defining a binding site; or (ii) a protein that mediates the production of a biomolecule defining a binding site; and (b) optionally, an effective amount of a therapeutic agent that binds to the binding site.
  • the biomolecule defining a binding site is bone tissue, and/or the protein that mediates the production of the biomolecule defining a binding site is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR), osteocalcin, osteopontin, bone sialoprotein, osteonectin, osteoprotegerin (OPG) and alkaline phosphatase.
  • BMP bone morphogenetic protein
  • BMPR bone morphogenetic protein receptor
  • osteocalcin osteopontin
  • bone sialoprotein osteonectin
  • osteoprotegerin OPG
  • alkaline phosphatase alkaline phosphatase
  • the protein defining the binding site is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR) and prostate specific membrane antigen (PSMA).
  • BMP bone morphogenetic protein
  • BMPR bone morphogenetic protein receptor
  • PSMA prostate specific membrane antigen
  • the BMP comprises SEQ ID NO: 1 or a functional fragment thereof, SEQ ID NO: 2 or a functional fragment thereof, SEQ ID NO: 3 or a functional fragment thereof, or SEQ ID NO: 4 or a functional fragment thereof, or an amino acid sequence having 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
  • the therapeutic agent comprises a radionuclide or an antibody.
  • the radionuclide is a bone targeting radionuclide, e.g., 89 Sr (e.g., 89 Sr chloride), 223 Ra (e.g., 223 Ra dichloride), 153 Sm (e.g, 153 Sm-EDTMP), 177 Lu, 90 Y, 186 Re (e.g., 186 Re HEDP), 117m Sn (e.g, 117m Sn DTP A), and 32 P.
  • the antibody is an antibody drug conjugate or an antibody radionuclide conjugate.
  • the delivery vehicle is a recombinant oncolytic virus.
  • the recombinant oncolytic virus is selected from an adenovirus, fowlpox virus, lentivirus, herpes virus, vesicular stomatitis virus (VSV), maraba virus, polio virus, and alpha virus.
  • the recombinant virus is a recombinant oncolytic adenovirus, e.g., a type 5 adenovirus (Ad5).
  • the exogenous nucleotide sequence is inserted into an Elb-l9K insertion site located between the start site of Elb-l9K and the start site of Elb-55K.
  • the Elb-l9K insertion site is located between the start site of Elb-l9K and the stop site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion of from about 100 to about 305, about 100 to about 300, about 100 to about 250, about 100 to about 200, about 100 to about 150, about 150 to about 305, about 150 to about 300, about 150 to about 250, or about 150 to about 200 nucleotides adjacent the start site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion of about 200 nucleotides, e.g., 203 nucleotides adjacent the start site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion corresponding to nucleotides 1714-1916 of the Ad5 genome (SEQ ID NO: 5), or, the exogenous nucleotide sequence is inserted between nucleotides corresponding to 1714 and 1916 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between CTGACCTC (SEQ ID NO: 6) and TCACCAGG (SEQ ID NO: 7), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, CTGACCTC (SEQ ID NO: 6), the exogenous nucleotide sequence, and
  • the exogenous nucleotide sequence is inserted into an E3 insertion site located between the stop site of pVIII and the start site of Fiber.
  • the E3 insertion site comprises a deletion of from about 500 to about 3185, from about 500 to about 3000, from about 500 to about 2500, from about 500 to about 2000, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 3185, from about 1000 to about 3000, from about 1000 to about 2500, from about 1000 to about 2000, from about 1000 to about 1500, from about 1500 to about 3185, from about 1500 to about 3000, from about 1500 to about 2000, from about 2000 to about 3185, from about 2000 to about 3000, from about 2000 to about 2500, from about 2500 to about 3185, from about 2500 to about 3000, or from about 3000 to about 3185 nucleotides.
  • the E3 insertion site is located between the stop site of E3-10.5K and the stop site of E3-14.7K. In certain embodiments, the E3 insertion site comprises a deletion of from about 500 to about 1551, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 1551, from about 1000 to about 1500, or from about 1500 to about 1551 nucleotides adjacent the stop site of E3-10.5K. In certain embodiments, the E3 insertion site comprises a deletion of about 1050 nucleotides adjacent the stop site of E3-10.5K, e.g., the E3 insertion site comprises a deletion of 1063 or 1064 nucleotides adjacent the stop site of E3-10.5K.
  • the E3 insertion site comprises a deletion corresponding to the Ad5 dl309 E3 deletion. In certain embodiments, the E3 insertion site comprises a deletion corresponding to nucleotides 29773-30836 of the Ad5 genome (SEQ ID NO: 5), or, the exogenous nucleotide sequence is inserted between nucleotides corresponding to 29773 and 30836 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between CAGTATGA (SEQ ID NO: 8) and TAATAAAAAA (SEQ ID NO: 9), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, CAGTATGA (SEQ ID NO: 8), the exogenous nucleotide sequence, and TAATAAAAAA (SEQ ID NO: 9).
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one Pea3 binding site, or a functional portion thereof, e.g., the virus may comprise a deletion of nucleotides corresponding to about -300 to about -250 upstream of the initiation site of E la or a deletion of nucleotides corresponding to -305 or -304 to -255 upstream of the initiation site of Ela.
  • the recombinant adenovirus may comprise a deletion of nucleotides corresponding to 195-244 of the Ad5 genome (SEQ ID NO: 5), and/or the recombinant adenovirus may comprise the sequence GGTGTTTTGG (SEQ ID NO: 10).
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one Pea3 binding site, or a functional portion thereof, and not comprise a deletion of an E2F binding site.
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one E2F binding site, or a functional portion thereof.
  • the recombinant oncolytic adenovirus may comprise a deletion of at least one E2F binding site, or a functional portion thereof, and not comprise a deletion of a Pea3 binding site.
  • the recombinant oncolytic adenovirus may comprise an Ela promoter having a deletion of a functional TATA box, e.g., the deletion of an entire TATA box.
  • the virus may comprise a deletion of nucleotides corresponding to -29 to -26, -33 to -26, -44 to +52, or -148 to +52 of the Ela promoter.
  • the deletion comprises a deletion of nucleotides corresponding to 353-552 of the Ad5 genome (SEQ ID NO: 5), and/or the Ela promoter comprises the sequence CTAGGACTG (SEQ ID NO: 11).
  • the exogenous nucleotide sequence is not operably linked to an exogenous promoter sequence.
  • the recombinant oncolytic adenovirus may selectively replicate in a hyperproliferative cell.
  • the recombinant oncolytic adenovirus may selectively express the protein defining a binding site or the protein that mediates the production of a biomolecule defining a binding site in a hyperproliferative cell.
  • the hyperproliferative cell may be a cancer cell, e.g., a lung cancer cell, a colon cancer cell, and a pancreatic cancer cell.
  • the recombinant oncolytic adenovirus comprises a nucleotide sequence of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, or SEQ ID NO: 15, or a nucleotide sequence having 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, or SEQ ID NO: 15.
  • the recombinant oncolytic virus further comprises an exogenous nucleotide sequence encoding a
  • the composition further comprises a bisphosphonate, hyaluronic acid, vitamin D, calcium, testosterone, estrogen, progesterone, a selective estrogen modulator (SERM), calcitonin, or a RANK or RANKL inhibitor (e.g., denosumab).
  • SERM selective estrogen modulator
  • FIGURE 1 is a bar graph showing the expression of BMP-2 by the TAV-BMP-2 virus in A549 cells as determined by ELISA.
  • FIGURE 2 is a bar graph showing the expression of BMP-7 by the TAV-BMP-7 virus in A549 cells as determined by ELISA.
  • FIGURE 3 depicts crystal violet staining of A549 cells at the indicated time points following infection with the TAV- A 19k, TAV-BMP-2, and TAV-BMP-7 viruses.
  • FIGURE 4 depicts tumor volumes of mice carrying subcutaneous ADS-12 tumors treated with vehicle, TAVA19k. or TAV-BMP-7 viruses. Lines show the mean tumor volume of 10 mice in each treatment group and error bars show SEM.
  • the invention is based, in part, upon the discovery that a delivery vehicle, e.g., a recombinant oncolytic virus, can be used to selectively express a protein defining a binding site or a protein that mediates the production of a biomolecule defining a binding site in a cancer cell, and thereby target the cancer cell for a treatment by a therapeutic agent that binds to the binding site.
  • a delivery vehicle e.g., a recombinant oncolytic virus
  • a delivery vehicle e.g., a recombinant oncolytic virus described herein
  • a delivery vehicle can be used to selectively express a bone morphogenetic protein (BMP) in a cancer cell, and cause a cancer to acquire a bone cell phenotype, e.g., an osteoblast-like cell phenotype, and/or mediate the production of bone tissue in a tumor.
  • BMP bone morphogenetic protein
  • Certain cancers and metastases are associated with increased levels of BMP expression, and cancer cells that acquire a bone cell phenotype have a greater likelihood of surviving and proliferating in bone.
  • the invention provides a method of treating a cancer in a subject in need thereof, the method comprising administering to the subject: (a) an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding: (i) a protein defining a binding site; or (ii) a protein that mediates the production of a biomolecule defining a binding site; and (b) an effective amount of a therapeutic agent that binds to the binding site.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: (a) an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding: (i) a protein defining a binding site; or (ii) a protein that mediates the production of a biomolecule defining a binding site; and (b) optionally, an effective amount of a therapeutic agent that binds to the binding site.
