US20180256655A1 - Newcastle Disease Viruses and Uses Thereof - Google Patents

Newcastle Disease Viruses and Uses Thereof Download PDF

Info

Publication number
US20180256655A1
US20180256655A1 US15/789,340 US201715789340A US2018256655A1 US 20180256655 A1 US20180256655 A1 US 20180256655A1 US 201715789340 A US201715789340 A US 201715789340A US 2018256655 A1 US2018256655 A1 US 2018256655A1
Authority
US
United States
Prior art keywords
cancer
ndv
cell
protein
receptor
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US15/789,340
Inventor
Peter Palese
Adolfo Garcia-Sastre
Dmitriy Zamarin
James Allison
Jedd D. Wolchok
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Icahn School of Medicine at Mount Sinai
Memorial Sloan Kettering Cancer Center
Original Assignee
Icahn School of Medicine at Mount Sinai
Memorial Sloan Kettering Cancer Center
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 Icahn School of Medicine at Mount Sinai, Memorial Sloan Kettering Cancer Center filed Critical Icahn School of Medicine at Mount Sinai
Priority to US15/789,340 priority Critical patent/US20180256655A1/en
Publication of US20180256655A1 publication Critical patent/US20180256655A1/en
Assigned to ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI reassignment ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GARCIA-SASTRE, ADOLFO, PALESE, PETER
Assigned to MEMORIAL SLOAN KETTERING CANCER CENTER reassignment MEMORIAL SLOAN KETTERING CANCER CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALLISON, JAMES, WOLCHOK, JEDD, ZAMARIN, DMITRIY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18121Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18132Use 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18133Use of viral protein as therapeutic agent other than vaccine, e.g. apoptosis inducing or anti-inflammatory
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18141Use of virus, viral particle or viral elements as a vector
    • C12N2760/18143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • chimeric Newcastle disease viruses engineered to express an agonist of a co-stimulatory signal of an immune cell and compositions comprising such viruses. Also described herein are chimeric Newcastle disease viruses engineered to express an antagonist of an inhibitory signal of an immune cell and compositions comprising such viruses. The chimeric Newcastle disease viruses and compositions are useful in the treatment of cancer. In addition, described herein are methods for treating cancer comprising administering Newcastle disease viruses in combination with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • Newcastle Disease Virus is a member of the Avulavirus genus in the Paramyxoviridae family, which has been shown to infect a number of avian species (Alexander, D J (1988). Newcastle disease, Newcastle disease virus—an avian paramyxovirus. Kluwer Academic Publishers: Dordrecht, The Netherlands. pp 1-22). NDV possesses a single-stranded RNA genome in negative sense and does not undergo recombination with the host genome or with other viruses (Alexander, D J (1988). Newcastle disease, Newcastle disease virus—an avian paramyxovirus. Kluwer Academic Publishers: Dordrecht, The Netherlands. pp 1-22).
  • the genomic RNA contains genes in the order of 3′-NP-P-M-F-HN-L-5′, described in further detail below. Two additional proteins, V and W, are produced by NDV from the P gene by alternative mRNAs that are generated by RNA editing.
  • the genomic RNA also contains a leader sequence at the 3′ end.
  • the structural elements of the virion include the virus envelope which is a lipid bilayer derived from the cell plasma membrane.
  • the glycoprotein, hemagglutinin-neuraminidase (HN) protrudes from the envelope allowing the virus to contain both hemagglutinin (e.g., receptor binding/fusogenic) and neuraminidase activities.
  • the fusion glycoprotein (F) which also interacts with the viral membrane, is first produced as an inactive precursor, then cleaved post-translationally to produce two disulfide linked polypeptides.
  • the active F protein is involved in penetration of NDV into host cells by facilitating fusion of the viral envelope with the host cell plasma membrane.
  • the matrix protein (M) is involved with viral assembly, and interacts with both the viral membrane as well as the nucleocapsid proteins.
  • the main protein subunit of the nucleocapsid is the nucleocapsid protein (NP) which confers helical symmetry on the capsid.
  • NP nucleocapsid protein
  • P phosphoprotein
  • L L protein
  • the phosphoprotein (P) which is subject to phosphorylation, is thought to play a regulatory role in transcription, and may also be involved in methylation, phosphorylation and polyadenylation.
  • the L gene which encodes an RNA-dependent RNA polymerase, is required for viral RNA synthesis together with the P protein.
  • the L protein which takes up nearly half of the coding capacity of the viral genome is the largest of the viral proteins, and plays an important role in both transcription and replication.
  • the V protein has been shown to inhibit interferon-alpha and to contribute to the virulence of NDV (Huang et al. (2003). Newcastle disease virus V protein is associated with viral pathogenesis and functions as an Alpha Interferon Antagonist. Journal of Virology 77: 8676-8685).
  • NDV Newcastle disease virus
  • Naturally-occurring strains of NDV have been used in multiple clinical trials against advanced human cancers (Sinkovics, J G, and Horvath, J C (2000). Newcastle disease virus (NDV): brief history of its oncolytic strains. J Clin Virol 16: 1-15; Lorence et al. (2007). Phase 1 clinical experience using intravenous administration of PV701, an oncolytic Newcastle disease virus. Curr Cancer Drug Targets 7: 157-167; Hotte et al.
  • NDVs Newcastle disease viruses
  • chimeric NDVs engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, wherein the agonist is expressed.
  • chimeric NDVs comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, wherein the antagonist is expressed.
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell and a mutated F protein that causes the NDV to be highly fusogenic, wherein the agonist and the mutated F protein are expressed.
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell and a mutated F protein with a mutated cleavage site, wherein the agonist and the mutated F protein are expressed.
  • the chimeric NDVs expressing the mutated F protein have increased fusogenic activity relative to the corresponding virus expressing the counterpart F protein without the mutations to the cleavage site.
  • the modified F protein is incorporated into the virion.
  • chimeric NDVs comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell and a mutated F protein that causes the NDV to be highly fusogenic, wherein the antagonist and the mutated F protein are expressed.
  • chimeric NDVs comprising a packaged genome which encodes antagonist of an inhibitory signal of an immune cell and a mutated F protein with a mutated cleavage site, wherein the antagonist and the mutated F protein are expressed.
  • the chimeric NDVs expressing the mutated F protein have increased fusogenic activity relative to the corresponding virus expressing the counterpart F protein without the mutations to the cleavage site.
  • the modified F protein is incorporated into the virion.
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell and a cytokine (e.g., interleukin (IL)-2), wherein the agonist and the cytokine are expressed.
  • a cytokine e.g., interleukin (IL)-2
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein that causes the NDV to be highly fusogenic, wherein the agonist, the cytokine and the mutated F protein are expressed.
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein with a mutated cleavage site, wherein the agonist, the cytokine and the mutated F protein are expressed.
  • a cytokine e.g., IL-2
  • a mutated F protein with a mutated cleavage site wherein the agonist, the cytokine and the mutated F protein are expressed.
  • the chimeric NDVs expressing the mutated F protein with the mutated cleavage site are highly fusogenic.
  • the mutated F protein is incorporated into the virion.
  • chimeric NDVs comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell of an immune cell and a cytokine (e.g., IL-2), wherein the antagonist and the cytokine are expressed.
  • a cytokine e.g., IL-2
  • chimeric NDVs comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein that causes the NDV to be highly fusogenic, wherein the antagonist, the cytokine and the mutated F protein are expressed.
  • chimeric NDVs comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein with a mutated cleavage site, wherein the antagonist, the cytokine and the mutated F protein are expressed.
  • the chimeric NDVs expressing the mutated F protein with the mutated cleavage site are highly fusogenic.
  • the mutated F protein is incorporated into the virion.
  • the agonist of a co-stimulatory signal of an immune cell is an agonist of a co-stimulatory receptor expressed by an immune cell.
  • co-stimulatory receptors include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell costimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-1BB (CD137), and CD40.
  • GITR glucocorticoid-induced tumor necrosis factor receptor
  • ICOS or CD278 Inducible T-cell costimulator
  • OX40 CD134
  • CD27, CD28, 4-1BB (CD137) CD40.
  • CD137 CD137
  • the antibody is an sc-Fv.
  • the antibody is a bispecific antibody that binds to two receptors on an immune cell.
  • the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell.
  • the antibody is a human or humanized antibody.
  • the ligand or an antibody is a chimeric protein comprising a NDV F protein or NDV HN protein. Methods for generating such chimeric proteins are known in the art. See, e.g., U.S. Patent Application Publication No. 2012-0122185, the disclosure of which is herein incorporated by reference in its entirety. Also see Park et al., PNAS 2006; 103:8203-8 and Murawski et al., J Virol 2010; 84:1110-23, the disclosures of which is herein incorporated by reference in their entireties.
  • the antagonist of an inhibitory signal of an immune cell is an antagonist of an inhibitory receptor expressed by an immune cell.
  • inhibitory receptors include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), and adenosine A2a receptor (A2aR).
  • CTLA-4 or CD52 cytotoxic T-lymphocyte-associated antigen 4
  • PD1 or CD279 programmed cell death protein 1
  • B and T-lymphocyte attenuator (BTLA) B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), and adenosine A2a receptor (A2aR).
  • the antibody is a monoclonal antibody. In another embodiment, the antibody is an sc-Fv. In specific embodiments, the antibody is a human or humanized antibody. In another specific embodiment, the antagonist of an inhibitory receptor is a soluble receptor or antibody (or an antigen-binding fragment thereof) that specifically binds to a ligand of the inhibitory receptor.
  • the NDVs described herein can be propagated in any cell, subject, tissue or organ susceptible to a NDV infection.
  • the NDVs described herein e.g., chimeric NDVs described herein
  • the NDVs described herein e.g., chimeric NDVs described herein
  • the NDVs described herein may be propagated in an embryonated egg.
  • presented herein are isolated cells, tissues or organs infected with an NDV described herein (e.g., a chimeric NDV described herein). See, e.g., Section 5.4, infra, for examples of cells, animals and eggs to infect with an NDV described herein (e.g., a chimeric NDV described herein).
  • presented herein are isolated cancer cells infected with an NDV described herein (e.g., a chimeric NDV described herein).
  • presented herein are cell lines infected with an NDV described herein (e.g., a chimeric NDV described herein).
  • presented herein are embryonated eggs infected with an NDV described herein (e.g., a chimeric NDV described herein).
  • compositions comprising an NDV described herein (e.g., a chimeric NDV described herein).
  • pharmaceutical compositions comprising an NDV described herein (e.g., a chimeric NDV described herein) and a pharmaceutically acceptable carrier.
  • pharmaceutical compositions comprising cancer cells infected with an NDV described herein (e.g., a chimeric NDV described herein), and a pharmaceutically acceptable carrier.
  • the cancer cells have been treated with gamma radiation prior to incorporation into the pharmaceutical composition.
  • the cancer cells have been treated with gamma radiation before infection with the NDV (e.g., chimeric NDV).
  • the cancer cells have been treated with gamma radiation after infection with the NDV (e.g., chimeric NDV).
  • NDV e.g., chimeric NDV
  • pharmaceutical compositions comprising protein concentrate from lysed NDV-infected cancer cells (e.g., chimeric-NDV infected cancer cells), and a pharmaceutically acceptable carrier.
  • a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in a cell line that is susceptible to an NDV infection; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition.
  • a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in an embryonated egg; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition.
  • an NDV described herein e.g., a chimeric NDV described herein
  • a method for treating cancer comprises infecting a cancer cell in a subject with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof.
  • a method for treating cancer comprises administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof.
  • an effective amount of a chimeric NDV described herein e.g., a chimeric NDV described in Section 5.2, infra
  • a composition comprising an effective amount of a chimeric NDV described herein is administered to a subject to treat cancer.
  • the chimeric NDV comprises a genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell).
  • the genome of the NDV also comprises a mutated F protein.
  • two or more chimeric NDVs are administered to a subject to treat cancer.
  • a method for treating cancer comprises administering to a subject in need thereof cancer cells infected with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or composition thereof.
  • the cancer cells have been treated with gamma radiation prior to administration to the subject or incorporation into the composition.
  • a method for treating cancer comprises administering to a subject in need thereof a protein concentrate or plasma membrane fragments from cancer cells infected with a chimeric NDV (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof.
  • the chimeric NDV comprises a genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell).
  • the genome of the NDV also comprises a mutated F protein.
  • presented herein are methods for treating cancer utilizing an NDV described herein (e.g., a chimeric NDV such as described in Section 5.2, infra) or a composition comprising such the NDV in combination with one or more other therapies.
  • methods for treating cancer comprising administering to a subject an NDV described herein (e.g., a chimeric NDV, such as described in Section 5.2.1, infra) and one or more other therapies.
  • methods for treating cancer comprising administering to a subject an effective amount of an NDV described herein or a composition comprising an effective amount of an NDV described herein, and one or more other therapies.
  • the NDV and one or more other therapies can be administered concurrently or sequentially to the subject. In certain embodiments, the NDV and one or more other therapies are administered in the same composition. In other embodiments, the NDV and one or more other therapies are administered in different compositions. The NDV and one or more other therapies can be administered by the same or different routes of administration to the subject.
  • any NDV type or strain may be used in a combination therapy disclosed herein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses.
  • the NDV used in a combination with one or more other therapies is a naturally-occurring strain.
  • the NDV used in combination with one or more other therapies is a chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising a cytokine (e.g., IL-2, IL-7, IL-15, IL-17, or IL-21).
  • the cytokine is expressed by cells infected with the chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising a tumor antigen.
  • the tumor antigen is expressed by cells infected with the chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising a pro-apoptotic molecule or an anti-apoptotic molecule.
  • the pro-apoptotic molecule or anti-apoptotic molecule is expressed by cells infected with the chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell).
  • the agonist and/or antagonist are expressed by cells infected with the chimeric NDV.
  • the genome of the NDV also comprises a mutated F protein.
  • the one or more therapies used in combination with an NDV described herein is one or more other therapies described in Section 5.6.4 infra.
  • the one or more therapies used in combination with an NDV described herein is an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell (see, e.g., Section 5.6.4.4.1, infra). See, e.g., Section 5.2.1, infra, for examples of agonists of a co-stimulatory signal of an immune cell and antagonists of an inhibitory signal of an immune cell.
  • the antagonist of an inhibitory signal of an immune cell is the anti-CTLA-4 antibody described in Section 6, infra.
  • the agonist of a co-stimulatory signal of an immune cell is the ICOS ligand described in Section 6, infra
  • the term “about” or “approximately” when used in conjunction with a number refers to any number within 1, 5 or 10% of the referenced number.
  • an agonist refers to a molecule(s) that binds to another molecule and induces a biological reaction.
  • an agonist is a molecule that binds to a receptor on a cell and triggers one or more signal transduction pathways.
  • an agonist includes an antibody or ligand that binds to a receptor on a cell and induces one or more signal transduction pathways.
  • the antibody or ligand binds to a receptor on a cell and induces one or more signal transduction pathways.
  • the agonist facilitates the interaction of the native ligand with the native receptor.
  • an antagonist refers to a molecule(s) that inhibits the action of another molecule without provoking a biological response itself.
  • an antagonist is a molecule that binds to a receptor on a cell and blocks or dampens the biological activity of an agonist.
  • an antagonist includes an antibody or ligand that binds to a receptor on a cell and blocks or dampens binding of the native ligand to the cell without inducing one or more signal transduction pathways.
  • Another example of an antagonist includes an antibody or soluble receptor that competes with the native receptor on cells for binding to the native ligand, and thus, blocks or dampens one or more signal transduction pathways induced when the native receptor binds to the native ligand.
  • antibody refers to molecules that contain an antigen binding site, e.g., immunoglobulins.
  • Antibodies include, but are not limited to, monoclonal antibodies, bispecific antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, polyclonal antibodies, single domain antibodies, camelized antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked bispecific Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id and anti-anti-Id antibodies to antibodies), and epitope-binding fragments of any of the above.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • an antibody is a human or humanized antibody.
  • an antibody is a monoclonal antibody or scFv.
  • an antibody is a human or humanized monoclonal antibody or scFv.
  • the antibody is a bispecific antibody.
  • the bispecific antibody specifically binds to a co-stimulatory receptor of an immune cell or an inhibitory receptor of an immune, and a receptor on a cancer cell. In some embodiments, the bispecific antibody specifically binds to two receptors immune cells, e.g., two co-stimulatory receptors on immune cells, two inhibitory receptors on immune cells, or one co-stimulatory receptor on immune cells and one inhibitory receptor on immune cells.
  • the term “derivative” in the context of proteins or polypeptides refers to: (a) a polypeptide that is at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% or is 40% to 65%, 50% to 90%, 65% to 90%, 70% to 90%, 75% to 95%, 80% to 95%, or 85% to 99% identical to a native polypeptide; (b) a polypeptide encoded by a nucleic acid sequence that is at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% or is 40% to 65%, 50% to 90%, 65% to 90%, 70% to 90%, 75% to 95%, 80% to 95%, or 85% to 99% identical a nucleic acid sequence encoding a native polypeptide; (c) a polypeptide that contains 1, 2, 3, 4, 5, 6, 7, 8,
  • Derivatives also include a polypeptide that comprises the amino acid sequence of a naturally occurring mature form of a mammalian polypeptide and a heterologous signal peptide amino acid sequence.
  • derivatives include polypeptides that have been chemically modified by, e.g., glycosylation, acetylation, pegylation, phosphorylation, amidation, derivitization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein moiety, etc.
  • derivatives include polypeptides comprising one or more non-classical amino acids.
  • a derivative is isolated.
  • a derivative retains one or more functions of the native polypeptide from which it was derived.