  • the invention provides methods for treating bone loss and/or hypercalcemia or promoting the growth of bone in a subject in need thereof using a delivery vehicle (e.g., a recombinant oncolytic virus) described herein.
  • a delivery vehicle e.g., a recombinant oncolytic virus
  • Delivery vehicles suitable for use in the invention include, e.g., viruses, bacteria (e.g., Listeria monocytogenes, Salmonella enterica or Serovar typhi murium), nanoparticles, liposomes, exosomes, or microemulsions.
  • viruses e.g., viruses, bacteria (e.g., Listeria monocytogenes, Salmonella enterica or Serovar typhi murium), nanoparticles, liposomes, exosomes, or microemulsions.
  • viruses are used herein to refer any of the obligate intracellular parasites having no protein-synthesizing or energy-generating mechanism.
  • the viral genome may be RNA or DNA.
  • the viruses useful in the practice of the present invention include recombinantly modified enveloped or non-enveloped DNA and RNA viruses, preferably selected from baculoviridiae, parvoviridiae, picomoviridiae, herpesviridiae, poxyiridae, or adenoviridiae.
  • a recombinantly modified virus is referred to herein as a“recombinant virus.”
  • a recombinant virus may, e.g., be modified by recombinant DNA techniques to be replication deficient, conditionally replicating, or replication competent, and/or be modified by recombinant DNA techniques to include expression of exogenous transgenes.
  • Chimeric viral vectors which exploit advantageous elements of each of the parent vector properties (See, e.g., Feng et al. (1997) NATURE BIOTECHNOLOGY 15:866-870) may also be useful in the practice of the present invention. Although it is generally favored to employ a virus from the species to be treated, in some instances it may be advantageous to use vectors derived from different species that possess favorable pathogenic features.
  • equine herpes virus vectors for human gene therapy are described in PCT Publication No. WO 98/27216.
  • the vectors are described as useful for the treatment of humans as the equine virus is not pathogenic to humans.
  • ovine adenoviral vectors may be used in human gene therapy as they are claimed to avoid the antibodies against the human adenoviral vectors.
  • Such vectors are described in PCT Publication No. WO 97/06826.
  • viruses useful in the practice of the invention include adenovirus, fowlpox virus, lentivirus, herpes virus, vesicular stomatitis virus (VSV), maraba virus, polio virus, and alpha virus.
  • the recombinant virus is an adenovirus.
  • Adenoviruses are medium-sized (90-100 nm), non-enveloped (naked), icosahedral viruses composed of a nucleocapsid and a double-stranded linear DNA genome. Adenoviruses replicate in the nucleus of mammalian cells using the host's replication machinery.
  • adenovirus refers to any virus in the genus Adenoviridiae including, but not limited to, human, bovine, ovine, equine, canine, porcine, murine, and simian adenovirus subgenera.
  • human adenoviruses includes the A-F subgenera as well as the individual serotypes thereof, the individual serotypes and A-F subgenera including but not limited to human adenovirus types 1, 2, 3, 4, 4a, 5, 6, 7, 8, 9, 10, 11 (Adl la and Adl lp), 12, 13, 14, 15, 16, 17, 18, 19, l9a, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 34a, 35, 35p, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, and 91.
  • Preferred are recombinant viruses derived from human adenovirus types 2 and 5.
  • all adenovirus type 5 nucleotide numbers are relative to the NCBI reference sequence AC_000008.1, which is depicted herein in SEQ ID NO: 5.
  • the adenovirus replication cycle has two phases: an early phase, during which 4 transcription units (El, E2, E3, and E4) are expressed, and a late phase which occurs after the onset of viral DNA synthesis, and during which late transcripts are expressed primarily from the major late promoter (MLP).
  • MLP major late promoter
  • the late messages encode most of the virus's structural proteins.
  • the gene products of El, E2 and E4 are responsible for transcriptional activation, cell transformation, viral DNA replication, as well as other viral functions, and are necessary tor viral growth.
  • operably linked refers to a linkage of polynucleotide elements in a functional relationship.
  • a nucleic acid sequence is "operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • a promoter or enhancer is operably linked to a gene if it affects the transcription of the gene.
  • Operably linked nucleotide sequences are typically contiguous. However, as enhancers generally function when separated from the promoter by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably linked but not directly flanked and may even function in trans from a different allele or chromosome.
  • the virus has one or more modifications to a regulatory sequence or promoter.
  • a modification to a regulatory sequence or promoter comprises a deletion, substitution, or addition of one or more nucleotides compared to the wild-type sequence of the regulatory sequence or promoter.
  • the modification of a regulatory sequence or promoter comprises a modification of sequence of a transcription factor binding site to reduce affinity for the transcription factor, for example, by deleting a portion thereof, or by inserting a single point mutation into the binding site.
  • the additional modified regulatory sequence enhances expression in neoplastic cells, but attenuates expression in normal cells.
  • the modified regulatory sequence is operably linked to a sequence encoding a protein.
  • at least one of the adenoviral Ela and Elb genes (coding regions) is operably linked to a modified regulatory sequence.
  • the Ela gene is operably linked to the modified regulatory sequence.
  • the Ela regulatory sequence contains five binding sites for the transcription factor Pea3, designated Pea3 I, Pea3 II, Pea3 III, Pea3 IV, and Pea3 V, where Pea3 I is the Pea3 binding site most proximal to the Ela start site, and Pea3 V is most distal.
  • the Ela regulatory sequence also contains binding sites for the transcription factor E2F, hereby designated E2F I and E2F II, where E2F I is the E2F binding site most proximal to the Ela start site, and E2F II is more distal. From the Ela start site, the binding sites are arranged: Pea3 I, E2F I, Pea3 II, E2F II, Pea3 III, Pea3 IV, and Pea3 V.
  • At least one of these seven binding sites, or a functional portion thereof, is deleted.
  • a "functional portion” is a portion of the binding site that, when deleted, decreases or even eliminates the functionality, e.g. binding affinity, of the binding site to its respective transcription factor (Pea3 or E2F) by, for example, at least 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100% relative to the complete sequence.
  • one or more entire binding sites are deleted.
  • a functional portion of one or more binding sites is deleted.
  • a "deleted binding site” encompasses both the deletion of an entire binding site and the deletion of a functional portion thereof. When two or more binding sites are deleted, any combination of entire binding site deletion and functional portion deletion may be used.
  • At least one Pea3 binding site, or a functional portion thereof, is deleted.
  • the deleted Pea3 binding site can be Pea3 I, Pea3 II, Pea3 III, Pea3 IV, and/or Pea3 V.
  • the deleted Pea3 binding site is Pea3 II, Pea3 III, Pea3 IV, and/or Pea3 V.
  • the deleted Pea3 binding site is Pea3 IV and/or Pea3 V.
  • the deleted Pea3 binding site is Pea3 II and/or Pea3
  • the deleted Pea3 binding site is both Pea3 II and Pea3 III. In certain embodiments, the Pea3 I binding site, or a functional portion thereof, is retained.
  • At least one E2F binding site, or a functional portion thereof is deleted. In certain embodiments, at least one E2F binding site, or a functional portion thereof, is retained. In certain embodiments, the retained E2F binding site is E2F I and/or E2F II. In certain embodiments, the retained E2F binding site is E2F II. In certain embodiments, the total deletion consists essentially of one or more of Pea3 II, Pea3 III, Pea3
  • the virus has a deletion of a 50 base pair region located from -304 to -255 upstream of the Ela initiation site, e.g., corresponding to 195-244 of the Ad5 genome (SEQ ID NO: 5), hereafter referred to as the TAV-255 deletion.
  • the TAV-255 deletion results in an Ela promoter that comprises the sequence GGTGTTTTGG (SEQ ID NO: 10).
  • the virus comprises an Ela promoter having a deletion of a functional TATA box, e.g., the deletion of an entire TATA box.
  • a “functional TATA box” refers to a TATA box that is capable of binding to a TATA box binding protein (TBP), e.g., a TATA box that has at least 100%, at least 90%, at least 80%, at least 70%, at least 60%, at least 50%, or at least 40%, of the TBP binding activity of a corresponding wild-type TATA box sequence.
  • TBP TATA box binding protein
  • a“non-functional TATA box” refers to a TATA box that, e.g., has less than 30%, less than 20%, less than 10%, or 0% of the TBP binding activity of a corresponding wild-type TATA box sequence.
  • Assays for determining whether a TBP binds to a TATA box are known in the art. Exemplary binding assays include electrophoretic mobility shift assays, chromatin immunoprecipitation assays, and DNAse footprinting assays.
  • the virus comprises a deletion of nucleotides corresponding to -27 to -24, -31 to -24, -44 to +54, or -146 to +54 of the adenovirus type 5 Ela promoter, which correspond, respectively, to nucleotides 472 to 475, 468 to 475, 455 to 552, and 353 to 552 of the Ad5 genome (SEQ ID NO: 5).
  • the virus comprises a deletion of nucleotides corresponding to -29 to -26, -33 to -26, -44 to +52, or -148 to +52 of the adenovirus type 5 Ela promoter.
  • the virus comprises a deletion of nucleotides corresponding to 353 to 552 of the Ad5 genome (SEQ ID NO: 5).
  • the virus comprises a polynucleotide deletion that results in an adenovirus comprising the sequence CTAGGACTG (SEQ ID NO: 11), AGTGCCCG (SEQ ID NO: 19), or TATTCCCG (SEQ ID NO: 20), which result from joining the two polynucleotide sequences that would otherwise flank the deleted polynucleotide sequence.
  • the virus comprises a polynucleotide deletion that results in an adenovirus comprising the sequence CTAGGACTG (SEQ ID NO: 11) .
  • the virus comprises an Ela promoter having a deletion of a functional CAAT box, e.g., the deletion of an entire CAAT box.
  • a “functional CAAT box” refers to a CAAT box that is capable of binding to a C/EBP or NF-Y protein, e.g., a CAAT box that has at least 100%, at least 90%, at least 80%, at least 70%, at least 60%, at least 50%, or at least 40%, of the a C/EBP or NF-Y binding activity of a corresponding wild-type CAAT box sequence.
  • a“non-functional CAAT box” refers to a CAAT box that, e.g., has less than 30%, less than 20%, less than 10%, or 0% of the a C/EBP or NF-Y binding activity of a corresponding wild-type CAAT box sequence.
  • Assays for determining whether a C/EBP or NF-Y protein binds to a CAAT box are known in the art. Exemplary binding assays include electrophoretic mobility shift assays, chromatin immunoprecipitation assays, and DNAse footprinting assays.
  • the virus comprises a deletion of nucleotides corresponding to -76 to -68 of the adenovirus type 5 Ela promoter, which corresponds to nucleotides 423 to 431 of the Ad5 genome (SEQ ID NO: 5).
  • the virus comprises a polynucleotide deletion that results in an adenovirus comprising the sequence TTCCGTGGCG (SEQ ID NO: 21), which results from joining the two polynucleotide sequences that would otherwise flank the deleted polynucleotide sequence.
  • the adenoviral Elb-l9k gene functions primarily as an anti-apoptotic gene and is a homolog of the cellular anti-apoptotic gene, BCL-2. Since host cell death prior to maturation of the progeny viral particles would restrict viral replication, Elb-l9k is expressed as part of the El cassette to prevent premature cell death thereby allowing the infection to proceed and yield mature virions. Accordingly, in certain embodiments, a recombinant virus is provided that includes an Elb-l9K insertion site, e.g., the adenovirus has an exogenous nucleotide sequence inserted into an Elb-l9K insertion site.
  • the Elb-l9K insertion site is located between the start site of Elb-l9K (i.e., the nucleotide sequence encoding the start codon of Elb-l9k, e.g., corresponding to nucleotides 1714-1716 of SEQ ID NO: 5) and the start site of Elb-55K (i.e., the nucleotide sequence encoding the start codon of Elb-55k, e.g., corresponding to nucleotides 2019-2021 of SEQ ID NO: 5).
  • an insertion between two sites for example, an insertion between (i) a start site of a first gene (e.g., Elb-l9k) and a start site of a second gene, (e.g., Elb-55K), (ii) a start site of a first gene and a stop site of a second gene, (iii) a stop site of a first gene and start site of a second gene, or (iv) a stop site of first gene and a stop site of a second gene, is understood to mean that all or a portion of the nucleotides constituting a given start site or a stop site surrounding the insertion may be present or absent in the final virus. Similarly, an insertion between two nucleotides is understood to mean that the nucleotides surrounding the insertion may be present or absent in the final virus.
  • the Elb-l9K insertion site is located between the start site of Elb-l9K (i.e., the nucleotide sequence encoding the start codon of Elb-l9k, e.g., corresponding to nucleotides 1714-1716 of SEQ ID NO: 5) and the stop site of Elb-l9K (i.e., the nucleotide sequence encoding the stop codon of Elb-l9k, e.g., corresponding to nucleotides 2242-2244 of SEQ ID NO: 5).
  • start site of Elb-l9K i.e., the nucleotide sequence encoding the start codon of Elb-l9k, e.g., corresponding to nucleotides 1714-1716 of SEQ ID NO: 5
  • the stop site of Elb-l9K i.e., the nucleotide sequence encoding the stop codon of Elb-l9k, e.g., corresponding
  • the Elb-l9K insertion site comprises a deletion of from about 100 to about 305, about 100 to about 300, about 100 to about 250, about 100 to about 200, about 100 to about 150, about 150 to about 305, about 150 to about 300, about 150 to about 250, or about 150 to about 200 nucleotides adjacent the start site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion of about 200 nucleotides, e.g., 203 nucleotides adjacent the start site of Elb-l9K.
  • the Elb-l9K insertion site comprises a deletion corresponding to nucleotides 1714-1916 of the Ad5 genome (SEQ ID NO: 5), or the exogenous nucleotide sequence is inserted between nucleotides corresponding to 1714 and 1916 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between CTGACCTC (SEQ ID NO: 6) and TCACCAGG (SEQ ID NO: 7), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, CTGACCTC (SEQ ID NO: 6), the exogenous nucleotide sequence, and TCACCAGG (SEQ ID NO: 7).
  • CTGACCTC (SEQ ID NO: 6) and TCACCAGG (SEQ ID NO: 7) define unique boundary sequences for the Elb- 19K insertion site within the Ad5 genome (SEQ ID NO: 5).
  • a deletion adjacent to a site for example, a deletion adjacent to a start site of a gene or a deletion adjacent to a stop site of a gene, is understood to mean that the deletion may include a deletion of all, a portion, or none of the nucleotides constituting a given start site or a stop site.
  • an exogenous nucleotide sequence is inserted into an E3 insertion site located between the stop site of pVIII (i.e., the nucleotide sequence encoding the stop codon of pVIII, e.g., corresponding to nucleotides 27855-27857 of SEQ ID NO: 5) and the start site of Fiber (i.e., the nucleotide sequence encoding the start codon of Fiber, e.g., corresponding to nucleotides 31042-31044 of SEQ ID NO: 5).
  • the E3 insertion site comprises a deletion of from about 500 to about 3185, from about 500 to about 3000, from about 500 to about 2500, from about 500 to about 2000, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 3185, from about 1000 to about 3000, from about 1000 to about 2500, from about 1000 to about 2000, from about 1000 to about 1500, from about 1500 to about 3185, from about 1500 to about 3000, from about 1500 to about 2000, from about 2000 to about 3185, from about 2000 to about 3000, from about 2000 to about 2500, from about 2500 to about 3185, from about 2500 to about 3000, or from about 3000 to about 3185 nucleotides.
  • the E3 insertion site is located between the stop site of E3-10.5K (i.e., the nucleotide sequence encoding the stop codon of E3-10.5K, e.g., corresponding to nucleotides 29770-29772 of SEQ ID NO: 5) and the stop site of E3-14.7K (i.e., the nucleotide sequence encoding the stop codon of E3-14.7K, e.g., corresponding to nucleotides 30837-30839 of SEQ ID NO: 5).
  • the E3 insertion site comprises a deletion of from about 500 to about 1551, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 1551, from about 1000 to about 1500, or from about 1500 to about 1551 nucleotides adjacent the stop site of E3-10.5K.
  • the E3 insertion site comprises a deletion of about 1050 nucleotides adjacent the stop site of E3-10.5K, e.g., the E3 insertion site comprises a deletion of 1063 or 1064 nucleotides adjacent the stop site of E3-10.5K.
  • the E3 insertion site comprises a deletion corresponding to the Ad5 dl309 E3 deletion.
  • the E3 insertion site comprises a deletion corresponding to nucleotides 29773-30836 of the Ad5 genome (SEQ ID NO: 5), or the exogenous nucleotide sequence is inserted between nucleotides corresponding to 29773 and 30836 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between CAGTATGA (SEQ ID NO: 8) and TAATAAAAAA (SEQ ID NO: 9), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, CAGTATGA (SEQ ID NO: 8), the exogenous nucleotide sequence, and TAATAAAAAA (SEQ ID NO: 9).
  • CAGTATGA (SEQ ID NO: 8) and TAATAAAAAA (SEQ ID NO: 9) define unique boundary sequences for an E3 insertion site within the Ad5 genome (SEQ ID NO: 5).
  • the E3 insertion site is located between stop site of E3- gpl9K (i.e.. the nucleotide sequence encoding the stop codon of E3-gpl9K, e.g., corresponding to nucleotides 29215-29217 of SEQ ID NO: 5) and the stop site of E3-14.7K (i.e., the nucleotide sequence encoding the stop codon of E3-14.7K, e.g., corresponding to nucleotides 30837-30839 of SEQ ID NO: 5).
  • stop site of E3- gpl9K i.e.. the nucleotide sequence encoding the stop codon of E3-gpl9K, e.g., corresponding to nucleotides 29215-29217 of SEQ ID NO: 5
  • the stop site of E3-14.7K i.e., the nucleotide sequence encoding the stop codon of E3-14.7K, e.
  • the E3 insertion site comprises a deletion of from about 500 to about 1824, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 1824, from about 1000 to about 1500, or from about 1500 to about 1824 nucleotides adjacent the stop site of E3-gpl9K. In certain embodiments, the E3 insertion site comprises a deletion of about 1600 nucleotides adjacent the stop site of E3-gpl9K. e.g., the E3 insertion site comprises a deletion of 1622 nucleotides adjacent the stop site of E3-gpl9K.