  • Percent identity can be determined using any method known to one of skill in the art. In a specific embodiment, the percent identity is determined using the “Best Fit” or “Gap” program of the Sequence Analysis Software Package (Version 10; Genetics Computer Group, Inc., University of Wisconsin Biotechnology Center, Madison, Wis.). Information regarding hybridization conditions (e.g., high, moderate, and typical stringency conditions) have been described, see, e.g., U.S. Patent Application Publication No. US 2005/0048549 (e.g., paragraphs 72-73).
  • fragment is the context of a fragment of a proteinaceous agent (e.g., a protein) refers to a fragment that is 8 or more contiguous amino acids, 10 or more contiguous amino acids, 15 or more contiguous amino acids, 20 or more contiguous amino acids, 25 or more contiguous amino acids, 50 or more contiguous amino acids, 75 or more contiguous amino acids, 100 or more contiguous amino acids, 150 or more contiguous amino acids, 200 or more contiguous amino acids, or in the range of between 10 to 300 contiguous amino acids, 10 to 200 contiguous amino acids, 10 to 250 contiguous amino acids, 10 to 150 contiguous amino acids, 10 to 100 contiguous amino acids, 10 to 50 contiguous amino acids, 50 to 100 contiguous amino acids, 50 to 150 contiguous amino acids, 50 to 200 contiguous amino acids, 50 to 250 contiguous amino acids, 50 to 300 contiguous amino acids, 25 to 50 contiguous amino acids, 25 to 75 contiguous amino acids,
  • a fragment of a proteinaceous agent retains one or more functions of the proteinaceous agent—in other words, it is a functional fragment.
  • a fragment of a proteinaceous agent retains the ability to interact with another protein and/or to induce, enhance or activate one or more signal transduction pathways.
  • the term “functional fragment,” in the context of a proteinaceous agent, refers to a portion of a proteinaceous agent that retains one or more activities or functions of the proteinaceous agent.
  • a functional fragment of an inhibitory receptor may retain the ability to bind one or more of its ligands.
  • a functional fragment of a ligand of a co-stimulatory receptor may retain the ability to bind to the receptor and/or induce, enhance or activate one or more signal transduction pathways mediated by the ligand binding to its co-stimulatory receptor.
  • heterologous refers an entity not found in nature to be associated with (e.g., encoded by and/or expressed by the genome of) a naturally occurring NDV.
  • yielderly human refers to a human 65 years or older.
  • human adult refers to a human that is 18 years or older.
  • human child refers to a human that is 1 year to 18 years old.
  • human toddler refers to a human that is 1 year to 3 years old.
  • human infant refers to a newborn to 1 year old year human.
  • cells infected with an NDV described herein that is engineered to express a mutated F protein have an increased ability to form syncytia relative to cells infected with the parental virus from which the virus is derived, which parental virus has an unmutated F protein.
  • the syncytia are quantitated microscopically by counting the number of nuclei per syncytium after a certain period of time (e.g., about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 36 hours, or about 36 hours to about 48 hours).
  • an interferon antagonist refers to an agent that reduces or inhibits the cellular interferon immune response.
  • an interferon antagonist is a proteinaceous agent that reduces or inhibits the cellular interferon immune response.
  • an interferon antagonist is a viral protein or polypeptide that reduces or inhibits the cellular interferon response.
  • an interferon antagonist is an agent that reduces or inhibits interferon expression and/or activity.
  • the interferon antagonist reduces or inhibits the expression and/or activity of type I IFN.
  • the interferon antagonist reduces or inhibits the expression and/or activity of type II IFN.
  • the interferon antagonist reduces or inhibits the expression and/or activity of type III IFN.
  • the interferon antagonist reduces or inhibits the expression and/or activity of either IFN- ⁇ , IFN- ⁇ or both.
  • the interferon antagonist reduces or inhibits the expression and/or activity of IFN- ⁇ .
  • the interferon antagonist reduces or inhibits the expression and/or activity of one, two or all of IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ .
  • the expression and/or activity of IFN- ⁇ , IFN- ⁇ and/or IFN- ⁇ in an embryonated egg or cell is reduced approximately 1 to approximately 100 fold, approximately 5 to approximately 80 fold, approximately 20 to approximately 80 fold, approximately 1 to approximately 10 fold, approximately 1 to approximately 5 fold, approximately 40 to approximately 80 fold, or 1, 2, 3, 4, 5, 7, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 fold by an interferon antagonist relative to the expression and/or activity of IFN- ⁇ , IFN- ⁇ , and/or IFN- ⁇ in a control embryonated egg or a cell not expressing or not contacted with such an interferon antagonist as measured by the techniques described herein or known to one skilled in the art.
  • the expression and/or activity of IFN- ⁇ , IFN- ⁇ and/or IFN- ⁇ in an embryonated egg or cell is reduced by at least 20% to 25%, at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, at least 80% to 85%, at least 85% to 90%, at least 90% to 95%, at least 95% to 99% or by 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% by an interferon antagonist relative to the expression and/or activity of IFN- ⁇ , IFN- ⁇ , and/or IFN- ⁇ in a control embryonated egg or a cell not expressing or not contacted with such an interferon antagonist as
  • IFN deficient systems or “IFN-deficient substrates” refer to systems, e.g., cells, cell lines and animals, such as mice, chickens, turkeys, rabbits, rats, horses etc., which do not produce one, two or more types of IFN, or do not produce any type of IFN, or produce low levels of one, two or more types of IFN, or produce low levels of any IFN (i.e., a reduction in any IFN expression of 5-10%, 10-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-90% or more when compared to IFN-competent systems under the same conditions), do not respond or respond less efficiently to one, two or more types of IFN, or do not respond to any type of IFN, have a delayed response to one, two or more types of IFN, and/or are deficient in the activity of antiviral genes induced by one, two or more types of IFN, or induced by any type of IFN
  • immunospecifically binds As used herein, the terms “immunospecifically binds,” “immunospecifically recognizes,” “specifically binds,” and “specifically recognizes” are analogous terms in the context of antibodies and refer to molecules that specifically bind to an antigen (e.g., epitope or immune complex) as understood by one skilled in the art.
  • an antigen e.g., epitope or immune complex
  • a molecule that specifically binds to an antigen may bind to other peptides or polypeptides with lower affinity as determined by, e.g., immunoassays (e.g., ELISA), surface plasmon resonance (e.g., BIAcore®), a KinEx assay (using, e.g., a KinExA 3000 instrument (Sapidyne Instruments, Boise, Id.)), or other assays known in the art.
  • immunoassays e.g., ELISA
  • surface plasmon resonance e.g., BIAcore®
  • KinEx assay using, e.g., a KinExA 3000 instrument (Sapidyne Instruments, Boise, Id.)
  • molecules that specifically bind to an antigen bind to the antigen with a dissociation constant (i.e., Ka) that is at least 2 logs, 2.5 logs, 3 logs, 3.5 logs, 4 logs or greater than the Ka when the molecules bind to another antigen.
  • a dissociation constant i.e., Ka
  • molecules that specifically bind to an antigen do not cross react with other proteins.
  • the term “monoclonal antibody” is a term of the art and generally refers to an antibody obtained from a population of homogenous or substantially homogeneous antibodies, and each monoclonal antibody will typically recognize a single epitope (e.g., single conformation epitope) on the antigen.
  • MOI multiplicity of infection
  • the term “native ligand” refers to any naturally occurring ligand that binds to a naturally occurring receptor.
  • the ligand is a mammalian ligand.
  • the ligand is a human ligand.
  • native polypeptide(s) in the context of proteins or polypeptides refers to any naturally occurring amino acid sequence, including immature or precursor and mature forms of a protein.
  • native polypeptide is a human protein or polypeptide.
  • the term “native receptor” refers to any naturally occurring receptor that binds to a naturally occurring ligand.
  • the receptor is a mammalian receptor.
  • the receptor is a human receptor.
  • the terms “subject” or “patient” are used interchangeably.
  • the terms “subject” and “subjects” refers to an animal.
  • the subject is a mammal including a non-primate (e.g., a camel, donkey, zebra, cow, horse, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, chimpanzee, and a human).
  • the subject is a non-human mammal.
  • the subject is a pet (e.g., dog or cat) or farm animal (e.g., a horse, pig or cow).
  • the subject is a human.
  • the mammal e.g., human
  • the mammal is 0 to 6 months old, 6 to 12 months old, 1 to 5 years old, 5 to 10 years old, 10 to 15 years old, 15 to 20 years old, 20 to 25 years old, 25 to 30 years old, 30 to 35 years old, 35 to 40 years old, 40 to 45 years old, 45 to 50 years old, 50 to 55 years old, 55 to 60 years old, 60 to 65 years old, 65 to 70 years old, 70 to 75 years old, 75 to 80 years old, 80 to 85 years old, 85 to 90 years old, 90 to 95 years old or 95 to 100 years old.
  • the subject is an animal that is not avian.
  • the terms “treat” and “treating” in the context of the administration of a therapy refers to a treatment/therapy from which a subject receives a beneficial effect, such as the reduction, decrease, attenuation, diminishment, stabilization, remission, suppression, inhibition or arrest of the development or progression of cancer, or a symptom thereof.
  • the treatment/therapy that a subject receives results in at least one or more of the following effects: (i) the reduction or amelioration of the severity of cancer and/or a symptom associated therewith; (ii) the reduction in the duration of a symptom associated with cancer; (iii) the prevention in the recurrence of a symptom associated with cancer; (iv) the regression of cancer and/or a symptom associated therewith; (v) the reduction in hospitalization of a subject; (vi) the reduction in hospitalization length; (vii) the increase in the survival of a subject; (viii) the inhibition of the progression of cancer and/or a symptom associated therewith; (ix) the enhancement or improvement the therapeutic effect of another therapy; (x) a reduction or elimination in the cancer cell population; (xi) a reduction in the growth of a tumor or neoplasm; (xii) a decrease in tumor size; (xiii) a reduction in the formation of a tumor; (xiv) eradication
  • the treatment/therapy that a subject receives does not cure cancer, but prevents the progression or worsening of the disease. In certain embodiments, the treatment/therapy that a subject receives does not prevent the onset/development of cancer, but may prevent the onset of cancer symptoms.
  • the term “in combination” in the context of the administration of (a) therapy(ies) to a subject refers to the use of more than one therapy.
  • the use of the term “in combination” does not restrict the order in which therapies are administered to a subject.
  • a first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject.
  • the terms “therapies” and “therapy” can refer to any protocol(s), method(s), and/or agent(s) that can be used in the treatment of cancer.
  • the terms “therapies” and “therapy” refer to biological therapy, supportive therapy, hormonal therapy, chemotherapy, immunotherapy and/or other therapies useful in the treatment of cancer.
  • a therapy includes adjuvant therapy.
  • the term “therapy” refers to a chimeric NDV described herein. In other embodiments, the term “therapy” refers to an agent that is not a chimeric NDV.
  • FIG. 1 NDV infection upregulates the expression of MHC I, MHC II, and ICAM-1 on the surface of in vitro infected B16-F10 cells (24 hours post-infection).
  • FIGS. 2A-2E Intratumoral NDV treatment leads to infiltration with macrophages, NK cells, CD8 and CD4 effector cells and decreases the frequency of Tregs.
  • FIGS. 3A-3C Therapy with NDV exhibits favorable effects on tumor microenvironment of distant tumors.
  • FIGS. 4A-4C Lymphocytes infiltrating distant tumors upregulate activation, lytic, and proliferation markers. Representative expression plots on CD4 effector cells (top) and the corresponding percentages in the CD4 effector, CD8, Tregs (bottom) are shown for A) CD44, B) Granzyme B, and C) Ki-67.
  • FIGS. 5A-5D NDV Monotherapy delays the growth of distant tumors and provides some protection against tumor rechallenge. Bilateral flank tumors were established as described in FIG. 2A and the animals were treated and followed for survival. A) Growth of right flank (treated) tumors. B) Growth of left flank (non-treated) tumors. C) Overall survival. Numbers in boxes indicate percent of animals free of tumors. D) Survival in animals cured of B16-F10 melanoma by NDV re-challenged on day 75 with B16-F10 melanoma cells. Representative results of two different experiments with 10 mice per group.
  • FIGS. 6A-6B Tumor-infiltrating lymphocytes from both treated and non-treated tumors upregulate CTLA-4 in response to NDV therapy.
  • FIG. 7A-7C Combination therapy with NDV and CTLA-4 blockade enhances anti-tumor effect in the injected and distant tumors.
  • Bilateral B16 flank tumors were established and the animals were treated as described in FIG. 2A with or without anti-CTLA-4 antibody 9H10.
  • FIG. 8 Combination therapy with NDV and anti-CTLA-4 is effective systemically against non-virus-permissive prostate TRAMP tumors.
  • Right (day 12) and left (day 3) flank TRAMP tumors were established and the animals were treated with NDV as described in FIG. 2A with or without systemic anti-CTLA-4 antibody. Growth of left flank (non-injected) tumors is shown. Numbers in boxes indicate percent of animals free of tumors.
  • FIG. 9A-9C NDV infection upregulates expression of PD-L1 in B16-F10 tumors.
  • FIGS. 10A-10F Combination therapy with NDV and anti-PD-1 is effective systemically against B16 melanoma and results in increased T cell infiltration with upregulation of activation markers.
  • D-E Tumor-infiltrating lymphocytes from distant tumors were isolated and stained for expression of ICOS (D) and Granzyme B (E).
  • F Tumor infiltrating lymphocytes were re-stimulated with dendritic cells loaded with tumor lysates and assessed for expression of IFN gamma by intracellular cytokine staining.
  • FIG. 11 Combination therapy with NDV and CTLA-4 induces upregulation of ICOS and CD4 effector cells in distant tumors and tumor-draining lymph nodes (TDLN).
  • TDLN tumor-draining lymph nodes
  • FIGS. 12A-12D Generation and in vitro evaluation of NDV-ICOSL virus.
  • FIGS. 13A-13C Combination therapy with NDV-mICOSL and anti-CTLA-4 protects mice from contralateral tumor challenge and results in long-term animal survival.
  • Animals were challenged with a larger tumor dose and treated with NDV as described in FIG. 2A with or without systemic anti-CTLA-4 antibody. Growth of left flank (non-injected) tumors is shown.
  • FIG. 14A-14B Combination therapy with NDV-mICOSL and anti-CTLA-4 protects mice from contralateral tumor challenge and results in long-term animal survival in the CT26 colon carcinoma model.
  • Animals were challenged with a larger tumor dose and treated with NDV as described in FIG. 2A with or without systemic anti-CTLA-4 antibody. Growth of left flank (non-injected) tumors is shown. Numbers in boxes indicate percent of animals protected from tumors.
  • FIGS. 15A-15C NDV treatment leads to distant B16 tumor infiltration with macrophages, NK cells, CD8 and CD4 effector cells and decreases the frequency of Tregs.
  • FIG. 16A-16B Lymphocytes infiltrating distant B16 tumors upregulate activation, lytic, and proliferation markers.
  • FIG. 17 Tumor infiltrating lymphocytes from treated animals secrete IFN-gamma in response to stimulation with DC's loaded with B16-F10 lysates. Representative dot plots are shown.
  • FIGS. 18A-18B Animals cured by combination therapy are protected from further tumor challenge.
  • FIG. 19A-19B Recombinant ICOSL-F chimeric protein is efficiently expressed on surface.
  • NDVs Newcastle disease viruses
  • chimeric NDVs engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • chimeric NDVs comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, wherein the agonist is expressed.
  • chimeric NDVs comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, wherein the antagonist is expressed.
  • NDVs described herein e.g., chimeric NDVs described herein.
  • the NDVs described herein can be propagated in any cell, subject, tissue, organ or animal susceptible to a NDV infection.
  • compositions comprising an NDV described herein (e.g., a chimeric NDV described herein).
  • pharmaceutical compositions comprising an NDV described herein (e.g., a chimeric NDV described herein) and a pharmaceutically acceptable carrier.
  • pharmaceutical compositions comprising cancer cells infected with an NDV described herein (e.g., a chimeric NDV described herein), and a pharmaceutically acceptable carrier.
  • pharmaceutical compositions comprising protein concentrate from lysed NDV-infected cancer cells (e.g., chimeric-NDV infected cancer cells), and a pharmaceutically acceptable carrier.
  • a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in a cell line that is susceptible to an NDV infection; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition.
  • a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in an embryonated egg; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition.
  • an NDV described herein e.g., a chimeric NDV described herein
  • a method for treating cancer comprises infecting a cancer cell in a subject with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof.
  • a method for treating cancer comprises administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof.
  • an effective amount of a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition comprising an effective amount of a chimeric NDV described herein is administered to a subject to treat cancer.
  • the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell), wherein the agonist and/or antagonist are expressed by the NDV.
  • the genome of the NDV also comprises a mutated F protein.
  • two or more chimeric NDVs are administered to a subject to treat cancer.
  • a method for treating cancer comprises administering to a subject in need thereof cancer cells infected with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or composition thereof.
  • the cancer cells have been treated with gamma radiation prior to administration to the subject or incorporation into the composition.
  • a method for treating cancer comprises administering to a subject in need thereof a protein concentrate or plasma membrane fragments from cancer cells infected with a chimeric NDV (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof.
  • the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell), wherein the agonist and/or antagonist are expressed by the NDV.
  • the genome of the NDV also comprises a mutated F protein, which is expressed by the NDV.
  • presented herein are methods for treating cancer utilizing an NDV described herein (e.g., a chimeric NDV such as described in Section 5.2, infra) or a composition comprising such the NDV in combination with one or more other therapies.
  • methods for treating cancer comprising administering to a subject an NDV described herein (e.g., a chimeric NDV, such as described in Section 5.2, infra) and one or more other therapies.