  • the E3 insertion site comprises a deletion corresponding to nucleotides 29218-30839 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between nucleotides corresponding to 29218 and 30839 of the Ad5 genome (SEQ ID NO: 5).
  • the exogenous nucleotide sequence is inserted between TGCCTTAA (SEQ ID NO: 17) and TAAAAAAAAAT (SEQ ID NO: 18), e.g., the recombinant adenovirus comprises, in a 5’ to 3’ orientation, TGCCTTAA (SEQ ID NO: 17), the exogenous nucleotide sequence, and TAAAAAAAAAT (SEQ ID NO: 18).
  • TGCCTTAA (SEQ ID NO: 17) and TAAAAAAAAAT (SEQ ID NO: 18) define unique boundary sequences for an E3 insertion site within the Ad5 genome (SEQ ID NO: 5).
  • the vims comprises an E4 deletion.
  • the E4 deletion is located between the start site of E4-ORF6/7 (i.e., the nucleotide sequence encoding the start codon of E4-ORF6/7, e.g., corresponding to nucleotides 34075-34077 of SEQ ID NO: 5) and the right inverted terminal repeat (ITR; e.g., corresponding to nucleotides 35836-35938 of SEQ ID NO: 5).
  • the E4 deletion is located between the start site of E4-ORF6/7 and the start site of E4-ORF1 (i.e..
  • the E4 deletion comprises a deletion of a nucleotide sequence between the start site of E4-ORF6/7 and the start site of E4-ORF1.
  • the E4 deletion comprises a deletion of from about 500 to about 2500, from about 500 to about 2000, from about 500 to about 1500, from about 500 to about 1000, from about 1000 to about 2500, from about 1000 to about 2000, from about 1000 to about 1500, from about 1500 to about 2500, from about 1500 to about 2000, or from about 2000 to about 2500 nucleotides.
  • the E4 deletion comprises a deletion of from about 250 to about 1500, from about 250 to about 1250, from about 250 to about 1000, from about 250 to about 750, from about 250 to about 500, from 500 to about 1500, from about 500 to about 1250, from about 500 to about 1000, from about 500 to about 750, from 750 to about 1500, from about 750 to about 1250, from about 750 to about 1000, from about 1000 to about 1500, or from about 1000 to about 1250 nucleotides adjacent the start site of E4-ORF6/7.
  • the E4 deletion comprises a deletion of about 1450 nucleotides adjacent the start site of E4-ORF6/7, e.g., the E4 deletion comprises a deletion of about 1449 nucleotides adjacent the start site of E4-ORF6/7. In certain embodiments, the E4 deletion comprises a deletion corresponding to nucleotides 34078-35526 of the Ad5 genome (SEQ ID NO: 5).
  • the virus includes an IX-E2 insertion site, e.g., the virus has an exogenous nucleotide sequence inserted into an IX-E2 insertion site.
  • the IX-E2 insertion site is located between the nucleotide sequence encoding the stop codon of IX and the nucleotide sequence encoding the stop codon of IVa2.
  • the nucleotide sequence is inserted between nucleotides corresponding to 4029 and 4093 of the Ad5 genome (SEQ ID NO: 5).
  • the nucleotide sequence is inserted between nucleotides corresponding to 4029 and 4050, nucleotides corresponding to 4051 and 4070, or nucleotides corresponding to 4071 and 4093 of the Ad5 genome (SEQ ID NO: 5).
  • the IX-E2 insertion site comprises a deletion of about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, or 60 nucleotides.
  • the virus includes an L5-E4 insertion site, e.g., the virus has an exogenous nucleotide sequence inserted into an L5-E4 insertion site.
  • the L5-E4 insertion site is located between the nucleotide sequence encoding the stop codon of Fiber and the nucleotide sequence encoding the stop codon of E4-ORF6 or E40RF6/7.
  • the nucleotide sequence is inserted between nucleotides corresponding to 32785 to 32916 of the Ad5 genome (SEQ ID NO: 5).
  • the nucleotide sequence is inserted between nucleotides corresponding to 32785 and 32800, nucleotides corresponding to 32801 and 32820, nucleotides corresponding to 32821 and 32840, nucleotides corresponding to 32841 and 32860, nucleotides
  • the L5-E4 insertion site comprises a deletion of about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, or 130 nucleotides.
  • the recombinant virus comprises a nucleotide sequence of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, or SEQ ID NO: 15, or a nucleotide sequence having 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, or SEQ ID NO: 15.
  • Sequence identity may be determined in various ways that are within the skill in the art, e.g., using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software.
  • BLAST Basic Local Alignment Search Tool
  • analysis using the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx (Karlin et al, ( 1990) PROC. NATL. ACAD. SCI. USA 87:2264-2268; Altschul, (1993) J. MOL. EVOL. 36, 290-300; Altschul et al., (1997) NUCLEIC ACIDS RES.
  • BLOSUM62 matrix (Henikoff et al, (1992) PROC. NATL. ACAD. SCI. USA 89: 10915-10919, fully incorporated by reference).
  • a recombinant virus is an oncolytic virus, e.g., a virus that exhibits tumor-selective replication and/or viral mediated lysis.
  • a recombinant virus of the invention exhibits selective expression of a protein, e.g., a protein defining a binding site or the protein that mediates the production of a biomolecule defining a binding site therapeutic transgene in a hyperproliferative cell, e.g., a cancer cell, relative to a non-hyperprobferative cell.
  • the expression of the protein in a non- hyperproliferative cell is about 90%, about 80%, about 70%, about 60%, about 50%, about 40%, about 30%, about 20%, about 10% , or about 5% of the expression of in a
  • the hyperproliferative cell may be a cancer cell, e.g., a carcinoma, sarcoma, leukemia, lymphoma, prostate cancer, lung cancer, gastrointestinal tract cancer, colorectal cancer, pancreatic cancer, breast cancer, ovarian cancer, cervical cancer, stomach cancer, thyroid cancer, mesothelioma, liver cancer, kidney cancer, skin cancer, head and neck cancer, or brain cancer cell.
  • a cancer cell e.g., a carcinoma, sarcoma, leukemia, lymphoma, prostate cancer, lung cancer, gastrointestinal tract cancer, colorectal cancer, pancreatic cancer, breast cancer, ovarian cancer, cervical cancer, stomach cancer, thyroid cancer, mesothelioma, liver cancer, kidney cancer, skin cancer, head and neck cancer, or brain cancer cell.
  • a disclosed virus is produced in a suitable host cell line using conventional techniques including culturing a transfected or infected host cell under suitable conditions so as to allow the production of infectious viral particles.
  • Nucleic acids encoding viral genes can be incorporated into plasmids and introduced into host cells through conventional transfection or transformation techniques.
  • Exemplary suitable host cells for production of disclosed viruses include human cell lines such as HeLa, Hela-S3, HEK293, 911, A549, HER96, or PER-C6 cells. Specific production and purification conditions will vary depending upon the virus and the production system employed.
  • adenovirus For adenovirus, the traditional method for the generation of viral particles is co-transfection followed by subsequent in vivo recombination of a shuttle plasmid (usually containing a small subset of tire adenoviral genome and optionally containing a potential transgene an expression cassette) and an adenoviral helper plasmid (containing most of the entire adenoviral genome).
  • a shuttle plasmid usually containing a small subset of tire adenoviral genome and optionally containing a potential transgene an expression cassette
  • adenoviral helper plasmid containing most of the entire adenoviral genome
  • Alternative technologies for the generation of adenovirus include utilization of the bacterial artificial chromosome (BAG) system, in vivo bacterial recombination in a recAT bacterial strain utilizing two plasmids containing complementary adenoviral sequences, and the yeast artificial chromosome (YAC) system.
  • BAG bacterial artificial chromosome
  • YAC yeast artificial chromosome
  • infectious viral particles are recovered from the culture and optionally purified.
  • Typical purification steps may include plaque purification, centrifugation, e.g., cesium chloride gradient centrifugation, clarification, enzymatic treatment, e.g., benzonase or protease treatment, chromatographic steps, e.g., ion exchange chromatography or filtration steps.
  • the invention provides methods and compositions comprising: (a) an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding: (i) a protein defining a binding site; or (ii) a protein that mediates the production of a biomolecule defining a binding site; and (b) an effective amount of a therapeutic agent that binds to the binding site.
  • the term“therapeutic agent that binds to a binding site” refers to any therapeutic agent (/. e. , any agent that imparts a therapeutic effect in a target cell, body fluid, tissue, organ, physiological system, or subject) that reacts or associates more frequently, more rapidly, with greater duration and/or with stronger affinity with a particular binding site (e.g., a binding site on a protein encoded by a delivery vehicle or a binding site on a biomolecule the production of which is mediated by a protein encoded by a delivery vehicle, e.g., bone tissue) than it does with an alternative binding site.