  • presented herein are methods for treating cancer comprising administering to a subject an effective amount of an NDV described herein or a composition comprising an effective amount of an NDV described herein, and one or more other therapies.
  • the NDV and one or more other therapies can be administered concurrently or sequentially to the subject. In certain embodiments, the NDV and one or more other therapies are administered in the same composition. In other embodiments, the NDV and one or more other therapies are administered in different compositions. The NDV and one or more other therapies can be administered by the same or different routes of administration to the subject.
  • any NDV type or strain may be used in a combination therapy disclosed herein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses.
  • the NDV used in a combination with one or more other therapies is a naturally-occurring strain.
  • the NDV used in combination with one or more other therapies is a chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising a cytokine (e.g., IL-2, IL-7, IL-15, IL-17 or IL-21).
  • the chimeric NDV comprises a packaged genome, the genome comprising a tumor antigen.
  • the tumor antigen is expressed by cells infected with the chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising a pro-apoptotic molecule (e.g., Bax, Bak, Bad, BID, Bc1-xS, Bim, Noxa, Puma, AIF, FasL, and TRAIL) or an anti-apoptotic molecule (e.g., Bc1-2, Bc1-xL, Mc1-1, and XIAP).
  • a pro-apoptotic molecule e.g., Bax, Bak, Bad, BID, Bc1-xS, Bim, Noxa, Puma, AIF, FasL, and TRAIL
  • an anti-apoptotic molecule e.g., Bc1-2, Bc1-xL, Mc1-1, and XIAP
  • the pro-apoptotic molecule or anti-apoptotic molecule is expressed by cells infected with the chimeric NDV.
  • the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell).
  • the agonist and/or antagonist are expressed by cells infected with the chimeric NDV.
  • the genome of the NDV also comprises a mutated F protein, a tumor antigen, a heterologous interferon antagonist, a pro-apoptotic molecule and/or an anti-apoptotic molecule.
  • the one or more therapies used in combination with an NDV described herein is one or more other therapies described in Section 5.6.4, infra.
  • the one or more therapies used in combination with an NDV described herein are an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • the antagonist of an inhibitory signal of an immune cell is the anti-CTLA-4 antibody described in Section 6, infra.
  • the agonist of a co-stimulatory signal of an immune cell is the ICOS ligand described in Section 6, infra
  • any NDV type or strain may be used in a combination therapy disclosed herein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses.
  • the NDV used in a combination therapy disclosed herein is a naturally-occurring strain.
  • the NDV is a lytic strain.
  • the NDV used in a combination therapy disclosed herein is a non-lytic strain.
  • the NDV used in a combination therapy disclosed herein is lentogenic strain.
  • the NDV is a mesogenic strain.
  • the NDV used in a combination therapy disclosed herein is a velogenic strain.
  • NDV strains include, but are not limited to, the 73-T strain, NDV HUJ strain, Ulster strain, MTH-68 strain, lentil strain, Hickman strain, PV701 strain, Hitchner B1 strain (see, e.g., Genbank No. AF309418 or NC_002617), La Sota strain (see, e.g., Genbank No. AY845400), YG97 strain, MET95 strain, Roakin strain, and F48E9 strain.
  • the NDV used in a combination therapy disclosed herein that is the Hitchner B1 strain.
  • the NDV used in a combination therapy disclosed herein is a B1 strain as identified by Genbank No.
  • the NDV used in a combination therapy disclosed herein is the NDV identified by ATCC No. VR2239. In another specific embodiment, the NDV used in a combination therapy disclosed herein is the La Sota strain.
  • the NDV used in a combination therapy disclosed herein is not pathogenic birds as assessed by a technique known to one of skill.
  • the NDV used in a combination therapy is no pathogenic as assessed by intracranial injection of 1-day-old chicks with the virus, and disease development and death as scored for 8 days.
  • the NDV used in a combination therapy disclosed herein has an intracranial pathogenicity index of less than 0.7, less than 0.6, less than 0.5, less than 0.4, less than 0.3, less than 0.2 or less than 0.1.
  • the NDV used in a combination therapy disclosed herein has an intracranial pathogenicity index is zero.
  • the NDV used in a combination therapy disclosed herein is a mesogenic strain that has been genetically engineered so as not be a considered pathogenic in birds as techniques known to one skilled in the art. In certain embodiments, the NDV used in a combination therapy disclosed herein is a velogenic strain that has been genetically engineered so as not be a considered pathogenic in birds as techniques known to one skilled in the art.
  • the NDV used in a combination therapy disclosed herein expresses a mutated F protein.
  • the NDV used in a combination therapy expresses a mutated F protein is highly fusogenic and able to form syncytia.
  • the mutated F protein is incorporated into the virion.
  • a genome of a NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein with a mutated cleavage site.
  • the NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein in which the cleavage site of the F protein is mutated to produce a polybasic amino acid sequence, which allows the protein to be cleaved by intracellular proteases, which makes the virus more effective in entering cells and forming syncytia.
  • the NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein in which the cleavage site of the F protein is replaced with one containing one or two extra arginine residues, allowing the mutant cleavage site to be activated by ubiquitously expressed proteases of the furin family.
  • Specific examples of NDVs that express such a mutated F protein include, but are not limited to, rNDV/F2aa and rNDV/F3aa.
  • the NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the NDV used in a combination therapy disclosed herein is attenuated such that the NDV remains, at least partially, infectious and can replicate in vivo, but only generate low titers resulting in subclinical levels of infection that are non-pathogenic (see, e.g., Khattar et al., 2009, J. Virol. 83:7779-7782).
  • the NDV is attenuated by deletion of the V protein.
  • Such attenuated NDVs may be especially suited for embodiments wherein the virus is administered to a subject in order to act as an immunogen, e.g., a live vaccine.
  • the viruses may be attenuated by any method known in the art.
  • the NDV used in a combination therapy disclosed herein does not comprise an NDV V protein encoding sequence.
  • the NDV used in a combination therapy disclosed herein expresses a mutated V protein. See, e.g., Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844, which is incorporated herein by reference, for examples of mutated V proteins.
  • a mesogenic or velogenic NDV strain used in a combination therapy disclosed herein expresses a mutated V protein, such as disclosed by Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844.
  • the NDV used in a combination therapy disclosed herein is an NDV disclosed in U.S. Pat. No. 7,442,379, U.S. Pat. No. 6,451,323, or U.S. Pat. No. 6,146,642, which is incorporated herein by reference in its entirety.
  • the NDV used in a combination therapy disclosed herein is genetically engineered to encode and express a heterologous peptide or protein.
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV known to one of skill in the art, or a chimeric NDV disclosed herein (see, e.g., Section 5.2, infra).
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express a tumor antigen (see below for examples of tumor antigens).
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express a heterologous interferon antagonist (see below for examples of heterologous interferon antagonists).
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV disclosed in U.S. patent application publication No. 2012/0058141, which is incorporated herein by reference in its entirety.
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV disclosed in U.S. patent application publication No. 2012/0122185, which is incorporated herein by reference in its entirety.
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express a cytokine, such as, e.g., IL-2, IL-7, IL-9, IL-15, IL-17, IL-21, IL-22, IFN-gamma, GM-CSF, and TNF-alpha.
  • the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express IL-2.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or Natural Killer (NK) cell.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell.
  • the NDV serves as the “backbone” that is engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or Natural Killer (NK) cell.
  • an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or Natural Killer (NK) cell.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein.
  • the mutated F protein is highly fusogenic and able to form syncytia.
  • the mutated F protein is incorporated into the virion.
  • the genome of a NDV engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell comprises an NDV V protein encoding sequence.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein with a mutated cleavage site.
  • the NDV is engineered to express a mutated F protein in which the cleavage site of the F protein is mutated to produce a polybasic amino acid sequence, which allows the protein to be cleaved by intracellular proteases, which makes the virus more effective in entering cells and forming syncytia.
  • the NDV is engineered to express a mutated F protein in which the cleavage site of the F protein is replaced with one containing one or two extra arginine residues, allowing the mutant cleavage site to be activated by ubiquitously expressed proteases of the furin family.
  • NDVs that express such a mutated F protein include, but are not limited to, rNDV/F2aa and rNDV/F3aa.
  • the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the mutated F protein is incorporated into the virion.
  • the genome of a NDV engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell comprises a mutated NDV V protein encoding sequence, such as disclosed by Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844.
  • the genome of a NDV engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell does not comprise an NDV V protein encoding sequence.
  • parental backbone of the chimeric NDV is a mesogenic or velogenic NDV strain that is engineered to express a mutated V protein, such as disclosed by Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a cytokine.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a cytokine.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a cytokine.
  • an immune cell such as, e.g., a T-lymphocyte or NK cell
  • cytokine include, but are not limited to, interleukin (IL)-2, IL-7, IL-9, IL-15, IL-17, IL-21, IL-22, interferon (IFN) gamma, GM-CSF, and tumor necrosis factor (TNF)-alpha.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a cytokine (e.g., IL-2, IL-7, IL-9, IL-15, IL-17, IL-21, IL-22, IFN-gamma, GM-CSF, and TNF-alpha).
  • the mutated F protein are highly fusogenic.
  • the mutated F protein has a mutant cleavage site (such as described herein).
  • the mutated F protein comprises the amino acid mutation L289A.
  • the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the mutated F protein is incorporated into the virion.
  • chimeric NDVs comprising a genome engineered to express (i) an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, and (ii) a tumor antigen.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a tumor antigen.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a tumor antigen.
  • Tumor antigens include tumor-associated antigens and tumor-specific antigens.
  • Specific examples of tumor antigens include, but are not limited to, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, N-acetylglucosaminyltransferase-V, p-15, gp100, MART-1/MelanA, TRP-1 (gp75), Tyrosinase, cyclin-dependent kinase 4, ⁇ -catenin, MUM-1, CDK4, HER-2/neu, human papillomavirus-E6, human papillomavirus E7, CD20, carcinoembryonic antigen (CEA), epidermal growth factor receptor, MUC-1, caspase-8, CD5, mucin-1, Lewisx, CA-125, p185HER2, IL-2R, Fap- ⁇ , tenascin, antigens associated with a metalloproteinase, and CAMPATH-1.
  • KS 1/4 pan-carcinoma antigen such as ovarian carcinoma antigen (CA125), prostatic acid phosphate, prostate specific antigen, melanoma-associated antigen p97, melanoma antigen gp75, high molecular weight melanoma antigen (HMW-MAA), prostate specific membrane antigen, CEA, polymorphic epithelial mucin antigen, milk fat globule antigen, colorectal tumor-associated antigens (such as: CEA, TAG-72, CO17-1A, GICA 19-9, CTA-1 and LEA), Burkitt's lymphoma antigen-38.13, CD19, B-lymphoma antigen-CD20, CD33, melanoma specific antigens (such as ganglioside GD2, ganglioside GD3, ganglioside GM2, ganglioside GM3), tumor-specific transplantation type of cell-surface antigen (TSTA) (such as ganglioside GD
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a tumor antigen.
  • the mutated F protein are highly fusogenic.
  • the mutated F protein has a mutant cleavage site (such as described herein).
  • the mutated F protein comprises the amino acid mutation L289A.
  • the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the mutated F protein is incorporated into the virion.
  • chimeric NDVs comprising a genome engineered to express (i) an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, and (ii) a heterologous interferon antagonist.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a heterologous interferon antagonist.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a heterologous interferon antagonist.
  • an antagonist of an inhibitory signal of an immune cell such as, e.g., a T-lymphocyte or NK cell
  • a heterologous interferon antagonist See, e.g., U.S. patent application publication No. 2012-0058141, which is incorporated herein by reference, for examples of chimeric NDV engineered to express heterologous interferon antagonists.
  • Interferon antagonists may be identified using any technique known to one of skill in the art, including, e.g., the techniques described in U.S. Pat. Nos. 6,635,416; 7,060,430; and 7,442,527; which are incorporated herein by reference in their entirety.
  • the heterologous interferon antagonist is a viral protein.
  • viral proteins may be obtained or derived from any virus and the virus may infect any species (e.g., the virus may infect humans or non-human mammals).
  • heterologous interferon antagonists include, without limitation, Nipah virus W protein, Nipah V protein, Ebola virus VP35 protein, vaccinia virus E3L protein, influenza virus NS1 protein, respiratory syncytial virus (RSV) NS2 protein, herpes simplex virus (HSV) type 1 ICP34.5 protein, Hepatitis C virus NS3-4 protease, dominant-negative cellular proteins that block the induction or response to innate immunity (e.g., STAT1, MyD88, IKK and TBK), and cellular regulators of the innate immune response (e.g., SOCS proteins, PIAS proteins, CPLD proteins, IkB protein, Atg5 protein, Pin1 protein, IRAK-M protein, and UBP43). See, e.g., U.S. patent application publication No. 2012-0058141, which is incorporated herein by reference in its entirety, for additional information regarding heterologous interferon antagonist.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a heterologous interferon antagonist.
  • the mutated F protein are highly fusogenic.
  • the mutated F protein has a mutant cleavage site (such as described herein).
  • the mutated F protein comprises the amino acid mutation L289A.
  • the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the mutated F protein is incorporated into the virion.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a pro-apoptotic molecule.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a pro-apoptotic molecule.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a pro-apoptotic molecule.
  • an antagonist of an inhibitory signal of an immune cell such as, e.g., a T-lymphocyte or NK cell
  • a pro-apoptotic molecule include, but are not limited to, Bax, Bak, Bad, BID, Bcl-xS, Bim, Noxa, Puma, AIF, FasL, and TRAIL.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a pro-apoptotic molecule.
  • the mutated F protein are highly fusogenic.
  • the mutated F protein has a mutant cleavage site (such as described herein).
  • the mutated F protein comprises the amino acid mutation L289A.
  • the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the mutated F protein is incorporated into the virion.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and an anti-apoptotic molecule.
  • a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and an anti-apoptotic molecule.
  • a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and an anti-apoptotic molecule.
  • an anti-apoptotic molecule include, but are not limited to, Bcl-2, Bcl-xL, Mcl-1, and XIAP.
  • chimeric NDVs comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and an anti-apoptotic molecule.
  • the mutated F protein are highly fusogenic.
  • the mutated F protein has a mutant cleavage site (such as described herein).
  • the mutated F protein comprises the amino acid mutation L289A.
  • the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A.
  • the mutated F protein is from a different type or strain of NDV than the backbone NDV.
  • the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site.
  • the mutated F protein is in addition to the backbone NDV F protein.
  • the mutated F protein replaces the backbone NDV F protein.
  • the mutated F protein is incorporated into the virion.
  • any NDV type or strain may be used as a backbone that is engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and in certain embodiments, engineered to express a cytokine, tumor antigen, heterologous interferon antagonist and/or mutated F protein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses.
  • the NDV used in a combination therapy disclosed herein is a naturally-occurring strain.
  • the NDV that serves as the backbone for genetic engineering is a lytic strain. In other embodiments, the NDV that serves as the backbone for genetic engineering is a non-lytic strain. In certain embodiments, the NDV that serves as the backbone for genetic engineering is lentogenic strain. In some embodiments, the NDV that serves as the backbone for genetic engineering is mesogenic strain. In other embodiments, the NDV that serves as the backbone for genetic engineering is a velogenic strain.
  • NDV strains include, but are not limited to, the 73-T strain, NDV HUJ strain, Ulster strain, MTH-68 strain, lentil strain, Hickman strain, PV701 strain, Hitchner B1 strain, La Sota strain (see, e.g., Genbank No. AY845400), YG97 strain, MET95 strain, Roakin strain, and F48E9 strain.
  • the NDV that serves as the backbone for genetic engineering is the Hitchner B1 strain.
  • the NDV that serves as the backbone for genetic engineering is a B1 strain as identified by Genbank No. AF309418 or NC_002617.
  • the NDV that serves as the backbone for genetic engineering is the NDV identified by ATCC No. VR2239.
  • the NDV that serves as the backbone for genetic engineering is the La Sota strain.
  • Attenuation, or further attenuation, of the chimeric NDV is desired such that the chimeric NDV remains, at least partially, infectious and can replicate in vivo, but only generate low titers resulting in subclinical levels of infection that are non-pathogenic (see, e.g., Khattar et al., 2009, J. Virol. 83:7779-7782).
  • the NDV is attenuated by deletion of the V protein.
  • Such attenuated chimeric NDVs may be especially suited for embodiments wherein the virus is administered to a subject in order to act as an immunogen, e.g., a live vaccine.
  • the viruses may be attenuated by any method known in the art.
  • a chimeric NDV in addition to expressing an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and in certain embodiments, a mutated F protein and a cytokine, a chimeric NDV is engineered to express a suicide gene (e.g., thymidine kinase) or another molecule that inhibits NDV replication or function (a gene that makes NDV sensitive to an antibiotic or an anti-viral agent).
  • a suicide gene e.g., thymidine kinase
  • another molecule that inhibits NDV replication or function a gene that makes NDV sensitive to an antibiotic or an anti-viral agent
  • a chimeric NDV in addition to expressing an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and in certain embodiments, a mutated F protein and a cytokine, a chimeric NDV is engineered to encode tissue-specific microRNA (miRNA) target sites (e.g., sites targeted by miR-21, miR-184, miR-133a/133b, miR-137, and/or miR-193a microRNAs).
  • miRNA tissue-specific microRNA
  • the tropism of the chimeric NDV is altered.
  • the tropism of the virus is altered by modification of the F protein cleavage site to be recognized by tissue-specific or tumor-specific proteases such as matrix metalloproteases (MMP) and urokinase.
  • MMP matrix metalloproteases
  • tropism of the virus is altered by introduction of tissue-specific miRNA target sites.
  • NDV HN protein is mutated to recognize tumor-specific receptor.
  • the agonist of a co-stimulatory signal and/or the antagonist of an inhibitory signal of an immune cell may be inserted into the genome of the backbone NDV between two transcription units.
  • the agonist of a co-stimulatory signal and/or the antagonist of an inhibitory signal of an immune cell is inserted into the genome of the backbone NDV between the M and P transcription units or between the HN and L transcription units.
  • the cytokine, and/or mutated F protein are inserted into the genome of the backbone NDV between two or more transcription units (e.g., between the M and P transcription units or between the HN and L transcription units).
  • the chimeric NDVs described herein may be engineered to express any agonist of a co-stimulatory signal and/or any antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage), known to one of skill in the art.
  • the agonist and/or antagonist is an agonist of a human co-stimulatory signal of an immune cell and/or antagoinst of a human inhibitory signal of an immune cell.
  • the agonist of a co-stimulatory signal is an agonist of a co-stimulatory molecule (e.g., co-stimulatory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+ T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • a co-stimulatory molecule e.g., co-stimulatory receptor
  • immune cells such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+ T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • co-stimulatory molecules include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell costimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-1BB (CD137), CD40, lymphotoxin alpha (LT alpha), and LIGHT (lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes).
  • the agonist is an agonist of a human co-stimulatory receptor of an immune cell.
  • the agonist of a co-stimulatory receptor is not an agonist of ICOS.
  • the antagonist is an antagonist of an inhibitory molecule (e.g., inhibitory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+ T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • an inhibitory molecule e.g., inhibitory receptor
  • immune cells such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+ T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • inhibitory molecules include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD160, and adenosine A2a receptor (A2aR).
  • CTLA-4 or CD52 cytotoxic T-lymphocyte-associated antigen 4
  • PD1 or CD279 programmed cell death protein 1
  • B and T-lymphocyte attenuator (BTLA) killer cell immunoglobulin-like receptor
  • KIR killer cell immunoglobulin-like receptor
  • LAG3 lymphocyte activation gene 3
  • TIM3 T-cell membrane protein 3
  • CD160 adenosine A2a receptor
  • A2aR adenosine A2a receptor
  • the antagonist is an antagonist of a human inhibitory receptor of an immune cell.
  • the agonist of a co-stimulatory receptor is an antibody or antigen-binding fragment thereof that specifically binds to the co-stimulatory receptor.
  • co-stimulatory receptors include GITR, ICOS, OX40, CD27, CD28, 4-1BB, or CD40.
  • the antibody is a monoclonal antibody.
  • the antibody is an sc-Fv.
  • the antibody is a bispecific antibody that binds to two receptors on an immune cell.
  • the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell.
  • the antibody is a human or humanized antibody.
  • the ligand or an antibody is a chimeric protein with NDV F protein or NDV HN protein. See, e.g., U.S. patent application Publication No. 2012/0122185, which is incorporated herein by reference for a description regarding generation of chimeric F or chimeric HN proteins.
  • the chimeric protein is the chimeric F protein described in Section 6, infra. The techniques described below for generating the chimeric ICOSL-F protein can be used to generate other chimeric F proteins or chimeric HN proteins.
  • the agonist of a co-stimulatory receptor is a ligand of the co-stimulatory receptor.
  • the ligand is fragment of a native ligand.
  • native ligands include B7RP1, CD137L, OX40L, CD70, herpes virus entry mediator (HVEM), CD80, and CD86.
  • HVEM herpes virus entry mediator
  • the nucleotide sequences encoding native ligands as well as the amino acid sequences of native ligands are known in the art.
  • B7RP1 GenBank human: NM_015259.4, NP_056074.1 murine: NM_015790.3, NP_056605.1
  • CD137L GenBank human: NM_003811.3, NP_003802.1, murine: NM_009404.3, NP_033430.1
  • OX40L GenBank human: NM_003326.3, NP_003317.1, murine: NM_009452.2, NP_033478.0
  • CD70 GenBank human: NM_001252.3, NP_001243.1, murine: NM_011617.2, AAD00274.1
  • CD80 GenBank human: NM_05191.3, NP_005182.1, murine: NM_009855.2, NP_033985.3
  • CD86 GenBank human: NM_005191.3, CAG46642.1, murine: NM_019388.3,
  • the ligand is a derivative of a native ligand.
  • the ligand is a fusion protein comprising at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and a heterologous amino acid sequence.
  • the fusion protein comprises at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and the Fc portion of an immunoglobulin or a fragment thereof.
  • An example of a ligand fusion protein is a 4-1BB ligand fused to Fc portion of immunoglobulin (described by Meseck M et al., J Immunother. 2011 34:175-82).
  • the antagonist of an inhibitory receptor is an antibody (or an antigen-binding fragment) or a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s).
  • native ligands for inhibitory receptors include PDL-1, PDL-2, B7-H3, B7-H4, HVEM, Ga19 and adenosine.
  • Specific examples of inhibitory receptors that bind to a native ligand include CTLA-4, PD-1, BTLA, KIR, LAG3, TIM3, and A2aR.
  • the antagonist of an inhibitory receptor is a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s).
  • the soluble receptor is a fragment of a native inhibitory receptor or a fragment of a derivative of a native inhibitory receptor that specifically binds to native ligand (e.g., the extracellular domain of a native inhibitory receptor or a derivative of an inhibitory receptor).
  • the soluble receptor is a fusion protein comprising at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor (e.g., the extracellular domain of the native inhibitory receptor or a derivative of the native inhibitory receptor), and a heterologous amino acid sequence.
  • the fusion protein comprises at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor, and the Fc portion of an immunoglobulin or a fragment thereof.
  • An example of a soluble receptor fusion protein is a LAG3-Ig fusion protein (described by Huard B et al., Eur J Immunol. 1995 25:2718-21).
  • the antagonist of an inhibitory receptor is an antibody (or an antigen-binding fragment) that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s).
  • the antibody is a monoclonal antibody.
  • the antibody is an scFv.
  • the antibody is a human or humanized antibody.
  • a specific example of an antibody to inhibitory ligand is anti-PD-L1 antibody (Iwai Y, et al. PNAS 2002; 99:12293-12297).
  • the antagonist of an inhibitory receptor is an antibody (or an antigen-binding fragment) or ligand that binds to the inhibitory receptor, but does not transduce an inhibitory signal(s).
  • inhibitory receptors include CTLA-4, PD1, BTLA, KIR, LAG3, TIM3, and A2aR.
  • the antibody is a monoclonal antibody.
  • the antibody is an scFv.
  • the antibody is a human or humanized antibody.
  • a specific example of an antibody to inhibitory receptor is anti-CTLA-4 antibody (Leach D R, et al. Science 1996; 271: 1734-1736).
  • Another example of an antibody to inhibitory receptor is anti-PD-1 antibody (Topalian S L, NEJM 2012; 28:3167-75).
  • a chimeric NDV described herein is engineered to an antagonist of CTLA-4, such as, e.g., Ipilimumab or Tremelimumab.
  • a chimeric NDV described herein is engineered to an antagonist of PD1, such as, e.g., MDX-1106 (BMS-936558), MK3475, CT-011, AMP-224, or MDX-1105.
  • a chimeric NDV described herein is engineered to express an antagonist of LAG3, such as, e.g., IMP321.
  • a chimeric NDV described herein is engineered to express an antibody (e.g., a monoclonal antibody or an antigen-binding fragment thereof, or scFv) that binds to B7-H3, such as, e.g., MGA271.
  • a chimeric NDV described herein is engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell described in Section 6, infra.
  • NDV described herein is engineered to express anti-CD28 scvFv, ICOSL, CD40L, OX40L, CD137L, GITRL, and/or CD70.
  • an agonist of a co-stimulatory signal of an immune cell induces (e.g., selectively) induces one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand.
  • an agonist of a co-stimulatory receptor induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands in the absence of the agonist.
  • an agonist of a co-stimulatory receptor induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not induce, or induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to
  • an agonist of a co-stimulatory signal of an immune cell activates or enhances (e.g., selectively activates or enhances) one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand.
  • an agonist of a co-stimulatory receptor activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of co-stimulatory receptor to one or more of its ligands in the absence of the agonist.
  • an agonist of a co-stimulatory receptor activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not activate or enhance, or activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5%
  • an antagonist of an inhibitory signal of an immune cell inhibits or reduces one or more of the signal transduction pathways induced by the binding of an inhibitory receptor to its ligand.
  • an antagonist of an inhibitory receptor inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands in the absence of the antagonist.
  • an antagonist of an inhibitory receptor (i) inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to the one particular ligand in the absence of the antagonist; and (ii) does not inhibit or reduce, or inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or
  • an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell induces, activates and/or enhances one or more immune activities, functions or responses.
  • the one or more immune activities, functions or responses can be in the form of, e.g., an antibody response (humoral response) or a cellular immune response, e.g., cytokine secretion (e.g., interferon-gamma), helper activity or cellular cytotoxicity.
  • expression of an activation marker on immune cells e.g., CD44, Granzyme, or Ki-67
  • expression of a co-stimulatory receptor on immune cells e.g., ICOS, CD28, OX40, or CD27
  • expression of a ligand for a co-stimulatory receptor e.g., B7HRP1, CD80, CD86, OX40L, or CD70
  • cytokine secretion infiltration of immune cells (e.g., T-lymphocytes, B lymphocytes and/or NK cells) to a tumor, antibody production, effector function, T cell activation, T cell differentiation, T cell proliferation, B cell differentiation, B cell proliferation, and/or NK cell proliferation is induced, activated and/or enhanced following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • myeloid-derived suppressor cell tumor infiltration and proliferation, Treg tumor infiltration, activation and proliferation, peripheral blood MDSC and Treg counts are inhibited following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • the NDVs described herein can be generated using the reverse genetics technique.
  • the reverse genetics technique involves the preparation of synthetic recombinant viral RNAs that contain the non-coding regions of the negative-strand, viral RNA which are essential for the recognition by viral polymerases and for packaging signals necessary to generate a mature virion.
  • the recombinant RNAs are synthesized from a recombinant DNA template and reconstituted in vitro with purified viral polymerase complex to form recombinant ribonucleoproteins (RNPs) which can be used to transfect cells.
  • RNPs ribonucleoproteins
  • the synthetic recombinant RNPs can be rescued into infectious virus particles.
  • the foregoing techniques are described in U.S. Pat. No. 5,166,057 issued Nov. 24, 1992; in U.S. Pat. No. 5,854,037 issued Dec. 29, 1998; in U.S. Pat. No. 6,146,642 issued Nov. 14, 2000; in European Patent Publication EP 0702085A1, published Feb. 20, 1996; in U.S. patent application Ser. No. 09/152,845; in International Patent Publications PCT WO97/12032 published Apr. 3, 1997; WO96/34625 published Nov. 7, 1996; in European Patent Publication EP A780475; WO 99/02657 published Jan. 21, 1999; WO 98/53078 published Nov.
  • the helper-free plasmid technology can also be utilized to engineer a NDV described herein.
  • a complete cDNA of a NDV e.g., the Hitchner B1 strain
  • a plasmid vector e.g., the Hitchner B1 strain
  • a nucleotide sequence encoding a heterologous amino acid sequence e.g., a nucleotide sequence encoding an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell
  • a nucleotide sequence encoding a heterologous amino acid sequence may be engineered into a NDV transcription unit so long as the insertion does not affect the ability of the virus to infect and replicate.
  • the single segment is positioned between a T7 promoter and the hepatitis delta virus ribozyme to produce an exact negative transcript from the T7 polymerase.
  • the plasmid vector and expression vectors comprising the necessary viral proteins are transfected into cells leading to production of recombinant viral particles (see, e.g., International Publication No.
  • Bicistronic techniques to produce multiple proteins from a single mRNA are known to one of skill in the art.
  • Bicistronic techniques allow the engineering of coding sequences of multiple proteins into a single mRNA through the use of IRES sequences.
  • IRES sequences direct the internal recruitment of ribozomes to the RNA molecule and allow downstream translation in a cap independent manner.
  • a coding region of one protein is inserted into the ORF of a second protein.
  • the insertion is flanked by an IRES and any untranslated signal sequences necessary for proper expression and/or function.
  • the insertion must not disrupt the open reading frame, polyadenylation or transcriptional promoters of the second protein (see e.g., Garcia-Sastre et al., 1994, J. Virol. 68:6254-6261 and Garcia-Sastre et al., 1994 Dev. Biol. Stand. 82:237-246, each of which are incorporated by reference herein in their entirety).
  • the NDVs described herein can be propagated in any substrate that allows the virus to grow to titers that permit the uses of the viruses described herein.
  • the substrate allows the NDVs described herein (e.g., the chimeric NDVs) to grow to titers comparable to those determined for the corresponding wild-type viruses.
  • the NDVs described herein may be grown in cells (e.g., avian cells, chicken cells, etc.) that are susceptible to infection by the viruses, embryonated eggs (e.g., chicken eggs or quail eggs) or animals (e.g., birds). Such methods are well-known to those skilled in the art.
  • the NDVs described herein may be propagated in cancer cells, e.g., carcinoma cells (e.g., breast cancer cells and prostate cancer cells), sarcoma cells, leukemia cells, lymphoma cells, and germ cell tumor cells (e.g., testicular cancer cells and ovarian cancer cells).
  • the NDVs described herein may be propagated in cell lines, e.g., cancer cell lines such as HeLa cells, MCF7 cells, THP-1 cells, U87 cells, DU145 cells, Lncap cells, and T47D cells.
  • the NDVs described herein are propagated in chicken cells or embryonated eggs. Representative chicken cells include, but are not limited to, chicken embryo fibroblasts and chicken embryo kidney cells.
  • the NDVs described herein are propagated in Vero cells.
  • the NDVs described herein are propagated in cancer cells in accordance with the methods described in Section 6, infra.
  • the NDVs described herein are propagated in chicken eggs or quail eggs.
  • a NDV virus described herein is first propagated in embryonated eggs and then propagated in cells (e.g., a cell line).
  • the NDVs described herein may be propagated in embryonated eggs, e.g., from 6 to 14 days old, 6 to 12 days old, 6 to 10 days old, 6 to 9 days old, 6 to 8 days old, or 10 to 12 days old. Young or immature embryonated eggs can be used to propagate the NDVs described herein (e.g., the chimeric NDVs).
  • Immature embryonated eggs encompass eggs which are less than ten day old eggs, e.g., eggs 6 to 9 days old or 6 to 8 days old that are IFN-deficient.
  • Immature embryonated eggs also encompass eggs which artificially mimic immature eggs up to, but less than ten day old, as a result of alterations to the growth conditions, e.g., changes in incubation temperatures; treating with drugs; or any other alteration which results in an egg with a retarded development, such that the IFN system is not fully developed as compared with ten to twelve day old eggs.
  • the NDVs described herein e.g., the chimeric NDVs
  • the growth and propagation viruses see, e.g., U.S. Pat. No. 6,852,522 and U.S. Pat. No. 7,494,808, both of which are hereby incorporated by reference in their entireties.
  • the NDVs described herein can be removed from cell culture and separated from cellular components, typically by well known clarification procedures, e.g., such as gradient centrifugation and column chromatography, and may be further purified as desired using procedures well known to those skilled in the art, e.g., plaque assays.
  • compositions are used in methods of treating cancer.
  • a NDV described herein e.g., the chimeric NDVs
  • plasma membrane fragments from NDV infected cells or whole cancer cells infected with NDV in compositions.
  • the compositions are pharmaceutical compositions, such as immunogenic formulations (e.g., vaccine formulations).
  • the compositions may be used in methods of treating cancer.
  • a pharmaceutical composition comprises a NDV described herein (e.g., the chimeric NDVs), in an admixture with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition further comprises one or more additional prophylactic or therapeutic agents, such as described in Section 5.6.4, infra.
  • a pharmaceutical composition comprises an effective amount of a NDV described herein (e.g., the chimeric NDVs), and optionally one or more additional prophylactic of therapeutic agents, in a pharmaceutically acceptable carrier.
  • the NDV e.g., a chimeric NDV
  • the NDV is the only active ingredient included in the pharmaceutical composition.
  • a pharmaceutical composition (e.g., an oncolysate vaccine) comprises a protein concentrate or a preparation of plasma membrane fragments from NDV infected cancer cells, in an admixture with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition further comprises one or more additional prophylactic or therapeutic agents, such as described in Section 5.6.4, infra.
  • a pharmaceutical composition (e.g., a whole cell vaccine) comprises cancer cells infected with NDV, in an admixture with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition further comprises one or more additional prophylactic or therapeutic agents, such as described in Section 5.6.4, infra.
  • compositions provided herein can be in any form that allows for the composition to be administered to a subject.
  • the pharmaceutical compositions are suitable for veterinary and/or human administration.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeiae for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the pharmaceutical composition is administered. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin. The formulation should suit the mode of administration.
  • the pharmaceutical compositions are formulated to be suitable for the intended route of administration to a subject.
  • the pharmaceutical composition may be formulated to be suitable for parenteral, intrapleural, inhalation, intraperitoneal, oral, intradermal, colorectal, intraperitoneal and intratumoral administration.
  • the pharmaceutical composition may be formulated for intravenous, oral, intraperitoneal, intranasal, intratracheal, subcutaneous, intramuscular, topical, pulmonary, or intratumoral administration.
  • a chimeric NDV described herein may be used in the treatment of cancer.
  • methods for treating cancer comprising administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof.
  • a method for treating cancer comprising administering to a subject in need thereof an effective amount of a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof.
  • a chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell, or a composition thereof is administered to a subject to treat cancer.