  • a therapeutic agent e. , any agent that imparts a therapeutic effect in a target cell, body fluid, tissue, organ, physiological system, or subject
  • a particular binding site e.g., a binding site on a protein encoded by a delivery vehicle or a binding site on a biomolecule the production of which is mediated by a protein encoded by a delivery vehicle, e.g.,
  • the therapeutic agent may, e.g., have affinity for the binding site stronger than 100 nM, 50 nM, 20 nM, 15 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM, as determined by surface plasmon resonance.
  • a biomolecule defining a binding site is understood to contemplate an individual biomolecule, e.g., a protein, and/or a collection of biomolecules, e.g., a tissue, e.g., bone tissue.
  • the protein that defines a binding site is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR) and prostate specific membrane antigen (PSMA), and/or the therapeutic agent that binds to the protein is selected from an anti-BMP antibody, an anti-BMPR antibody, and an anti-PSMA antibody.
  • BMP bone morphogenetic protein
  • BMPR bone morphogenetic protein receptor
  • PSMA prostate specific membrane antigen
  • the BMP may, for example, be selected from BMP-2, BMP-4, BMP-6 and BMP- 7, and accordingly, the anti-BMP antibody may, for example, be selected from an anti-BMP - 2 antibody, an anti-BMP -4 antibody, an anti-BMP-6 antibody, and an anti-BMP-7 antibody.
  • the therapeutic agent is an antibody.
  • antibodies are multimeric proteins that contain four polypeptide chains. Two of the polypeptide chains are called immunoglobulin heavy chains (H chains), and two of the polypeptide chains are called immunoglobulin light chains (L chains).
  • the immunoglobulin heavy and light chains are connected by an interchain disulfide bond.
  • the immunoglobulin heavy chains are connected by interchain disulfide bonds.
  • a light chain consists of one variable region (V L ) and one constant region (C L ).
  • the heavy chain consists of one variable region (V H ) and at least three constant regions (CHi, CH 2 and CH 3 ).
  • the variable regions determine the binding specificity of the antibody. [0090]
  • Each variable region contains three hypervariable regions known as
  • CDRs complementarity determining regions flanked by four relatively conserved regions known as framework regions (FRs).
  • FRs framework regions
  • the extent of the FRs and CDRs has been defined (Rabat, E.A., et al. (1991) SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, FIFTH EDITION, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; and Chothia, C. et al. (1987) J. MOL. BIOL. 196:901-917).
  • the three CDRs referred to as CDRi, CDR 2 , and CDR3, contribute to the antibody binding specificity.
  • Naturally occurring antibodies have been used as starting material for engineered antibodies, such as chimeric antibodies and humanized antibodies.
  • antibody means an intact antibody (e.g., an intact monoclonal antibody) or antigen-binding fragment of an antibody, including an intact antibody or antigen-binding fragment that has been modified, engineered or chemically conjugated.
  • antibodies that have been modified or engineered are chimeric antibodies, humanized antibodies, and multispecific antibodies (e.g., bispecific antibodies).
  • antigen-binding fragments include Fab, Fab’, F(ab’)2, Fv, single chain antibodies (e.g., scFv), minibodies, and diabodies.
  • an antibody conjugated to a toxin, radioisotope, cytokine, or enzyme is an example of a chemically conjugated antibody.
  • the antibody the antibody is selected from an antibody drug conjugate and an antibody radionuclide conjugate.
  • an antibody may mediate an“effector functions,” e.g., a biological activity attributable to the Fc region of an antibody that varies with the antibody isotype.
  • effector functions include Clq binding and complement dependent cytotoxicity, Fc receptor binding, antibody-dependent cell mediated cytotoxicity (ADCC), phagocytosis, down regulation of cell surface receptors (e.g., B cell receptors); and B cell activation.
  • the therapeutic agent is a cell, e.g., a T-cell.
  • the T-cell can be any T-cell, such as a cultured T-cell, e.g., a primary T-cell, or a T-cell from a cultured T-cell line, e.g., Jurkat, SupTi, etc., or a T-cell obtained from a mammal. If obtained from a mammal, the T-cell can be obtained from numerous sources, including but not limited to blood, bone marrow, lymph node, the thymus, or other tissues or fluids. T-cells can also be enriched for or purified.
  • the T-cell is a human T-cell, which can be an autologous or heterologous cell.
  • the T-cell can be any type of T-cell and can be of any developmental stage, including but not limited to, CD4+/CD8+ double positive T-cells, CD4+ helper T-cells, e.g., Thl and Th2 cells, CD4+ T-cells, CD8+ T-cells (e.g., cytotoxic T-cells), tumor infiltrating lymphocytes (TILs), memory T-cells (e.g., central memory T-cells and effector memory T-cells), naive T-cells, and the like.
  • the cells can include autologous cells derived from a subject to be treated, or alternatively allogenic cells derived from a donor.
  • a T-cell binds to a binding site through a T-cell receptor.
  • the T-cell receptor may be an endogenous or a recombinant T-cell receptor.
  • T-cell receptors comprise two chains referred to as the a- and b-chains, that form a pair on the surface of a T- cell to form a heterodimeric receptor.
  • the T-cell receptor is involved in recognition of MHC- restricted antigens.
  • Each of a- and b- chain comprises two regions, a constant region and a variable region.
  • Each variable region of the a- and b- chains defines three loops, referred to as complementary determining regions (CDRs) known as CDRi, CDR 2 , and CDR 3 that confer the T-cell receptor with antigen binding activity and binding specificity.
  • CDRs complementary determining regions
  • a T-cell binds to a binding site through a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • CARs refers to any artificial receptor including an antigen-specific binding moiety and one or more signaling chains derived from an immune receptor.
  • CARs comprise a single chain fragment variable (scFv) of an antibody specific for an antigen coupled via hinge and transmembrane regions to cytoplasmic domains of T-cell signaling molecules, e.g. a T-cell costimulatory domain (e.g., from CD28, CD 137, 0X40, ICOS, and CD27) in tandem with a T-cell triggering domain (e.g. from CD3z).
  • a T-cell expressing a chimeric antigen receptor is referred to as a CAR T-cell.
  • the protein that mediates the production of a biomolecule defining a binding site is selected from a bone morphogenetic protein (BMP), a bone morphogenetic protein receptor (BMPR), osteocalcin, osteopontin, bone sialoprotein, osteonectin, osteoprotegerin (OPG) and alkaline phosphatase and/or the biomolecule defining a binding site is bone tissue, e.g., expression of a BMP, a BMPR, osteocalcin, osteopontin, bone sialoprotein, osteonectin, OPG or alkaline phosphatase by the delivery vehicle mediates the production of bone tissue.
  • BMP bone morphogenetic protein
  • BMPR bone morphogenetic protein receptor
  • OPG osteoprotegerin
  • alkaline phosphatase alkaline phosphatase
  • the biomolecule defining a binding site is bone tissue, e.g., expression of a BMP, a BMPR, osteocalcin, osteopontin, bone si
  • the BMP may, for example, be selected from BMP-2, BMP-4, BMP-6 or BMP-7.
  • expression of a BMP, a BMPR, osteocalcin, osteopontin, bone sialoprotein, osteonectin, OPG or alkaline phosphatase by the delivery vehicle in a cancer cell results in the cancer cell acquiring an osteoblast-like cell phenotype.
  • the therapeutic agent that binds to a biomolecule, e.g., bone is a radionuclide, i.e., an atom that emits radiation and, e.g., kills surrounding cancer cells.
  • Radionuclides may, e.g., be g-emitting radionuclides, b-emitting radionuclides, a- emitting radionuclides, and positron-emitting radionuclides.
  • Exemplary radionuclides include 32 P, 47 Sc, 64 Cu, 67 Cu, 89 Sr, 86 Y, 87 Y, 90 Y, 105 Rh, m Ag, m In, 117m Sn, 149 Pm, 153 Sm, 166 Ho, 177 Lu, I S6 RC. 188 Re, 211 At, 212 Bi, and 223 Ra.
  • the radionuclide is a bone targeting (i.e., a bone-homing) radionuclide.
  • a bone-targeting radionuclide is a calcium mimetic (e.g., Sr and Ra).
  • a bone homing radionuclide is conjugated to a ligand (e.g., ethylenediaminetetramethylene phosphonate (EDTMP), e.g., 153 Sm-EDTMP), which binds to calcium.
  • a ligand e.g., ethylenediaminetetramethylene phosphonate (EDTMP), e.g., 153 Sm-EDTMP
  • the radionuclide is selected from 89 Sr (e.g., 89 Sr chloride, i.e., MetastronTM),
  • 223 Ra e.g., 223 Ra dichloride, i.e., Xofigo ®
  • 153 Sm e.g., 153 Sm-EDTMP, i.e., 153 Sm
  • lexidronam i.e., Quadramet ®
  • 177 Lu 90 Y
  • 186 Re e.g, 186 Re HEDP
  • 117m Sn e.g, 117m Sn DTP A
  • 32 P 32 P.
  • a bone morphogenetic protein may, for example, be selected from BMP-2, BMP -4, BMP-6 or BMP-7.