  • a chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell, or a composition thereof is administered to a subject to treat cancer.
  • a chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell and a mutated F protein or a composition thereof is administered to a subject to treat cancer.
  • a chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell and a mutated F protein or a composition thereof is administered to a subject to treat cancer.
  • a chimeric NDV (e.g., a chimeric NDV described in Section 5.2, supra) described herein or a composition thereof, an oncolysate vaccine, or a whole cell cancer vaccine used in a method for treating cancer may be used as any line of therapy (e.g., a first, second, third, fourth or fifth line therapy).
  • a chimeric NDV described herein is the only active ingredient administered to treat cancer.
  • a chimeric NDV described herein is the only active ingredient in a composition administered to treat cancer.
  • the chimeric NDV (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof may be administered locally or systemically to a subject.
  • the chimeric NDV e.g., a chimeric NDV described in Section 5.2, supra
  • composition may be administered parenterally (e.g., intravenously or subcutanously), intratumorally, intrapleurally, intranasally, intraperitoneally, orally, by inhalation, topically or intradermally to a subject.
  • the methods described herein include the treatment of cancer for which no treatment is available.
  • a chimeric NDV described herein e.g., a chimeric NDV described in Section 5.2, supra
  • a composition thereof is administered to a subject to treat cancer as an alternative to other conventional therapies.
  • a method for treating cancer comprising administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof and one or more additional therapies, such as described in Section 5.6.4, infra.
  • one or more therapies are administered to a subject in combination with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof to treat cancer.
  • the additional therapies are currently being used, have been used or are known to be useful in treating cancer.
  • a chimeric NDV described herein e.g., a chimeric NDV described in Section 5.2, supra
  • a composition thereof is administered to a subject in combination with a supportive therapy, a pain relief therapy, or other therapy that does not have a therapeutic effect on cancer.
  • the one or more additional therapies administered in combination with a chimeric NDV described herein is one or more of the therapies described in Section 5.6.4.1, infra.
  • a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) and one or more additional therapies are administered in the same composition.
  • a chimeric NDV and one or more additional therapies are administered in different compositions.
  • two, three or multiple NDVs are administered to a subject to treat cancer.
  • the second or more chimeric NDVs used in accordance with methods described herein that comprise administration of two, three or multiple NDVs to a subject to treat cancer may be naturally occurring chimeric NDVs or engineered chimeric NDVs that have been engineered to express heterologous amino acid sequence (e.g., a cytokine).
  • the first and second chimeric NDVs may be part of the same pharmaceutical composition or different pharmaceutical compositions.
  • first chimeric NDV and the second chimeric NDV are administered by the same route of administration (e.g., both are administered intratumorally or intravenously). In other embodiments, the first chimeric NDV and the second chimeric NDV are administered by different routes of administration (e.g., one is administered intratumorally and the other is administered intravenously).
  • a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell is administered to a patient to treat cancer in combination with a second chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell.
  • a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune is administered in combination with a second chimeric NDV engineered to express one, two or more of the following: a cytokine (e.g., IL-2), a heterologous interferon antagonist, a tumor antigen, a pro-apopototic molecule, and/or anti-apoptotic molecule.
  • the first chimeric NDV, the second chimeric NDV, or both express a mutated F protein that increases the fusogenic activity of the chimeric NDV.
  • the first chimeric NDV, the second chimeric NDV or both express a mutated F protein with a mutation in the cleavage site (such as described herein).
  • a first composition e.g., a pharmaceutical composition
  • a second composition e.g., a pharmaceutical composition
  • a second composition comprising a second chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell
  • a first composition comprising a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune is administered in combination with a second composition (e.g., a pharmaceutical composition) comprising a second chimeric NDV engineered to express one, two or more of the following: a cytokine (e.g., IL-2), a heterologous interferon antagonist, a tumor antigen, a pro-apopototic molecule, and/or anti-apoptotic molecule.
  • a cytokine e.g., IL-2
  • a heterologous interferon antagonist e.g., IL-2
  • the first chimeric NDV, the second chimeric NDV, or both express a mutated F protein that increases the fusogenic activity of the chimeric NDV.
  • the first chimeric NDV, the second chimeric NDV or both express a mutated F protein with a mutation in the cleavage site (such as described herein).
  • an NDV described herein may be used in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra (e.g., Section 5.6.4.1, infra), in the treatment of cancer.
  • additional therapies such as described herein in Section 5.6.4, infra (e.g., Section 5.6.4.1, infra)
  • methods for treating cancer comprising administering to a subject in need thereof an NDV described herein (e.g., an NDV described in Section 5.1, supra) or a composition thereof and one or more additional therapies, such as described herein in Section 5.6.4, infra. (e.g., Section 5.6.4.1).
  • a method for treating cancer comprising administering to a subject in need thereof an effective amount of an NDV described herein (e.g., an NDV described in Section 5.1, supra) or a composition thereof and an effective amount of one or more additional therapies, such as described in Section 5.6.4, infra. (e.g., Section 5.6.4.1).
  • an NDV described herein e.g., an NDV described in Section 5.1, supra
  • one or more additional therapies such as described in Section 5.6.4, infra (e.g., Section 5.6.4.1)
  • an NDV e.g., an NDV described in Section 5.1, supra
  • one or more additional therapies are administered in different compositions.
  • NDV e.g., an NDV described in Section 5.1, supra
  • a composition thereof, an oncolysate vaccine, or a whole cell cancer vaccine in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra may be used as any line of therapy (e.g., a first, second, third, fourth or fifth line therapy) for treating cancer in accordance with a method described herein.
  • whole cancer cells infected with a chimeric NDV described herein can be used to treat cancer.
  • a chimeric NDV described herein e.g., a chimeric NDV described in Section 5.2, supra
  • a cancer cell or a population of cancer cells may be contacted with a cancer cell or a population of cancer cells and the infected cancer cell or population of cancer cells may be administered to a subject to treat cancer.
  • the cancer cells are subjected to gamma radiation prior to infection with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra).
  • the cancer cells are subjected to gamma radiation after infection with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra).
  • the cancer cells are treated prior to administration to a subject so that the cancer cells cannot multiply in the subject.
  • the cancer cells cannot multiply in the subject and the virus cannot infect the subject.
  • the cancer cells are subjected to gamma radiation prior to administration to subject.
  • the cancer cells are sonicated prior to administration to a subject.
  • the cancer cells are treated with mitomycin C prior to administration to a subject.
  • the cancer cells are treated by freezing and thawing prior to administration to a subject.
  • the cancer cells are treated with heat treatment prior to administration to a subject.
  • the cancer cells may be administered locally or systemically to a subject.
  • the cancer cells may be administered parenterally (e.g., intravenously or subcutaneously), intratumorally, intransally, orally, by inhalation, intrapleurally, topically or intradermally to a subject.
  • the cancer cells are administered intratumorally or to the skin (e.g., intradermally) of a subject.
  • the cancer cells used may be autologous or allogeneic.
  • the backbone of the chimeric NDV is a non-lytic strain.
  • the cancer cells may be administered to a subject alone or in combination with an additional therapy.
  • the cancer cells are preferably in a pharmaceutical composition.
  • the cancer cells are administered in combination with one or more additional therapies, such as described in Section 5.6.4, infra.
  • the cancer cells and one or more additional therapies are administered in the same composition.
  • the cancer cells and one or more additional therapies are administered in different compositions.
  • whole cancer cells infected with an NDV described herein may be used in combination with one or more additional therapies described herein in Section 5.6.4, infra, in the treatment of cancer.
  • methods for treating cancer comprising administering to a subject in need thereof whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) in combination with one or more additional therapies described herein in Section 5.6.4, infra.
  • a method for treating cancer comprising administering to a subject in need thereof an effective amount of whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) in combination with an effective amount of one or more additional therapies described in Section 5.6.4, infra.
  • whole cancer cells infected with an NDV described herein e.g., an NDV described in Section 5.1, supra
  • one or more additional therapies described in Section 5.6.4, infra are administered in the same composition.
  • whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) and one or more additional therapies are administered in different compositions.
  • a protein concentrate or plasma membrane preparation from lysed cancer cells infected with a chimeric NDV can be used to treat cancer.
  • a plasma membrane preparation comprising fragments from cancer cells infected with a chimeric NDV described herein can be used to treat cancer.
  • a protein concentrate from cancer cells infected with a chimeric NDV described herein can be used to treat cancer. Techniques known to one of skill in the art may be used to produce the protein concentrate or plasma membrane preparation.
  • a chimeric NDV described herein may be contacted with a cancer cell or a population of cancer cells and the infected cancer cell or population of cancer cells may be lysed using techniques known to one of skill in the art to obtain protein concentrate or plasma membrane fragments of the NDV-infected cancer cells, and the protein concentrate or plasma membrane fragments of the NDV-infected cancer cells may be administered to a subject to treat cancer.
  • the protein concentrate or plasma membrane fragments may be administered locally or systemically to a subject.
  • the protein concentrate or plasma membrane fragments may be administered parenterally, intratumorally, intransally, intrapleurally, orally, by inhalation, topically or intradermally to a subject.
  • a protein concentrate or plasma membrane preparation is administered intratumorally or to the skin (e.g., intradermally) of a subject.
  • the cancer cells used to produce the protein concentrate or plasma membrane preparation may be autologous or allogeneic.
  • the backbone of the chimeric NDV is a lytic strain.
  • the protein concentrate or plasma membrane preparation may be administered to a subject alone or in combination with an additional therapy.
  • the protein concentrate or plasma membrane preparation is preferably in a pharmaceutical composition.
  • the protein concentrate or plasma membrane preparation is administered in combination with one or more additional therapies, such as described in Section 5.6.4, infra (e.g., Section 5.6.4.1)
  • additional therapies such as described in Section 5.6.4, infra (e.g., Section 5.6.4.1)
  • the protein concentrate or plasma membrane preparation and one or more additional therapies are administered in the same composition.
  • the protein concentrate or plasma membrane preparation and one or more additional therapies are administered in different compositions.
  • a protein concentrate or plasma membrane preparation from lysed cancer cells infected with an NDV may be used in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra (e.g., Section 5.6.4.1), in the treatment of cancer.
  • additional therapies such as described herein in Section 5.6.4, infra (e.g., Section 5.6.4.1)
  • methods for treating cancer comprising administering to a subject in need thereof a protein concentrate or plasma membrane preparation from lysed cancer cells infected with an NDV (e.g., an NDV described in Section 5.1, supra) in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra. (e.g., Section 5.6.4.1).
  • a method for treating cancer comprising administering to a subject in need thereof an effective amount of a protein concentrate or plasma membrane preparation from lysed cancer cells infected with an NDV (e.g., an NDV described in Section 5.1, supra) in combination with an effective amount of one or more additional therapies, such as described in Section 5.6.4, infra. (e.g., Section 5.6.4.1).
  • the protein concentrate or plasma membrane preparation and one or more additional therapies such as described in Section 5.6.4, infra, are administered in the same composition.
  • the protein concentrate or plasma membrane preparation and one or more additional therapies are administered in different compositions.
  • the chimeric NDVs described herein can be used to produce antibodies which can be used in diagnostic immunoassays, passive immunotherapy, and the generation of antiidiotypic antibodies.
  • a chimeric NDV described herein e.g., a chimeric NDV described in Section 5.2, supra
  • a subject e.g., a mouse, rat, pig, horse, donkey, bird or human
  • a subject e.g., a mouse, rat, pig, horse, donkey, bird or human
  • an NDV described herein e.g., an NDV described in Section 5.1 or 5.2, supra
  • a subject e.g., a mouse, rat, pig, horse, donkey, bird, or human
  • additional therapies such as described in Section 5.6.4, infra
  • the generated antibodies may be isolated by standard techniques known in the art (e.g., immunoaffinity chromatography, centrifugation, precipitation, etc.) and used in diagnostic immunoassays, passive immunotherapy and generation of antiidiotypic antibodies.
  • the antibodies isolated from subjects administered a chimeric NDV described herein e.g., a chimeric NDV described in Section 5.2, supra
  • isolated from subjects administered an NDV described herein e.g., an NDV described in Section 5.1 or 5.2, supra
  • additional therapies such as described in Section 5.6.4, infra
  • any immunoassay system known in the art may be used for this purpose including but not limited to competitive and noncompetitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assays), “sandwich” immunoassays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays and immunoelectrophoresis assays, to name but a few.
  • radioimmunoassays ELISA (enzyme linked immunosorbent assays), “sandwich” immunoassays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays and immunoelectrophoresis assays, to name
  • an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject suffering from cancer.
  • an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject predisposed or susceptible to cancer.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject diagnosed with cancer.
  • specific examples of the types of cancer are described herein.
  • the subject has metastatic cancer.
  • the subject has stage 1, stage 2 or stage 3 cancer.
  • the subject is in remission.
  • the subject has a recurrence of cancer.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human that is 0 to 6 months old, 6 to 12 months old, 6 to 18 months old, 18 to 36 months old, 1 to 5 years old, 5 to 10 years old, 10 to 15 years old, 15 to 20 years old, 20 to 25 years old, 25 to 30 years old, 30 to 35 years old, 35 to 40 years old, 40 to 45 years old, 45 to 50 years old, 50 to 55 years old, 55 to 60 years old, 60 to 65 years old, 65 to 70 years old, 70 to 75 years old, 75 to 80 years old, 80 to 85 years old, 85 to 90 years old, 90 to 95 years old or 95 to 100 years old.
  • an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human infant.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human toddler.
  • an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human child.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human adult.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to an elderly human.
  • an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject in an immunocompromised state or immunosuppressed state or at risk for becoming immunocompromised or immunosuppressed.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject receiving or recovering from immunosuppressive therapy.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject that has or is at risk of getting cancer.
  • the subject is, will or has undergone surgery, chemotherapy and/or radiation therapy.
  • the patient has undergone surgery to remove the tumor or neoplasm.
  • the patient is administered an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein following surgery to remove a tumor or neoplasm.
  • the patient is administered an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein prior to undergoing surgery to remove a tumor or neoplasm.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject that has, will have or had a tissue transplant, organ transplant or transfusion.
  • an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a patient who has proven refractory to therapies other than the chimeric NDV or composition thereof, oncolysate, whole cell vaccine, or a combination therapy but are no longer on these therapies.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a patient who has proven refractory to chemotherapy.
  • a cancer is refractory to a therapy means that at least some significant portion of the cancer cells are not killed or their cell division arrested.
  • the determination of whether the cancer cells are refractory can be made either in vivo or in vitro by any method known in the art for assaying the effect of a therapy on cancer cells, using the art-accepted meanings of “refractory” in such a context.
  • refractory patient is a patient refractory to a standard therapy.
  • a patient with cancer is refractory to a therapy when the tumor or neoplasm has not significantly been eradicated and/or the symptoms have not been significantly alleviated.
  • the determination of whether a patient is refractory can be made either in vivo or in vitro by any method known in the art for assaying the effectiveness of a treatment of cancer, using art-accepted meanings of “refractory” in such a context.
  • the patient to be treated in accordance with the methods described herein is a patient already being treated with antibiotics, anti-virals, anti-fungals, or other biological therapy/immunotherapy or anti-cancer therapy.
  • these patients are refractory patients, and patients who are too young for conventional therapies.
  • the subject being administered an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein has not received therapy prior to the administration of the chimeric NDV or composition, the oncolysate vaccine, or the whole cell vaccine, or the combination therapy.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a patient to prevent the onset of cancer in a patient at risk of developing cancer.
  • compounds are administered to a patient who are susceptible to adverse reactions to conventional therapies.
  • the subject being administered an NDV e.g., a chimeric NDV or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein has not received prior therapy.
  • an NDV e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject who has received a therapy prior to administration of the NDV (e.g., a chimeric NDV) or composition, the oncolysate vaccine, the whole cell vaccine, or the combination therapy.
  • the subject administered an NDV e.g., a chimeric NDV
  • an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein experienced adverse side effects to a prior therapy or a prior therapy was discontinued due to unacceptable levels of toxicity to the subject.
  • an NDV or a composition thereof, an oncolysate vaccine, or a whole cell vaccine which will be effective in the treatment of cancer will depend on the nature of the cancer, the route of administration, the general health of the subject, etc. and should be decided according to the judgment of a medical practitioner. Standard clinical techniques, such as in vitro assays, may optionally be employed to help identify optimal dosage ranges.
  • suitable dosage ranges of an NDV for administration are generally about 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 5 ⁇ 10 4 , 10 5 , 5 ⁇ 10 5 , 10 6 , 5 ⁇ 10 6 , 10 7 , 5 ⁇ 10 7 , 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , 1 ⁇ 10 1 °, 5 ⁇ 10 10 , 1 ⁇ 10 11 , 5 ⁇ 10 11 or 10 12 pfu, and most preferably about 10 4 to about 10 12 , 10 6 to 10 12 , 10 8 to 10 12 , 10 9 to 10 12 or 10 9 to 10 11 , and can be administered to a subject once, twice, three, four or more times with intervals as often as needed.
  • Dosage ranges of oncolysate vaccines for administration may include 0.001 mg, 0.005 mg, 0.01 mg, 0.05 mg. 0.1 mg. 0.5 mg, 1.0 mg, 2.0 mg. 3.0 mg, 4.0 mg, 5.0 mg, 10.0 mg, 0.001 mg to 10.0 mg, 0.01 mg to 1.0 mg, 0.1 mg to 1 mg, and 0.1 mg to 5.0 mg, and can be administered to a subject once, twice, three or more times with intervals as often as needed.
  • Dosage ranges of whole cell vaccines for administration may include 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 5 ⁇ 10 4 , 10 5 , 5 ⁇ 10 5 , 10 6 , 5 ⁇ 10 6 , 10 7 , 5 ⁇ 10 7 , 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , 1 ⁇ 10 10 , 5 ⁇ 10 10 , 1 ⁇ 10 11 , 5 ⁇ 10 11 or 10 12 cells, and can be administered to a subject once, twice, three or more times with intervals as often as needed.
  • dosages similar to those currently being used in clinical trials for NDV, oncolysate vaccines or whole cell vaccines are administered to a subject. Effective doses may be extrapolated from dose response curves derived from in vitro or animal model test systems.
  • an NDV e.g., a chimeric NDV
  • a composition thereof is administered to a subject as a single dose followed by a second dose 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 1 to 3 weeks, 1 to 2 weeks later.
  • booster inoculations may be administered to the subject at 6 to 12 month intervals following the second inoculation.
  • an oncolysate vaccine or a whole cell vaccine is administered to a subject as a single dose followed by a second dose 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 1 to 3 weeks, 1 to 2 weeks later.
  • administration of the same NDV e.g., chimeric NDV
  • a composition thereof, oncolysate vaccine, or whole cell vaccine may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 6 says, 7 days, 10 days, 14 days, 15 days, 21 days, 28 days, 30 days, 45 days, 2 months, 75 days, 3 months, or at least 6 months.
  • administration of the same NDV (e.g., a NDV) or a composition thereof, oncolysate vaccine, or whole cell vaccine may be repeated and the administrations may be separated by 1 to 14 days, 1 to 7 days, 7 to 14 days, 1 to 30 days, 15 to 30 days, 15 to 45 days, 15 to 75 days, 15 to 90 days, 1 to 3 months, 3 to 6 months, 3 to 12 months, or 6 to 12 months.
  • a first NDV e.g., a first chimeric NDV
  • a composition thereof is administered to a subject followed by the administration of a second NDV (e.g., a second chimeric NDV) or a composition thereof.
  • the first and second NDVs (e.g., the first and second chimeric NDVs) or compositions thereof may be separated by at least 1 day, 2 days, 3 days, 5 days, 6 days, 7 days, 10 days, 14 days, 15 days, 21 days, 28 days, 30 days, 45 days, 2 months, 75 days, 3 months, or at least 6 months.
  • the first and second NDVs (e.g., the first and second chimeric NDVs) or compositions thereof may be separated by 1 to 14 days, 1 to 7 days, 7 to 14 days, 1 to 30 days, 15 to 30 days, 15 to 45 days, 15 to 75 days, 15 to 90 days, 1 to 3 months, 3 to 6 months, 3 to 12 months, or 6 to 12 months.
  • an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject in combination with one or more additional therapies, such as a therapy described in Section 5.6.4, infra.
  • the dosage of the other one or more additional therapies will depend upon various factors including, e.g., the therapy, the nature of the cancer, the route of administration, the general health of the subject, etc. and should be decided according to the judgment of a medical practitioner.
  • the dose of the other therapy is the dose and/or frequency of administration of the therapy recommended for the therapy for use as a single agent is used in accordance with the methods disclosed herein.
  • the dose of the other therapy is a lower dose and/or less frequent administration of the therapy than recommended for the therapy for use as a single agent is used in accordance with the methods disclosed herein.
  • Recommended doses for approved therapies can be found in the Physician's Desk Reference.
  • an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject concurrently with the administration of one or more additional therapies.
  • an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject every 3 to 7 days, 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 2 to 4 weeks, 1 to 3 weeks, or 1 to 2 weeks and one or more additional therapies (such as described in Section 5.6.4, infra) is administered every 3 to 7 days, 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 1 to 3 weeks, or 1 to 2 weeks.
  • an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject every 1 to 2 weeks and one or more additional therapies (such as described in Section 5.6.4, infra) is administered every 2 to 4 weeks.
  • an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject every week and one or more additional therapies (such as described in Section 5.6.4, infra) is administered every 2 weeks.
  • cancers that can be treated in accordance with the methods described herein include, but are not limited to: leukemias, such as but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias, such as, myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia leukemias and myelodysplastic syndrome; chronic leukemias, such as but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as but not limited to Hodgkin's disease, non-Hodgkin's disease; multiple myelomas such as but not limited to smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, placancer cell leukemia, s
  • cancers include myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangioendotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma and papillary adenocarcinomas (for a review of such disorders, see Fishman et al., 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al., 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).
  • the chimeric NDVs described herein or compositions thereof, an oncolysate vaccine described herein, a whole cell vaccine herein, or a combination therapy described herein are useful in the treatment of a variety of cancers and abnormal proliferative diseases, including (but not limited to) the following: carcinoma, including that of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid and skin; including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T cell lymphoma, Burkitt's lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyo
  • cancers associated with aberrations in apoptosis are treated in accordance with the methods described herein.
  • Such cancers may include, but are not limited to, follicular lymphomas, carcinomas with p53 mutations, hormone dependent tumors of the breast, prostate and ovary, and precancerous lesions such as familial adenomatous polyposis, and myelodysplastic syndromes.
  • malignancy or dysproliferative changes such as metaplasias and dysplasias
  • hyperproliferative disorders of the skin, lung, liver, bone, brain, stomach, colon, breast, prostate, bladder, kidney, pancreas, ovary, and/or uterus are treated in accordance with the methods described herein.
  • a sarcoma or melanoma is treated in accordance with the methods described herein.
  • the cancer being treated in accordance with the methods described herein is leukemia, lymphoma or myeloma (e.g., multiple myeloma).
  • leukemias and other blood-borne cancers that can be treated in accordance with the methods described herein include, but are not limited to, acute lymphoblastic leukemia “ALL”, acute lymphoblastic B-cell leukemia, acute lymphoblastic T-cell leukemia, acute myeloblastic leukemia “AML”, acute promyelocytic leukemia “APL”, acute monoblastic leukemia, acute erythroleukemic leukemia, acute megakaryoblastic leukemia, acute myelomonocytic leukemia, acute nonlymphocyctic leukemia, acute undifferentiated leukemia, chronic myelocytic leukemia “CIVIL”, chronic lymphocytic leukemia “CLL”, and hairy cell leukemia.
  • ALL acute lymphoblastic leukemia
  • ALL
  • lymphomas that can be treated in accordance with the methods described herein include, but are not limited to, Hodgkin's disease, non-Hodgkin's Lymphoma, Multiple myeloma, Waldenstrom's macroglobulinemia, Heavy chain disease, and Polycythemia vera.
  • the cancer being treated in accordance with the methods described herein is a solid tumor.
  • solid tumors that can be treated in accordance with the methods described herein include, but are not limited to fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, colorectal cancer, kidney cancer, pancreatic cancer, bone cancer, breast cancer, ovarian cancer, prostate cancer, esophageal cancer, stomach cancer, oral cancer, nasal cancer, throat cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, pa
  • the cancer being treated in accordance with the methods described herein is a cancer that has a poor prognosis and/or has a poor response to conventional therapies, such as chemotherapy and radiation.
  • the cancer being treated in accordance with the methods described herein is malignant melanoma, malignant glioma, renal cell carcinoma, pancreatic adenocarcinoma, malignant pleural mesothelioma, lung adenocarcinoma, lung small cell carcinoma, lung squamous cell carcinoma, anaplastic thyroid cancer, and head and neck squamous cell carcinoma.
  • Additional therapies that can be used in a combination with an NDV described herein or a composition thereof, an oncolysate vaccine, or a whole cell vaccine for the treatment of cancer include, but are not limited to, small molecules, synthetic drugs, peptides (including cyclic peptides), polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides or peptides), antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules.
  • the additional therapy is a chemotherapeutic agent.
  • an NDV described herein or a composition thereof, an oncolysate vaccine, or a whole cell vaccine is used in combination with radiation therapy comprising the use of x-rays, gamma rays and other sources of radiation to destroy cancer cells.
  • the radiation therapy is administered as external beam radiation or teletherapy, wherein the radiation is directed from a remote source.
  • the radiation therapy is administered as internal therapy or brachytherapy wherein a radioactive source is placed inside the body close to cancer cells and/or a tumor mass.
  • anti-cancer agents that may be used in combination with an NDV described herein or a composition thereof include: hormonal agents (e.g., aromatase inhibitor, selective estrogen receptor modulator (SERM), and estrogen receptor antagonist), chemotherapeutic agents (e.g., microtubule disassembly blocker, antimetabolite, topoisomerase inhibitor, and DNA crosslinker or damaging agent), anti-angiogenic agents (e.g., VEGF antagonist, receptor antagonist, integrin antagonist, vascular targeting agent (VTA)/vascular disrupting agent (VDA)), radiation therapy, and conventional surgery.
  • hormonal agents e.g., aromatase inhibitor, selective estrogen receptor modulator (SERM), and estrogen receptor antagonist
  • chemotherapeutic agents e.g., microtubule disassembly blocker, antimetabolite, topoisomerase inhibitor, and DNA crosslinker or damaging agent
  • anti-angiogenic agents e.g., VEGF antagonist, receptor antagonist, integrin antagonist,
  • Non-limiting examples of hormonal agents that may be used in combination with an NDV described herein or a composition thereof include aromatase inhibitors, SERMs, and estrogen receptor antagonists.
  • Hormonal agents that are aromatase inhibitors may be steroidal or nonsteroidal.
  • Non-limiting examples of nonsteroidal hormonal agents include letrozole, anastrozole, aminoglutethimide, fadrozole, and vorozole.
  • Non-limiting examples of steroidal hormonal agents include aromasin (exemestane), formestane, and testolactone.
  • Non-limiting examples of hormonal agents that are SERMs include tamoxifen (branded/marketed as Nolvadex®), afimoxifene, arzoxifene, avalycoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, and toremifene.
  • Non-limiting examples of hormonal agents that are estrogen receptor antagonists include fulvestrant.
  • Other hormonal agents include but are not limited to abiraterone and lonaprisan.
  • Non-limiting examples of chemotherapeutic agents that may be used in combination with an NDV described herein or a composition thereof, an oncolysate vaccine, or a whole cell vaccine include microtubule disasssembly blocker, antimetabolite, topoisomerase inhibitor, and DNA crosslinker or damaging agent.
  • Chemotherapeutic agents that are microtubule disassembly blockers include, but are not limited to, taxenes (e.g., paclitaxel (branded/marketed as TAXOL®), docetaxel, abraxane, larotaxel, ortataxel, and tesetaxel); epothilones (e.g., ixabepilone); and vinca alkaloids (e.g., vinorelbine, vinblastine, vindesine, and vincristine (branded/marketed as ONCOVIN®)).
  • taxenes e.g., paclitaxel (branded/marketed as TAXOL®), docetaxel, abraxane, larotaxel, ortataxel, and tesetaxel
  • epothilones e.g., ixabepilone
  • vinca alkaloids e.g., vinorelbine, vinblastine, vindesine, and vin
  • Chemotherapeutic agents that are antimetabolites include, but are not limited to, folate antimetabolites (e.g., methotrexate, aminopterin, pemetrexed, raltitrexed); purine antimetabolites (e.g., cladribine, clofarabine, fludarabine, mercaptopurine, pentostatin, thioguanine); pyrimidine antimetabolites (e.g., 5-fluorouracil, capecitabine, gemcitabine) (GEMZAR®, cytarabine, decitabine, floxuridine, tegafur); and deoxyribonucleotide antimetabolites (e.g., hydroxyurea).
  • folate antimetabolites e.g., methotrexate, aminopterin, pemetrexed, raltitrexed
  • purine antimetabolites e.g., cladribine, clofarabine, fludarabine, mercapto
  • Chemotherapeutic agents that are topoisomerase inhibitors include, but are not limited to, class I (camptotheca) topoisomerase inhibitors (e.g., topotecan (branded/marketed as HYCAMTIN®) irinotecan, rubitecan, and belotecan); class II (podophyllum) topoisomerase inhibitors (e.g., etoposide or VP-16, and teniposide); anthracyclines (e.g., doxorubicin, epirubicin, Doxil, aclarubicin, amrubicin, daunorubicin, idarubicin, pirarubicin, valrubicin, and zorubicin); and anthracenediones (e.g., mitoxantrone, and pixantrone).
  • class I camptotheca
  • topotecan branded/marketed as HYCAMTIN®
  • irinotecan ir
  • Chemotherapeutic agents that are DNA crosslinkers include, but are not limited to, alkylating agents (e.g., cyclophosphamide, mechlorethamine, ifosfamide (branded/marketed as IFEX®), trofosfamide, chlorambucil, melphalan, prednimustine, bendamustine, uramustine, estramustine, carmustine (branded/marketed as BiCNU®), lomustine, semustine, fotemustine, nimustine, ranimustine, streptozocin, busulfan, mannosulfan, treosulfan, carboquone, N,N′N′-triethylenethiophosphoramide, triaziquone, triethylenemelamine); alkylating-like agents (e.g., carboplatin (branded/marketed as PARAPLATIN®), cisplatin, oxaliplatin, nedaplatin, triplatin tetranit
  • an NDV described herein e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the following: any agonist of a co-stimulatory signal of an immune cell (such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage) and/or any antagonist of an inhibitory signal of an immune cell (such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage), known to one of skill in the art.
  • any agonist of a co-stimulatory signal of an immune cell such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage)
  • an NDV described herein e.g., a chimeric NDV
  • an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the agonists of a co-stimulatory signal of an immune cell described in Section 5.2.1, supra.
  • an NDV described herein e.g., a chimeric NDV
  • an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the antagonists of an inhibitory signal of an immune cell described in Section 5.2.1, supra.
  • an NDV described herein e.g., a chimeric NDV
  • a composition thereof, an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the agonists of a co-stimulatory signal of an immune cell and/or one or more of the antagonists of an inhibitory signal of an immune cell described in Section 6, infra (e.g., an anti-CTLA-4 antibody or an ICOS-L)
  • Viral assays include those that measure altered viral replication (as determined, e.g., by plaque formation) or the production of viral proteins (as determined, e.g., by western blot analysis) or viral RNAs (as determined, e.g., by RT-PCR or northern blot analysis) in cultured cells in vitro using methods which are well known in the art.
  • NDVs described herein can be assessed by any method known in the art or described herein (e.g., in cell culture (e.g., cultures of chicken embryonic kidney cells or cultures of chicken embryonic fibroblasts (CEF)).
  • Viral titer may be determined by inoculating serial dilutions of a NDV described herein into cell cultures (e.g., CEF, MDCK, EFK-2 cells, Vero cells, primary human umbilical vein endothelial cells (HUVEC), H292 human epithelial cell line or HeLa cells), chick embryos, or live animals (e.g., avians). After incubation of the virus for a specified time, the virus is isolated using standard methods.
  • Physical quantitation of the virus titer can be performed using PCR applied to viral supernatants (Quinn & Trevor, 1997; Morgan et al., 1990), hemagglutination assays, tissue culture infectious doses (TCID50) or egg infectious doses (EID50).
  • TCID50 tissue culture infectious doses
  • EID50 egg infectious doses
  • incorporación of nucleotide sequences encoding a heterologous peptide or protein e.g., a cytokine, a mutated F protein, a mutated V protein, or miRNA target site into the genome of a chimeric NDV described herein can be assessed by any method known in the art or described herein (e.g., in cell culture, an animal model or viral culture in embryonated eggs).
  • a heterologous peptide or protein e.g., a cytokine, a mutated F protein, a mutated V protein, or miRNA target site into the genome of a chimeric NDV described herein
  • a heterologous peptide or protein e.g., a cytokine, a mutated F protein, a mutated V protein, or miRNA target site into the genome of a chimeric NDV described herein
  • viral particles from cell culture of the allantoic fluid of embryonated eggs can be purified by centri
  • Immunofluorescence-based approaches may also be used to detect virus and assess viral growth. Such approaches are well known to those of skill in the art, e.g., fluorescence microscopy and flow cytometry (see Section 6, infra).
  • Antibodies generated by the NDVs described herein may be characterized in a variety of ways well-known to one of skill in the art (e.g., ELISA, Surface Plasmon resonance display (BIAcore), Western blot, immunofluorescence, immunostaining and/or microneutralization assays).
  • antibodies generated by the chimeric NDVs described herein may be assayed for the ability to specifically bind to an antigen of the virus or a heterologous peptide or protein.
  • Such an assay may be performed in solution (e.g., Houghten, 1992, Bio/Techniques 13:412 421), on beads (Lam, 1991, Nature 354:82 84), on chips (Fodor, 1993, Nature 364:555 556), on bacteria (U.S. Pat. No. 5,223,409), on spores (U.S. Pat. Nos. 5,571,698; 5,403,484; and 5,223,409), on plasmids (Cull et al., 1992, Proc. Natl. Acad. Sci. USA 89:1865 1869) or on phage (Scott and Smith, 1990, Science 249:386 390; Cwirla et al., 1990, Proc. Natl. Acad. Sci. USA 87:6378 6382; and Felici, 1991, J. Mol. Biol. 222:301 310) (each of these references is incorporated herein in its entirety by reference).
  • Antibodies generated by the chimeric NDVs described herein that have been identified to specifically bind to an antigen of the virus or a heterologous peptide or protein can be assayed for their specificity to said antigen of the virus or heterologous peptide or protein.
  • the antibodies may be assayed for specific binding to an antigen of the virus or a heterologous peptide or protein and for their cross-reactivity with other antigens by any method known in the art.
  • Immunoassays which can be used to analyze specific binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement
  • the binding affinity of an antibody to an antigen and the off-rate of an antibody-antigen interaction can be determined by competitive binding assays.
  • a surface plasmon resonance assay e.g., BIAcore kinetic analysis
  • KinExA assay Blake, et al., Analytical Biochem., 1999, 272:123-134 may be used to determine the binding on and off rates of antibodies to an antigen of the chimeric NDVs described herein.