  • An exemplary nucleotide sequence of BMP -2 cloned into an exemplary recombinant adenovirus delivery vehicle is as follows, where the flanking adenoviral sequence including restriction sites is underlined:
  • An exemplary nucleotide sequence of BMP -4 cloned into an exemplary recombinant adenovirus delivery vehicle is as follows, where the flanking adenoviral sequence including restriction sites is underlined:
  • An exemplary nucleotide sequence of BMP-6 cloned into an exemplary recombinant adenovirus delivery vehicle is as follows, where the flanking adenoviral sequence including restriction sites is underlined: ATCTGACCTCGTCGACATGCCGGGGCTGGGGCGGAGGGCGCAGTGGCTGTGCTGGTGGTGGG GGCTGCTGTGCAGCTGCTGCGGGCCCCCGCCGCTGCGGCCGCCCTTGCCCGCTGCCGCGGCC GCCGGGGGGCAGCTGCTGGGGGACGGCGGGAGCCCCGGCCGCACGGAGCAGCCGCC GCCGTCGCCGCAGTCCTCCTCGGGCTTCCTGTACCGGCGGCTCAAGACGCAGGAGAAGCGGG AGATGCAGAAGGAGATCTTGTCGGTGCTGGGGCTCCCGCACCGGCCCCGGCCCCTGCACGGC CTCCAACAGCCGCAGCCCCCGGCGCTCCGGCAGCAGGAGGAGCAGCAGCAGCAGCT GCCTCGCGGAGCT GCCTCGCGGAGCT G
  • An exemplary nucleotide sequence of BMP-7 cloned into an exemplary recombinant adenovirus delivery vehicle is as follows, where the flanking adenoviral sequence including restriction sites is underlined:
  • a BMP comprises SEQ ID NO: 1 or a functional fragment thereof, SEQ ID NO: 2 or a functional fragment thereof, SEQ ID NO: 3 or a functional fragment thereof, or SEQ ID NO: 4 or a functional fragment thereof, or an amino acid sequence having 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
  • a functional fragment of a BMP refers to a fragment that exhibits at least 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of the activity, e.g., BMP receptor binding activity, of a corresponding full length BMP protein.
  • a disclosed delivery vehicle may further comprise an exogenous nucleotide sequence that encodes for a therapeutic transgene.
  • a therapeutic transgene may encode a therapeutic nucleic acid, e.g., an antisense RNA or ribozyme RNA.
  • the therapeutic transgene may encode a therapeutic peptide or polypeptide, e.g., a costimulatory molecule, an antiangiogenic and/or antivascular molecule, an anti-fibrotic molecule, an anti-inflammatory or pro-inflammatory molecule, a monoclonal antibody or fragment thereof, a pro-apoptotic molecule, an enzyme, an immunomodulatory molecule, a cytokine, a cytokine inhibitor (e.g., a cytokine trap), an oncoprotein, a tumor suppressor, a lytic peptide, a vaccine antigen, and a molecule which complements genetic defects in somatic cells
  • a therapeutic peptide or polypeptide e.g., a costimulatory molecule, an antiangiogenic and/or antivascular molecule, an anti-fibrotic molecule, an anti-inflammatory or pro-inflammatory molecule, a monoclonal antibody or fragment thereof, a pro-apoptotic molecule, an enzyme, an immunomodulatory molecule
  • the therapeutic transgene encodes a therapeutic polypeptide, or a fragment thereof, selected from acetylcholine, an androgen-receptor, an anti-PD-l antibody heavy chain and/or light chain, an anti-PD-Ll antibody heavy chain and/or light chain, BORIS/CTCFL, BRAF, CD19, CD20, CD30, CD80, CD86, CD137, CD137L, CD154, CEA, DKKl/Wnt, EGFRvIII, FGF, gplOO, Her-2/neu, ICAM, IL-l, IL-3, IL-4, IL-5, IL-6, IL-8, IL-9, IL-l 7, IL-23A/pl9, p40, IL-24, IL-27, IL-27A/p28, IL- 27B/EBI3, IL-35, interferon-gamma, KRAS, MAGE, MAGE-A3, MART1, melan-A
  • the therapeutic transgene encodes a cancer antigen derived from 9D7, androgen receptor, a BAGE family protein, b-catenin, BING-4, BRAF, BRCA1/2, a CAGE family protein, calcium-activated chloride channel 2, CD 19, CD20, CD30, CDK4, CEA, CML66, CT9, CT10, cyclin-Bl, EGFRvIII, Ep-CAM, EphA3, fibronectin, a GAGE family protein, gpl00/pmell7, Her-2/neu, HPV E6, HPV E7, Ig, immature laminin receptor, a MAGE family protein (e.g., MAGE-A3), MART- 1 /melan-A, MART2, MC1R, mesothelin, a mucin family protein (e.g., MUC-l), NY-ESO-l/LAGE-l, P.polypeptide, p53, podocalyx
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent preferably is combined with a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier means buffers, carriers, and excipients suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the carrier(s) should be“acceptable” in the sense of being compatible with the other ingredients of the formulations and not deleterious to the recipient.
  • Pharmaceutically acceptable carriers include buffers, solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is known in the art.
  • compositions containing recombinant viruses disclosed herein can be presented in a dosage unit form and can be prepared by any suitable method.
  • a pharmaceutical composition should be formulated to be compatible with its intended route of administration. Examples of routes of administration are intravenous (IV), intradermal, inhalation, intraocular, intranasal, transdermal, topical, transmucosal, rectal, oral, parenteral, subcutaneous, intramuscular, ophthalmic, epidural, intratracheal, sublingual, buccal, vaginal, and nasal administration.
  • An exemplary route of administration is IV infusion.
  • Useful formulations can be prepared by methods known in the pharmaceutical art. For example, see Remington's Pharmaceutical Sciences, 18th ed. (Mack Publishing Company, 1990).
  • Formulation components suitable for parenteral administration include a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as EDTA;
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS).
  • the carrier should be stable under the conditions of manufacture and storage, and should be preserved against microorganisms.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol), and suitable mixtures thereof.
  • compositions preferably are sterile. Sterilization can be accomplished by any suitable method, e.g., filtration through sterile filtration membranes. Where the composition is lyophilized, filter sterilization can be conducted prior to or following lyophilization and reconstitution.
  • an effective amount refers to the amount of an active component (e.g., the amount of a recombinant virus of the present invention) sufficient to effect beneficial or desired results.
  • An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • a therapeutically effective amount of active component is in the range of 0.1 mg/kg to 100 mg/kg, e.g., 1 mg/kg to 100 mg/kg, 1 mg/kg to 10 mg/kg, 1 mg/kg to 5 mg/kg, 10 mg/kg, 7.5 mg/kg, 5 mg/kg, or 2.5 mg/kg.
  • a therapeutically effective amount of a recombinant virus is in the range of 10 2 to 10 15 plaque forming units (pfiis), e.g., l0 2 to 10 10 , l0 2 to 10 5 , l0 5 to 10 15 , l0 5 to 10 10 , or l0 10 to l0 15 plaque forming units.
  • a therapeutically effective amount of a radionuclide virus is in the range of 1-500000 kBq/kg, e.g., 1-200000 kBq/kg, 1-150000 kBq/kg, 1-100000 kBq/kg, 1-50000 kBq/kg, 1-20000 kBq/kg, 1-10000 kBq/kg, 1-5000 kBq/kg, 1-2000 kBq/kg, 1-1000 kBq/kg, 1-500 kBq/kg, 1-100 kBq/kg, 1-50 kBq/kg, or 1-20 kBq/kg.
  • the amount administered will depend on variables such as the type and extent of disease or indication to be treated, the overall health of the subject, the in vivo potency of the virus, the
  • the initial dosage can be increased beyond the upper level in order to rapidly achieve the desired blood-level or tissue- level.
  • the initial dosage can be smaller than the optimum, and the daily dosage may be progressively increased during the course of treatment.
  • Human dosage can be optimized, e.g., in a conventional Phase I dose escalation study designed to run from 0.5 mg/kg to 20 mg/kg.
  • Dosing frequency can vary, depending on factors such as route of administration, dosage amount, the half-life of the recombinant virus, and the disease being treated. Exemplary dosing frequencies are once per day, once per week and once every two weeks.
  • a preferred route of administration is parenteral, e.g., intravenous infusion.
  • the methods and compositions disclosed herein can be used to treat various medical indications.
  • the methods and compositions disclosed herein can be used to treat cancers.
  • the cancer cells are exposed to a therapeutically effective amount of a delivery vehicle (e.g., a recombinant virus) and/or a therapeutic agent so as to inhibit or reduce proliferation of the cancer cells.
  • a delivery vehicle e.g., a recombinant virus
  • the invention provides a method of treating a cancer in a subject.
  • the method comprises administering to the subject an effective amount of a delivery vehicle and/or a therapeutic agent to treat the cancer in the subject.
  • administering an effective amount of a delivery vehicle and/or a therapeutic agent to a subject reduces tumor load in that subject by at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%.
  • the methods and compositions disclosed herein can be used to treat bone loss and/or hypercalcemia, e.g., bone loss and/or hypercalcemia associated with a cancer.
  • the invention provides a method of bone loss and/or hypercalcemia in a subject, e.g., a subject with cancer.
  • the method comprises administering to the subject an effective amount of a delivery vehicle comprising an exogenous nucleotide sequence encoding a protein that mediates the production of bone tissue to treat the bone loss and/or hypercalcemia in a subject.
  • “treat”,“treating” and“treatment” mean the treatment of a disease in a subject, e.g., in a human. This includes: (a) inhibiting the disease, i.e., arresting its development; and (b) relieving the disease, i.e., causing regression of the disease state.
  • the terms“subject” and“patient” refer to an organism to be treated by the methods and compositions described herein. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and more preferably includes humans.
  • Examples of cancers include solid tumors, soft tissue tumors, hematopoietic tumors and metastatic lesions.
  • hematopoietic tumors include, leukemia, acute leukemia, acute lymphoblastic leukemia (ALL), B-cell, T-cell or FAB ALL, acute myeloid leukemia (AML), chronic myelocytic leukemia (CML), chronic lymphocytic leukemia (CLL), e.g., transformed CLL, diffuse large B-cell lymphomas (DLBCL), follicular lymphoma, hairy cell leukemia, myelodyplastic syndrome (MDS), a lymphoma, Hodgkin's disease, a malignant lymphoma, non-Hodgkin's lymphoma, Burkitt's lymphoma, multiple myeloma, or Richter’s Syndrome (Richter’s Transformation).
  • solid tumors include malignancies, e.g., sarcomas, adenocarcinomas, and carcinomas, of the various organ systems, such as those affecting head and neck (including pharynx), thyroid, lung (small cell or non-small cell lung carcinoma (NSCLC)), breast, lymphoid, gastrointestinal (e.g., oral, esophageal, stomach, liver, pancreas, small intestine, colon and rectum, anal canal), genitals and genitourinary tract (e.g., renal, urothelial, bladder, ovarian, uterine, cervical, endometrial, prostate, testicular), CNS (e.g., neural or glial cells, e.g., neuroblastoma or glioma), or skin (e.g, melanoma).
  • malignancies e.g., sarcomas, adenocarcinomas, and carcinomas
  • various organ systems such as those
  • the cancer is selected from melanoma, squamous cell carcinoma of the skin, basal cell carcinoma, head and neck cancer, breast cancer, anal cancer, cervical cancer, non-small cell lung cancer, mesothelioma, small cell lung cancer, renal cell carcinoma, prostate cancer, gastroesophageal cancer, colorectal cancer, testicular cancer, bladder cancer, ovarian cancer, liver cancer, hepatocellular carcinoma, cholangiocarcinoma, brain and central nervous system cancer, thyroid cancer, parathyroid cancer (e.g, parathyroid carcinoma), endometrial cancer, neuroendocrine cancer, lymphoma (e.g., Hodgkin and non- Hodgkin), leukemia, merkel cell carcinoma, gastrointestinal stromal tumors, multiple myeloma, uterine cancer, a sarcoma, kidney cancer, ocular cancer, pancreatic cancer, and a germ cell cancer (e.g., ovarian germ cell cancer).
  • melanoma s
  • the invention also provides a method of generating bone tissue in a subject in need thereof.
  • the method comprises administering to the subject an effective amount of a delivery vehicle described herein to generate bone tissue in the subject.
  • the generation of bone tissue may, e.g., treat a non-union or fracture in the subject, or be used for a spinal fusion or dental implant in the subject.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent are administered to the subject in combination with one or more therapies, e.g., surgery, radiation, chemotherapy, immunotherapy, hormone therapy, or virotherapy.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent are administered to the subject in combination with a therapy that promotes bone strength and/or growth, e.g., an anti-resorptive therapy.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent are administered to the subject in combination with a bisphosphonate, testosterone, hyaluronic acid, vitamin D, calcium, testosterone, estrogen, progesterone, a selective estrogen modulator (SERM), calcitonin, a RANK or RANKL inhibitor (e.g., denosumab), pulsed ultrasound (e.g., low intensity pulsed ultrasound (LIPU)), electrical stimulation, extracorporeal shockwave therapy (ESWT), low energy laser therapy, and/or pulsed electromagnetic field (PEMF) therapy.
  • a bisphosphonate e.g., testosterone, hyaluronic acid
  • vitamin D calcium
  • testosterone, estrogen, progesterone e.g., progesterone
  • SERM selective estrogen modulator
  • calcitonin calcitonin
  • a RANK or RANKL inhibitor e.g., denosumab
  • pulsed ultrasound e.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent are administered to the subject in combination with a bisphosphonate.
  • exemplary bisphosphonates include disodium pamidronate, alendronate, etidronate, tiludronate, risedronate, zoledronic acid, sodium clodronate, and ibandronic acid.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent of the invention is administered in combination with a tyrosine kinase inhibitor, e.g., erlotinib.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent of the invention is administered in combination with a checkpoint inhibitor, e.g., an anti-CTLA-4 antibody, an anti -PD- 1 antibody, or an anti-PD-Ll antibody.
  • a checkpoint inhibitor e.g., an anti-CTLA-4 antibody, an anti -PD- 1 antibody, or an anti-PD-Ll antibody.
  • Exemplary anti-PD-l antibodies include, for example, nivolumab (Opdivo®, Bristol-Myers Squibb Co.), pembrolizumab (Keytruda®, Merck Sharp & Dohme Corp.), PDR001 (Novartis Pharmaceuticals), and pidilizumab (CT-011, Cure Tech).
  • Exemplary anti-PD-Ll antibodies include, for example, atezolizumab (Tecentriq®, Genentech), duvalumab (AstraZeneca), MEDI4736, avelumab, and BMS 936559 (Bristol Myers Squibb Co.).
  • the term administered "in combination,” as used herein, is understood to mean that two (or more) different treatments are delivered to the subject during the course of the subject’s affliction with the disorder, such that the effects of the treatments on the subject overlap at a point in time.
  • the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous" or “concurrent delivery.”
  • the delivery of one treatment ends before the delivery of the other treatment begins.
  • the treatment is more effective because of combined administration.
  • the second treatment is more effective, e.g.
  • delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive.
  • the delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • the effective amount of a delivery vehicle (e.g., a recombinant virus) and/or a therapeutic agent of the invention is identified by measuring an immune response to an antigen in the subject and/or the method of treating the subject further comprises measuring an immune response to an antigen in the subject.
  • a delivery vehicle e.g., a recombinant virus
  • a therapeutic agent of the invention is identified by measuring an immune response to an antigen in the subject and/or the method of treating the subject further comprises measuring an immune response to an antigen in the subject.
  • Hyperproliferative diseases e.g., cancers, may be characterized by immunosuppression, and measuring an immune response to an antigen in the subject may be indicative of the level of
  • measuring an immune response to an antigen in the subject may be indicative of the efficacy of the treatment and/or the effective amount of the recombinant virus.
  • the immune response to the antigen in the subject may be measured by any method known in the art.
  • the immune response to the antigen is measured by injecting the subject with the antigen at an injection site on the skin of the subject and measuring the size of an induration or amount of inflammation at the injection site.
  • the immune response to the antigen is measured by release of a cytokine from a cell of the subject (e.g., interferon gamma, IL-4 and/or IL-5) upon exposure to the antigen.
  • compositions, devices, and systems are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions, devices, and systems of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.
  • viruses, compositions, systems, processes and methods, or features thereof are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual subcombination of the members of such groups and ranges. By way of other examples, an integer in the range of 1 to 20 is specifically intended to individually disclose 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12,
  • This Example describes the construction of a recombinant adenovirus type 5 (Ad5) that expresses a protein that defines a binding site or a protein that mediates the production of a biomolecule that defines a binding site for a therapeutic agent, e.g., BMP -2 or BMP-7.
  • Ad5 a recombinant adenovirus type 5
  • a plasmid carrying the 5' portion of the adenovirus type 5 genomic sequence was modified to carry the deletion of a nucleotide region located from -304 to -255 upstream of the Ela initiation site, which renders Ela expression cancer-selective (as previously described in U.S. Patent No. 9,073,980).
  • the modified plasmid is hereafter referred to as the TAV plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV virus.
  • the TAV plasmid was further modified to carry a Sall site at the start of the Elb- l9k region and an Xhol site 200 base pairs 3' of the Sall site to facilitate insertion of therapeutic transgenes.
  • a Sall site at the start of the Elb- l9k region and an Xhol site 200 base pairs 3' of the Sall site to facilitate insertion of therapeutic transgenes.
  • the plasmid was cut with Sall and Xhol and self-ligated.
  • the nucleotide sequence of the modified Elb-l9k region is as follows, with the residual bases from the fused Sall and Xhol sites underlined:
  • the modified plasmid is hereafter referred to as the TAV-Al9k plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV-Al9k virus.
  • Exogenous nucleotide sequences encoding BMP -2 or BMP-7 were cloned into the modified Elb-l9k region.
  • Bone morphogenetic proteins (BMPs) are osteoinductive growth factors that belong to the transforming growth factor b (TGF-b) family.
  • TGF-b transforming growth factor b
  • the modified plasmid is hereafter referred to as the TAV-BMP-2 plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV-BMP-2 virus.
  • a nucleotide sequence encoding exemplary protein BMP-7 was cloned in to the modified Elb-l9k region of the TAV-Al9k plasmid.
  • the modified plasmid is hereafter referred to as the TAV-BMP-7 plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV-BMP-7 virus.
  • This Example describes the construction of a recombinant adenovirus type 5 (Ad5) that expresses a protein that defines a binding site or a protein that mediates the production of a biomolecule that defines a binding site for a therapeutic agent, e.g., BMP -4 or BMP-6.