  • IFN induction and release by an NDV described herein may be determined using techniques known to one of skill in the art or described herein.
  • the amount of IFN induced in cells following infection with an NDV described herein may be determined using an immunoassay (e.g., an ELISA or Western blot assay) to measure IFN expression or to measure the expression of a protein whose expression is induced by IFN.
  • the amount of IFN induced may be measured at the RNA level by assays, such as Northern blots and quantitative RT-PCR, known to one of skill in the art.
  • the amount of IFN released may be measured using an ELISPOT assay. (See, e.g., the methods described in Section 6, below.)
  • an activation marker, co-stimulatory molecule, ligand, or inhibitory molecule by immune cells are known to one of skill in the art.
  • the expression of an activation marker, co-stimulatory molecule, ligand, or inhibitory molecule by an immune cell can be assessed by flow cytometry.
  • techniques described in Section 6, infra are used to assess the expression of an activation marker, co-stimulatory molecule, ligand, or inhibitory molecule by an immune cell.
  • the NDVs described herein or compositions thereof, oncolysate vaccines described herein, whole cell vaccines described herein, or combination therapies described herein are tested for cytotoxicity in mammalian, preferably human, cell lines (see, e.g., the cytotoxicity assay described in Section 6, infra).
  • cytotoxicity is assessed in one or more of the following non-limiting examples of cell lines: U937, a human monocyte cell line; primary peripheral blood mononuclear cells (PBMC); Huh7, a human hepatoblastoma cell line; HL60 cells, HT1080, HEK 293T and 293H, MLPC cells, human embryonic kidney cell lines; human melanoma cell lines, such as SkMe12, SkMe1-119 and SkMe1-197; THP-1, monocytic cells; a HeLa cell line; and neuroblastoma cells lines, such as MC-IXC, SK-N-MC, SK-N-MC, SK-N-DZ, SH-SY5Y, and BE(2)-C.
  • cytotoxicity is assessed in various cancer cells.
  • the ToxLite assay is used to assess cytotoxicity.
  • cell proliferation can be assayed by measuring Bromodeoxyuridine (BrdU) incorporation, ( 3 H) thymidine incorporation, by direct cell count, or by detecting changes in transcription, translation or activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers (Rb, cdc2, cyclin A, D1, D2, D3, E, etc).
  • PrdU Bromodeoxyuridine
  • 3 H thymidine incorporation
  • thymidine incorporation by direct cell count, or by detecting changes in transcription, translation or activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers (Rb, cdc2, cyclin A, D1, D2, D3, E, etc).
  • the levels of such protein and mRNA and activity can be determined by any method well known in the art.
  • protein can be quantitated by known immunodiagnostic methods such as ELISA, Western blotting or immuno
  • mRNA can be quantitated using methods that are well known and routine in the art, for example, using northern analysis, RNase protection, or polymerase chain reaction in connection with reverse transcription.
  • Cell viability can be assessed by using trypan-blue staining or other cell death or viability markers known in the art.
  • the level of cellular ATP is measured to determined cell viability.
  • an NDV described herein or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy kills cancer cells but does not kill healthy (i.e., non-cancerous) cells.
  • an NDV described herein or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy preferentially kills cancer cells but does not kill healthy (i.e., non-cancerous) cells.
  • cell viability is measured in three-day and seven-day periods using an assay standard in the art, such as the CellTiter-Glo Assay Kit (Promega) which measures levels of intracellular ATP. A reduction in cellular ATP is indicative of a cytotoxic effect.
  • cell viability can be measured in the neutral red uptake assay.
  • visual observation for morphological changes may include enlargement, granularity, cells with ragged edges, a filmy appearance, rounding, detachment from the surface of the well, or other changes.
  • the NDVs described herein or compositions thereof, oncolysate vaccines, whole cell vaccines or combination therapies can be tested for in vivo toxicity in animal models (see, e.g., the animal models described in Section 6, below).
  • animal models, described herein and/or others known in the art, used to test the effects of compounds on cancer can also be used to determine the in vivo toxicity of the NDVs described herein or compositions thereof, oncolysate vaccines, whole cell vaccines, or combination therapies.
  • animals are administered a range of pfu of an NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra).
  • the animals are monitored over time for lethality, weight loss or failure to gain weight, and/or levels of serum markers that may be indicative of tissue damage (e.g., creatine phosphokinase level as an indicator of general tissue damage, level of glutamic oxalic acid transaminase or pyruvic acid transaminase as indicators for possible liver damage).
  • tissue damage e.g., creatine phosphokinase level as an indicator of general tissue damage, level of glutamic oxalic acid transaminase or pyruvic acid transaminase as indicators for possible liver damage.
  • serum markers e.g., creatine phosphokinase level as an indicator of general tissue damage, level of glutamic oxalic acid transaminase or pyruvic acid transaminase as indicators for possible liver damage.
  • the toxicity and/or efficacy of an NDV described herein or a composition thereof, an oncolysate vaccine described herein, a whole cell vaccine described herein, or a combination therapy described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Therapies that exhibits large therapeutic indices are preferred. While therapies that exhibits toxic side effects may be used, care should be taken to design a delivery system that targets such therapies to the site of affected tissue in order to minimize potential damage to noncancerous cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the therapies for use in subjects.
  • the dosage of such agents lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the chimeric NDV that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC50 i.e., the concentration of the chimeric NDV that achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the NDVs described herein or compositions thereof, oncolysate vaccines described herein, whole cell vaccines described herein, or combination therapies described herein can be tested for biological activity using animal models for cancer.
  • animal model systems include, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, dogs, rabbits, etc.
  • the anti-cancer activity of an NDV described herein or combination therapy is tested in a mouse model system.
  • Such model systems are widely used and well-known to the skilled artisan such as the SCID mouse model or transgenic mice.
  • the anti-cancer activity of an NDV described herein or a composition thereof, oncolysate vaccine described herein, whole cell vaccine described herein, or a combination therapy described herein can be determined by administering the NDV or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy to an animal model and verifying that the NDV or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy is effective in reducing the severity of cancer, reducing the symptoms of cancer, reducing cancer metastasis, and/or reducing the size of a tumor in said animal model (see, e.g., Section 6, below).
  • animal models for cancer in general include, include, but are not limited to, spontaneously occurring tumors of companion animals (see, e.g., Vail & MacEwen, 2000, Cancer Invest 18(8):781-92).
  • animal models for lung cancer include, but are not limited to, lung cancer animal models described by Zhang & Roth (1994, In-vivo 8(5):755-69) and a transgenic mouse model with disrupted p53 function (see, e.g. Morris et al., 1998, J La State Med Soc 150(4): 179-85).
  • An example of an animal model for breast cancer includes, but is not limited to, a transgenic mouse that over expresses cyclin D1 (see, e.g., Hosokawa et al., 2001, Transgenic Res 10(5):471-8).
  • An example of an animal model for colon cancer includes, but is not limited to, a TCR b and p53 double knockout mouse (see, e.g., Kado et al., 2001, Cancer Res. 61(6):2395-8).
  • Examples of animal models for pancreatic cancer include, but are not limited to, a metastatic model of PancO2 murine pancreatic adenocarcinoma (see, e.g., Wang et al., 2001, Int. J.
  • Pancreatol. 29(1):37-46) and nu-nu mice generated in subcutaneous pancreatic tumors see, e.g., Ghaneh et al., 2001, Gene Ther. 8(3):199-208.
  • animal models for non-Hodgkin's lymphoma include, but are not limited to, a severe combined immunodeficiency (“SCID”) mouse (see, e.g., Bryant et al., 2000, Lab Invest 80(4):553-73) and an IgHmu-HOX11 transgenic mouse (see, e.g., Hough et al., 1998, Proc. Natl. Acad. Sci. USA 95(23):13853-8).
  • SCID severe combined immunodeficiency
  • an animal model for esophageal cancer includes, but is not limited to, a mouse transgenic for the human papillomavirus type 16 E7 oncogene (see, e.g., Herber et al., 1996, J. Virol. 70(3):1873-81).
  • animal models for colorectal carcinomas include, but are not limited to, Apc mouse models (see, e.g., Fodde & Smits, 2001, Trends Mol Med 7(8):369 73 and Kuraguchi et al., 2000).
  • the animal models for cancer described in Section 6, infra are used to assess efficacy of an NDV or composition thereof, an oncolysate, a whole cell vaccine, or a combination therapy.
  • This example demonstrates the therapeutic efficacy of NDV therapy in combination with immune checkpoint modulators that are immunostimulatory in the treatment of cancer.
  • mice BALB/c mice (6-8 weeks old), and WT C57BL/6 mice were purchased from Jackson Laboratory. All mice were maintained in microisolator cages and treated in accordance with the NIH and American Association of Laboratory Animal Care regulations. All mouse procedures and experiments for this study were approved by the Memorial Sloan-Kettering Cancer Center Institutional Animal Care and Use Committee.
  • the murine cancer cell lines for melanoma (B16-F10), and colon carcinoma (CT26 and MC38) were maintained in RPMI medium supplemented with 10% fetal calf serum and penicillin with streptomycin.
  • the murine prostate cancer cell line TRAMP-C2 was maintained in DMEM medium supplemented with 5% fetal calf serum (FCS; Mediatech, Inc.). 5% Nu Serum IV (BD Biosciences) HEPES, 2-ME, pen/strep, L-glut, 5 ⁇ g/mL insulin (Sigma), and 10 nmol/L DHT (Sigma).
  • Therapeutic anti-CTLA-4 (clone 9H10), anti-PD-1 (clone RMP1-14), and anti-PD-L1 monoclonal antibodies were produced by BioXcell. Antibodies used for flow cytometry were purchased from eBioscience, Biolegend, Invitrogen, and BD Pharmingen.
  • NDV LaSota strain Recombinant lentogenic NDV LaSota strain was used for all experiments.
  • a DNA fragment encoding the murine ICOSL flanked by the appropriate NDV-specific RNA transcriptional signals was inserted into the SacII site created between the P and M genes of pT7NDV/LS.
  • Viruses were rescued from cDNA using methods described previously and sequenced by reverse transcription PCR for insert fidelity.
  • Virus titers were determined by serial dilution and immunofluorescence in Vero cells.
  • Recombinant ICOSL-F fusion construct was generated by PCR amplification of the ICOSL DNA encoding the extracellular domain (amino acids 1-277) with flanking EcoRI and MluI restriction sites, and the NDV F DNA encoding the F transmembrane and intracellular domains (amino acids 501-554) with flanking MluI and XhoI restriction sites.
  • the resultant DNA fragments were assembled in pCAGGS vector utilizing 3-part ligation.
  • B16-F10 cells were infected in 6-well dishes at MOI 2 in triplicate. Twenty-four hours later, the cells were harvested by mechanical scraping and processed for surface labeling and quantification by flow cytometry. For virus growth curve experiments, B16-F10 cells were incubated at room temperature with the virus in 6-well culture dishes at the indicated MOIs in a total volume of 100 ⁇ l. One hour after the incubation, the infection media was aspirated and the cells were incubated at 37° C. in 1 ml of DMEM with 10% chick allantoic fluid.
  • Bilateral flank tumor models were established to monitor for therapeutic efficacy in both injected and systemic tumors. Treatment schedules and cell doses were established for each tumor model to achieve 10-20% tumor clearance by NDV or anti-CTLA-4/anti-PD-1 as single agents.
  • NDV-WT wild-type NDV
  • B16F10 tumors were implanted by injection of 2 ⁇ 10 5 B16F10 cells in the right flank i.d. on day 0 and 5 ⁇ 10 4 cells in the left flank on day 4. On days 7, 10, 13, and 16 the mice were treated with 4 intratumoral injections of 2 ⁇ 10 7 pfu of NDV in PBS in a total volume of 100 ⁇ l.
  • mice received 4 i.p. injections of anti-CTLA-4 antibody (100 ⁇ g) or anti-PD-1 antibody (250 ⁇ g).
  • Control groups received a corresponding dose of isotype antibody i.p. and intratumoral injection of PBS. Tumor size and incidence were monitored over time by measurement with a caliper.
  • TRAMP-C2 For the TRAMP-C2 model, 5 ⁇ 10 5 cells were implanted in right flank on day 0 and 5 ⁇ 10 5 cells were implanted in the left flank on day 8. Treatment was performed on days 11, 14, 17, and 20 in the similar fashion to above.
  • B16F10 tumors were implanted by injection of 2 ⁇ 10 5 B16F10 cells in the right flank i.d. on day 0 and 1 ⁇ 10 5 cells in the left flank on day 4. Treatment was carried out as above.
  • tumors were implanted by injection of 1 ⁇ 10 6 CT26 cells in the right flank i.d. on day 0 and 1 ⁇ 10 6 cells in the left flank on day 2. Treatment was carried out as above on days 6, 9, and 12.
  • B16F10 tumors were implanted by injection of 2 ⁇ 10 5 B16F10 cells in the right flank i.d. on day 0 and 2 ⁇ 10 5 cells in the left flank on day 4.
  • the mice were treated with 3 intratumoral injections of 2 ⁇ 10 7 pfu of NDV, and 100 ⁇ g of i.p. anti-CTLA-4 antibody or 250 ⁇ g of i.p. anti-PD-1 antibody, where specified.
  • mice were sacrificed by CO2 inhalation. Tumors and tumor-draining lymph nodes were removed using forceps and surgical scissors and weighed.
  • Tumors from each group were minced with scissors prior to incubation with 1.67 Wünsch 1J/int, Liberase and 0.2 mg/mL DNase for 30 minutes at 37° C. Tumors were homogenized by repeated pipetting and filtered through a 70- ⁇ m nylon filter. Cell suspensions were washed once with complete RPMI and purified on a Ficoll gradient to eliminate dead cells. Cells from tumor draining lymph nodes were isolated by grinding the lymph nodes through a 70- ⁇ m nylon filter.
  • Cells isolated from tumors or tumor-draining lymph nodes were processed for surface labeling with several antibody panels staining CD45, CD3, CD4, CD8, CD44, PD-1, ICOS, CD11c, CD19, NK1.1, CD11b, F4/80, Ly6C and Ly6G.
  • Fixable viability dye eFluor780 (eBioscience) was used to distinguish the live cells.
  • Cells were further permeabilized using FoxP3 fixation and permeabilization kit (eBioscience) and stained for Ki-67, FoxP3, Granzyme B, CTLA-4, and IFN gamma. Data was acquired using the LSRII Flow cytometer (BD Biosciences) and analyzed using FlowJo software (Treestar).
  • Spleens from na ⁇ ve mice were isolated and digested with 1.67 Wunsch U/mL Liberase and 0.2 mg/mL DNase for 30 minutes at 37° C. The resulting cell suspensions were filtered through 70 ⁇ m nylon filter and washed once with complete RPMI. CD11c+ dendritic cells were purified by positive selection using Miltenyi magnetic beads. Isolated dendritic cells were cultured overnight with recombinant GM-CSF and B16-F10 tumor lysates and were purified on Ficoll gradient.
  • T cells Cell suspensions from tumors or tumor-draining lymph nodes were pooled and enriched for T cells using a Miltenyi T-cell purification kit. Isolated T cells were counted and co-cultured for 8 hours with dendritic cells loaded with B16-F10 tumor cell lysates in the presence of 20 U/ml IL-2 (R and D) plus Brefeldin A (BD Bioscience). After restimulation, lymphocytes were processed for flow cytometry as above.
  • IL-2 R and D
  • Brefeldin A BD Bioscience
  • NDV infection In order to characterize the anti-tumor immune response induced by Newcastle disease virus (NDV) infection, the expression of MHC I and MHC II molecules as well as ICAM-1 on the surface of in vitro infected cells was assessed. As shown in FIG. 1 , NDV infection in B16 melanoma cells induces upregulation of MHC class I and II molecules as well as adhesion molecule ICAM-1, all of which are thought to be important for recruitment of tumor-specific lymphocytes and activation of anti-tumor immune response.
  • NDV infection in B16 melanoma cells induces upregulation of MHC class I and II molecules as well as adhesion molecule ICAM-1, all of which are thought to be important for recruitment of tumor-specific lymphocytes and activation of anti-tumor immune response.
  • the anti-tumor immune response induced by NDV infection in vivo was assessed in a murine melanoma model and an established 2-flank model that allowed us to monitor responses both in the virus-injected tumors as well as distant tumors which do not receive the virus.
  • the virus-infected tumors show dramatic infiltration with immune cells such as NK cells, macrophages, and CD8 and CD4 cells, but not regulatory T cells. Since part of this immune response could be a response to virus, rather than tumor, the immune response with respect to contralateral tumors was assessed ( FIG. 3 ). Interestingly, these tumors demonstrated a similar degree of increased CD8 and CD4 effector, but not T reg infiltrate.
  • NDV monotherapy was effective in controlling the treated tumors ( FIG. 5A ), but only marginally slowed down the growth of the contralateral tumors ( FIG. 5B ). Mice that cleared the tumors, however, demonstrated some degree of protection against further tumor challenge ( FIG. 5D ), suggesting that NDV therapy can induce a lasting immunity.
  • NDV infected tumor cells both in vitro and in vivo had upregulated the expression of the inhibitory PD-L1 ligand on the surface of the cells. This effect was not just a result of a direct virus infection, but was also seen when non-infected cells were treated with UV-inactivated supernatants from the virus infected cells ( FIG. 9B ) and in contralateral, noninfected, tumors ( FIG. 9C ). This prompted testing combination therapy with NDV and anti-PD-1 antibody.
  • NDV therapy in combination with anti-PD-1 in the aggressive B16 melanoma model resulted in cures in the majority of animals, an effect that was associated with increased tumor infiltration with activated effector lymphocytes ( FIG. 10 ).