  • Ad5 a recombinant adenovirus type 5
  • a plasmid carrying the 5' portion of the adenovirus type 5 genomic sequence is modified to carry the deletion of a nucleotide region located from -304 to -255 upstream of the Ela initiation site, which renders Ela expression cancer-selective (as previously described in U.S. Patent No. 9,073,980).
  • the modified plasmid is hereafter referred to as the TAV plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV virus.
  • the TAV plasmid is further modified to carry a Sall site at the start of the Elb- l9k region and an Xhol site 200 base pairs 3' of the Sall site to facilitate insertion of therapeutic transgenes.
  • a Sall site at the start of the Elb- l9k region and an Xhol site 200 base pairs 3' of the Sall site to facilitate insertion of therapeutic transgenes.
  • the plasmid is cut with Sall and Xhol and self-ligated.
  • the nucleotide sequence of the modified Elb-l9k region is as follows, with the residual bases from the fused Sall and Xhol sites underlined:
  • TAV-A 19k plasmid ATCTTGGTTACATCTGACCTCGTCGAGTCACCAGGCGCTTTTCCAA ( SEQ ID NO: 16).
  • TAV-D 19k virus any resulting viral particles produced therefrom are hereafter referred to as the TAV-D 19k virus.
  • BMPs Bone morphogenetic proteins
  • TGF-b transforming growth factor b
  • a nucleotide sequence encoding exemplary protein BMP-4 is cloned in to the modified Elb-l9k region of the TAV-Al9k plasmid.
  • the modified plasmid is hereafter referred to as the TAV-BMP-4 plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV-BMP-4 virus.
  • a nucleotide sequence encoding exemplary protein BMP-6 is cloned in to the modified Elb-l9k region of the TAV- Al9k plasmid.
  • the modified plasmid is hereafter referred to as the TAV-BMP-6 plasmid, and any resulting viral particles produced therefrom are hereafter referred to as the TAV-BMP-6 virus.
  • This Example describes the expression of BMPs from recombinant viruses.
  • A549 cells human lung cancer cells
  • (i) were infected with the TAV-BMP-2 virus, prepared as described in Example 1, at a multiplicity of infection (MOI) of 5,
  • (ii) were infected with the TAV-Al9k virus, without the BMP -2 gene, prepared as described in example 1, at an MOI of 5, or (ii) kept as non-infected controls.
  • BMP -2 levels in collected media were assayed by ELISA four days after infection. As depicted in FIGURE 1, BMP -2 expression from the TAV-BMP-2 virus resulted in approximately 10 ng/ml of BMP -2 in the collected media.
  • A549 cells were similarly (i) were infected with the TAV-BMP-7 virus, prepared as described in Example 1, at a multiplicity of infection (MOI) of 5, (ii) were infected with the TAV-Al9k virus, without the BMP-7 gene, prepared as described in example 1, at an MOI of 5, or (ii) kept as non-infected controls.
  • BMP-7 levels in conditioned media were measured by ELISA four days after infection. As shown in FIGURE 2, BMP-7 expression from the TAV-BMP-7 virus resulted in approximately 8 ng/ml of BMP-7 in the collected media.
  • Example 4 Cytotoxicity of BMP Expressing Viruses
  • This Example describes cytotoxicity resulting from BMP expressing recombinant viruses.
  • A549 cells human lung cancer cells were infected with the TAV-A19k.
  • TAV-A19k human lung cancer cells
  • TAV-BMP-7 viruses produced as described in Example 1.
  • Cells were stained with crystal violet, which stains viable cells blue, at the indicated time points after infection.
  • infection with the TAV-BMP-2 or TAV-BMP-7 results in cell death starting at approximately 6 days after infection.
  • This Example describes treatment of lung cancer in a mouse model with BMP expressing viruses.
  • mice were injected subcutaneously with ADS-12 (mouse lung cancer) cells and allowed to form tumors. After the tumors became large enough to treat, the mice were treated with TAV-Al9k, TAV-BMP-2, or TAV-BMP-7 (produced as described in Example 1), each at a dose of 1E9 PFU (plaque forming units). Viruses were injected intratumorally every four days for a total of three doses. Buffer without virus was used as a control. Tumor volumes of mice treated with buffer, TAV-Al9k, and TAV-BMP-7 are shown in FIGURE 4. Tumor volumes after treatment with TAV-BMP-2 were not smaller than with TAV-Al9k and are omitted for clarity.
  • ADS-12 mamouse lung cancer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Mycology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Optics & Photonics (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Developmental Biology & Embryology (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des compositions comprenant (a) un véhicule d'administration comprenant une séquence nucléotidique exogène codant pour une protéine qui définit un site de liaison (par exemple, une protéine morphogénétique osseuse (BMP) ou un antigène membranaire spécifique de la prostate (PSMA)), ou une protéine qui induit la production d'une biomolécule qui définit un site de liaison (par exemple, une BMP); et (b) un agent thérapeutique facultatif qui se lie au site de liaison, qui peut être utilisé pour le traitement du cancer, le traitement de la perte osseuse et/ou de l'hypercalcémie, ou la génération de tissu osseux chez un sujet.
PCT/US2019/041484 2018-07-11 2019-07-11 Procédés et compositions pour cibler des cellules cancéreuses pour un traitement WO2020014539A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862696450P 2018-07-11 2018-07-11
US62/696,450 2018-07-11

Publications (1)

Publication Number Publication Date
WO2020014539A1 true WO2020014539A1 (fr) 2020-01-16

Family

ID=69141719

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/041484 WO2020014539A1 (fr) 2018-07-11 2019-07-11 Procédés et compositions pour cibler des cellules cancéreuses pour un traitement

Country Status (1)

Country Link
WO (1) WO2020014539A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11077156B2 (en) 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11793843B2 (en) 2019-01-10 2023-10-24 Janssen Biotech, Inc. Prostate neoantigens and their uses
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
US12018289B2 (en) 2019-11-18 2024-06-25 Janssen Biotech, Inc. Vaccines based on mutant CALR and JAK2 and their uses

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BALMAYOR, E. R. ET AL.: "Gene Therapy for Bone Engineering", FRONT BIOENG BIOTECHNOL, vol. 3, 2015, XP055676397 *
DUGGAL, R. ET AL.: "Vaccinia virus expressing bone morphogenetic protein-4 in novel glioblastoma orthotopic models facilitates enhanced tumor regression and long-term survival", J TRANSL MED, vol. 11, no. 155, 2013, XP021155522 *
FAJARDO, C. A. ET AL.: "Oncolytic Adenoviral Delivery of an EGFR-Targeting T- cell Engager Improves Antitumor Efficacy", CANCER RES., vol. 77, no. 8, 2017, pages 2052 - 2063, XP055388559, DOI: 10.1158/0008-5472.CAN-16-1708 *
MORIUCHI, S. ET AL.: "Double suicide gene therapy using a replication defective herpes simplex virus vector reveals reciprocal interference in a malignant glioma model", GENE THER, vol. 9, no. 9, 2002, pages 584 - 591, XP055676400 *
PEERLINCK, I. ET AL.: "Targeted radionuclide therapy using a Wnt-targeted replicating adenovirus encoding the Na/I symporter", CLIN CANCER RES., vol. 15, no. 21, 2009, pages 6595 - 6601, XP055676395 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11077156B2 (en) 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
US11793843B2 (en) 2019-01-10 2023-10-24 Janssen Biotech, Inc. Prostate neoantigens and their uses
US12018289B2 (en) 2019-11-18 2024-06-25 Janssen Biotech, Inc. Vaccines based on mutant CALR and JAK2 and their uses

Similar Documents

Publication Publication Date Title
WO2020014539A1 (fr) Procédés et compositions pour cibler des cellules cancéreuses pour un traitement
JP7295192B2 (ja) 改変された腫瘍溶解性ウイルス
JP2022024161A (ja) 免疫療法のための組成物および方法
JP2023107931A (ja) 免疫療法のための組成物および方法
US11253608B2 (en) Recombinant adenoviruses carrying transgenes
US11999973B2 (en) Method for producing recombinant oncolytic adenovirus with modified E1a cancer-specific promoter
US20190352616A1 (en) Multiple transgene recombinant adenovirus
US20200155625A1 (en) Anti-angiogenic adenovirus
US20190352669A1 (en) Tumor selective tata-box and caat-box mutants
CN111263808A (zh) 一种靶向HPK1的gRNA和一种编辑HPK1基因的方法
KR20210089707A (ko) 항-liv1 면역세포 암 치료법
IL295405A (en) Chimeric antigen receptors with cd28 mutations and their use
WO2004101777A1 (fr) Virus recombine contenant un anticorps de therapie antitumorale intact comportant des regions humaines constantes et son utilisation
CN112996909B (zh) 表达pd-1结合蛋白的溶瘤病毒及其应用
US20200078415A1 (en) Multiple transgene recombinant adenovirus
WO2023172989A2 (fr) Agonistes et antagonistes du récepteur epo
CN117660358A (zh) 表达分泌型融合蛋白的工程化免疫细胞及其应用
CN117615786A (zh) 嵌合抗原受体及其使用方法

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: 19833415

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19833415

Country of ref document: EP

Kind code of ref document: A1