  • NDV-ICOSL NDV expressing murine ICOSL
  • FIG. 12 In vitro characterization of the virus revealed that it had similar replicative and lytic properties to the parental NDV strain ( FIG. 12 ).
  • NDV-ICOSL demonstrated significant advantage over the parental NDV virus when used in combination with CTLA-4 blockade, with long-term survival in the majority of treated animals ( FIG. 13 ). This effect was not limited to B16 melanoma and was demonstrated for CT26 colon carcinoma in the Balb/C mouse strain, suggesting that this therapeutic strategy could be translatable to different tumor types ( FIG. 14 ).
  • FIGS. 15 and 16 Analysis of B16 tumors from the treated animals demonstrated significant infiltration with different immune cell subtypes with upregulation of the activation markers. These lymphocytes were tumor-specific and demonstrated secretion of IFN gamma in response to stimulation with dendritic cells loaded with tumor lysates ( FIG. 17 ). Finally, animals that were cured of their B16 or CT26 tumors were re-challenged with tumor cells and demonstrated complete protection against tumor re-challenge ( FIG. 18 ).
  • a chimeric protein consisting of the extracellular domain of the ICOSL (amino acids 1-277) and the transmembrane and intracellular domains of the NDV F protein (amino acids 501-554) was generated ( FIG. 19A ).
  • Transfection of the resultant construct into B16-F10 cells resulted in increased expression of the chimeric ICOSL-F ligand on the surface of the transfected cells, when compared to the transfected native ICOSL, suggesting that the regulatory mechanisms governing the transport of NDV F protein to the surface can be utilized to increase the surface expression of immune stimulatory ligands.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Endocrinology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Described herein are chimeric Newcastle disease viruses engineered to express an agonist of a co-stimulatory signal of an immune cell and compositions comprising such viruses. Also described herein are chimeric Newcastle disease viruses engineered to express an antagonist of an inhibitory signal of an immune cell and compositions comprising such viruses. The chimeric Newcastle disease viruses and compositions are useful in the treatment of cancer. In addition, described herein are methods for treating cancer comprising administering Newcastle disease viruses in combination with an agonist of a co-stimulatory signal of an immune and/or an antagonist of an inhibitory signal of an immune cell.

Description

  • This application is a divisional of U.S. patent application Ser. No. 14/205,776, filed Mar. 12, 2014, which claims priority to U.S. Provisional Application No. 61/782,994, filed on Mar. 14, 2013, each of which is incorporated by reference herein in its entirety.
  • This invention was made government support under T32 CA009207 and HHSN26620070010C awarded by the National Institutes of Health. The government has certain rights in the invention.
  • 1. INTRODUCTION
  • Described herein are chimeric Newcastle disease viruses engineered to express an agonist of a co-stimulatory signal of an immune cell and compositions comprising such viruses. Also described herein are chimeric Newcastle disease viruses engineered to express an antagonist of an inhibitory signal of an immune cell and compositions comprising such viruses. The chimeric Newcastle disease viruses and compositions are useful in the treatment of cancer. In addition, described herein are methods for treating cancer comprising administering Newcastle disease viruses in combination with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • 2. BACKGROUND
  • Newcastle Disease Virus (NDV) is a member of the Avulavirus genus in the Paramyxoviridae family, which has been shown to infect a number of avian species (Alexander, D J (1988). Newcastle disease, Newcastle disease virus—an avian paramyxovirus. Kluwer Academic Publishers: Dordrecht, The Netherlands. pp 1-22). NDV possesses a single-stranded RNA genome in negative sense and does not undergo recombination with the host genome or with other viruses (Alexander, D J (1988). Newcastle disease, Newcastle disease virus—an avian paramyxovirus. Kluwer Academic Publishers: Dordrecht, The Netherlands. pp 1-22). The genomic RNA contains genes in the order of 3′-NP-P-M-F-HN-L-5′, described in further detail below. Two additional proteins, V and W, are produced by NDV from the P gene by alternative mRNAs that are generated by RNA editing. The genomic RNA also contains a leader sequence at the 3′ end.
  • The structural elements of the virion include the virus envelope which is a lipid bilayer derived from the cell plasma membrane. The glycoprotein, hemagglutinin-neuraminidase (HN) protrudes from the envelope allowing the virus to contain both hemagglutinin (e.g., receptor binding/fusogenic) and neuraminidase activities. The fusion glycoprotein (F), which also interacts with the viral membrane, is first produced as an inactive precursor, then cleaved post-translationally to produce two disulfide linked polypeptides. The active F protein is involved in penetration of NDV into host cells by facilitating fusion of the viral envelope with the host cell plasma membrane. The matrix protein (M), is involved with viral assembly, and interacts with both the viral membrane as well as the nucleocapsid proteins.
  • The main protein subunit of the nucleocapsid is the nucleocapsid protein (NP) which confers helical symmetry on the capsid. In association with the nucleocapsid are the P and L proteins. The phosphoprotein (P), which is subject to phosphorylation, is thought to play a regulatory role in transcription, and may also be involved in methylation, phosphorylation and polyadenylation. The L gene, which encodes an RNA-dependent RNA polymerase, is required for viral RNA synthesis together with the P protein. The L protein, which takes up nearly half of the coding capacity of the viral genome is the largest of the viral proteins, and plays an important role in both transcription and replication. The V protein has been shown to inhibit interferon-alpha and to contribute to the virulence of NDV (Huang et al. (2003). Newcastle disease virus V protein is associated with viral pathogenesis and functions as an Alpha Interferon Antagonist. Journal of Virology 77: 8676-8685).
  • Naturally-occurring NDV has been reported to be an effective oncolytic agent in a variety of animal tumor models (Sinkovics, J G, and Horvath, J C (2000). Newcastle disease virus (NDV): brief history of its oncolytic strains. J Clin Virol 16: 1-15). Naturally-occurring strains of NDV have been used in multiple clinical trials against advanced human cancers (Sinkovics, J G, and Horvath, J C (2000). Newcastle disease virus (NDV): brief history of its oncolytic strains. J Clin Virol 16: 1-15; Lorence et al. (2007). Phase 1 clinical experience using intravenous administration of PV701, an oncolytic Newcastle disease virus. Curr Cancer Drug Targets 7: 157-167; Hotte et al. (2007). An optimized clinical regimen for the oncolytic virus PV701. Clin Cancer Res 13: 977-985; Freeman et al. (2006). Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther 13: 221-228; Pecora et al. (2002). Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol 20: 2251-2266; Csatary et al. (2004). MTH-68/H oncolytic viral treatment in human high-grade gliomas. J Neurooncol 67: 83-93). However, the success of naturally-occurring strains of NDV in these clinical trials for advanced human cancers was only marginal (Hotte et al. (2007). An optimized clinical regimen for the oncolytic virus PV701. Clin Cancer Res 13: 977-985; Freeman et al. (2006). Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther 13: 221-228; Pecora et al. (2002). Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol 20: 2251-2266). As such, there remains a need for NDV-based therapies useful in the treatment of cancer, especially advanced cancer.
  • 3. SUMMARY
  • In one aspect, presented herein are chimeric Newcastle disease viruses (NDVs) engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell. In a specific embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, wherein the agonist is expressed. In a specific embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, wherein the antagonist is expressed.
  • In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell and a mutated F protein that causes the NDV to be highly fusogenic, wherein the agonist and the mutated F protein are expressed. In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell and a mutated F protein with a mutated cleavage site, wherein the agonist and the mutated F protein are expressed. In a specific embodiment, the chimeric NDVs expressing the mutated F protein have increased fusogenic activity relative to the corresponding virus expressing the counterpart F protein without the mutations to the cleavage site. In another specific embodiment, the modified F protein is incorporated into the virion.
  • In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell and a mutated F protein that causes the NDV to be highly fusogenic, wherein the antagonist and the mutated F protein are expressed. In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes antagonist of an inhibitory signal of an immune cell and a mutated F protein with a mutated cleavage site, wherein the antagonist and the mutated F protein are expressed. In a specific embodiment, the chimeric NDVs expressing the mutated F protein have increased fusogenic activity relative to the corresponding virus expressing the counterpart F protein without the mutations to the cleavage site. In another specific embodiment, the modified F protein is incorporated into the virion.
  • In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell and a cytokine (e.g., interleukin (IL)-2), wherein the agonist and the cytokine are expressed. In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein that causes the NDV to be highly fusogenic, wherein the agonist, the cytokine and the mutated F protein are expressed. In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein with a mutated cleavage site, wherein the agonist, the cytokine and the mutated F protein are expressed. In a specific embodiment, the chimeric NDVs expressing the mutated F protein with the mutated cleavage site are highly fusogenic. In another specific embodiment, the mutated F protein is incorporated into the virion.
  • In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell of an immune cell and a cytokine (e.g., IL-2), wherein the antagonist and the cytokine are expressed. In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein that causes the NDV to be highly fusogenic, wherein the antagonist, the cytokine and the mutated F protein are expressed. In another embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, a cytokine (e.g., IL-2) and a mutated F protein with a mutated cleavage site, wherein the antagonist, the cytokine and the mutated F protein are expressed. In a specific embodiment, the chimeric NDVs expressing the mutated F protein with the mutated cleavage site are highly fusogenic. In another specific embodiment, the mutated F protein is incorporated into the virion.
  • In a specific embodiment, the agonist of a co-stimulatory signal of an immune cell is an agonist of a co-stimulatory receptor expressed by an immune cell. Specific examples of co-stimulatory receptors include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell costimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-1BB (CD137), and CD40. In a specific embodiment, the agonist of a co-stimulatory receptor expressed by an immune cell is an antibody (or an antigen-binding fragment thereof) or ligand that specifically binds to the co-stimulatory receptor. In one embodiment, the antibody is a monoclonal antibody. In another embodiment, the antibody is an sc-Fv. In a specific embodiment the antibody is a bispecific antibody that binds to two receptors on an immune cell. In one embodiment, the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell. In specific embodiments, the antibody is a human or humanized antibody. In other embodiments, the ligand or an antibody is a chimeric protein comprising a NDV F protein or NDV HN protein. Methods for generating such chimeric proteins are known in the art. See, e.g., U.S. Patent Application Publication No. 2012-0122185, the disclosure of which is herein incorporated by reference in its entirety. Also see Park et al., PNAS 2006; 103:8203-8 and Murawski et al., J Virol 2010; 84:1110-23, the disclosures of which is herein incorporated by reference in their entireties.
  • In a specific embodiment, the antagonist of an inhibitory signal of an immune cell is an antagonist of an inhibitory receptor expressed by an immune cell. Specific examples of inhibitory receptors include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), and adenosine A2a receptor (A2aR). In a specific embodiment, the antagonist of an inhibitory receptor expressed by an immune cell is an antibody (or an antigen-binding fragment thereof) that specifically binds to the co-stimulatory receptor. In one embodiment, the antibody is a monoclonal antibody. In another embodiment, the antibody is an sc-Fv. In specific embodiments, the antibody is a human or humanized antibody. In another specific embodiment, the antagonist of an inhibitory receptor is a soluble receptor or antibody (or an antigen-binding fragment thereof) that specifically binds to a ligand of the inhibitory receptor.
  • In another aspect, presented herein are methods for propagating the NDVs described herein (e.g., chimeric NDVs described herein). The NDVs described herein (e.g., chimeric NDVs described herein) can be propagated in any cell, subject, tissue or organ susceptible to a NDV infection. In one embodiment, the NDVs described herein (e.g., chimeric NDVs described herein) may be propagated in a cell line. In another embodiment, the NDVs described herein (e.g., chimeric NDVs described herein) may be propagated in cancer cells. In another embodiment, the NDVs described herein (e.g., chimeric NDVs described herein) may be propagated in an embryonated egg. In certain embodiments, presented herein are isolated cells, tissues or organs infected with an NDV described herein (e.g., a chimeric NDV described herein). See, e.g., Section 5.4, infra, for examples of cells, animals and eggs to infect with an NDV described herein (e.g., a chimeric NDV described herein). In specific embodiments, presented herein are isolated cancer cells infected with an NDV described herein (e.g., a chimeric NDV described herein). In certain embodiments, presented herein are cell lines infected with an NDV described herein (e.g., a chimeric NDV described herein). In other embodiments, presented herein are embryonated eggs infected with an NDV described herein (e.g., a chimeric NDV described herein).
  • In another aspect, presented herein are compositions comprising an NDV described herein (e.g., a chimeric NDV described herein). In a specific embodiment, presented herein are pharmaceutical compositions comprising an NDV described herein (e.g., a chimeric NDV described herein) and a pharmaceutically acceptable carrier. In another embodiment, presented herein are pharmaceutical compositions comprising cancer cells infected with an NDV described herein (e.g., a chimeric NDV described herein), and a pharmaceutically acceptable carrier. In specific embodiments, the cancer cells have been treated with gamma radiation prior to incorporation into the pharmaceutical composition. In specific embodiments, the cancer cells have been treated with gamma radiation before infection with the NDV (e.g., chimeric NDV). In other specific embodiments, the cancer cells have been treated with gamma radiation after infection with the NDV (e.g., chimeric NDV). In another embodiment, presented herein are pharmaceutical compositions comprising protein concentrate from lysed NDV-infected cancer cells (e.g., chimeric-NDV infected cancer cells), and a pharmaceutically acceptable carrier.
  • In another aspect, presented herein are methods for producing pharmaceutical compositions comprising an NDV described herein (e.g., a chimeric NDV described herein). In one embodiment, a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in a cell line that is susceptible to an NDV infection; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition. In another embodiment, a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in an embryonated egg; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition.
  • In another aspect, presented herein are methods for treating cancer utilizing a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition comprising such a chimeric NDV. In a specific embodiment, a method for treating cancer comprises infecting a cancer cell in a subject with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof. In another embodiment, a method for treating cancer comprises administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof. In specific embodiments, an effective amount of a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition comprising an effective amount of a chimeric NDV described herein is administered to a subject to treat cancer. In specific embodiments, the chimeric NDV comprises a genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell). In certain embodiments, the genome of the NDV also comprises a mutated F protein. In certain embodiments, two or more chimeric NDVs are administered to a subject to treat cancer.
  • In another embodiment, a method for treating cancer comprises administering to a subject in need thereof cancer cells infected with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or composition thereof. In specific embodiments, the cancer cells have been treated with gamma radiation prior to administration to the subject or incorporation into the composition. In another embodiment, a method for treating cancer comprises administering to a subject in need thereof a protein concentrate or plasma membrane fragments from cancer cells infected with a chimeric NDV (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof. In specific embodiments, the chimeric NDV comprises a genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell). In certain embodiments, the genome of the NDV also comprises a mutated F protein.
  • In another aspect, presented herein are methods for treating cancer utilizing an NDV described herein (e.g., a chimeric NDV such as described in Section 5.2, infra) or a composition comprising such the NDV in combination with one or more other therapies. In one embodiment, presented herein are methods for treating cancer comprising administering to a subject an NDV described herein (e.g., a chimeric NDV, such as described in Section 5.2.1, infra) and one or more other therapies. In another embodiment, presented herein are methods for treating cancer comprising administering to a subject an effective amount of an NDV described herein or a composition comprising an effective amount of an NDV described herein, and one or more other therapies. The NDV and one or more other therapies can be administered concurrently or sequentially to the subject. In certain embodiments, the NDV and one or more other therapies are administered in the same composition. In other embodiments, the NDV and one or more other therapies are administered in different compositions. The NDV and one or more other therapies can be administered by the same or different routes of administration to the subject.
  • Any NDV type or strain may be used in a combination therapy disclosed herein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses. In a specific embodiment, the NDV used in a combination with one or more other therapies is a naturally-occurring strain. In another embodiment, the NDV used in combination with one or more other therapies is a chimeric NDV. In a specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising a cytokine (e.g., IL-2, IL-7, IL-15, IL-17, or IL-21). In specific embodiments, the cytokine is expressed by cells infected with the chimeric NDV. In a specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising a tumor antigen. In specific embodiments, the tumor antigen is expressed by cells infected with the chimeric NDV. In a specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising a pro-apoptotic molecule or an anti-apoptotic molecule. In specific embodiments, the pro-apoptotic molecule or anti-apoptotic molecule is expressed by cells infected with the chimeric NDV.
  • In another specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell). In specific embodiments, the agonist and/or antagonist are expressed by cells infected with the chimeric NDV. In certain embodiments, the genome of the NDV also comprises a mutated F protein. In certain embodiments, the one or more therapies used in combination with an NDV described herein is one or more other therapies described in Section 5.6.4 infra. In particular embodiments, the one or more therapies used in combination with an NDV described herein is an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell (see, e.g., Section 5.6.4.4.1, infra). See, e.g., Section 5.2.1, infra, for examples of agonists of a co-stimulatory signal of an immune cell and antagonists of an inhibitory signal of an immune cell. In a specific embodiment, the antagonist of an inhibitory signal of an immune cell is the anti-CTLA-4 antibody described in Section 6, infra. In another specific embodiment, the agonist of a co-stimulatory signal of an immune cell is the ICOS ligand described in Section 6, infra
  • 3.1 Terminology
  • As used herein, the term “about” or “approximately” when used in conjunction with a number refers to any number within 1, 5 or 10% of the referenced number.
  • As used herein, the term “agonist(s)” refers to a molecule(s) that binds to another molecule and induces a biological reaction. In a specific embodiment, an agonist is a molecule that binds to a receptor on a cell and triggers one or more signal transduction pathways. For example, an agonist includes an antibody or ligand that binds to a receptor on a cell and induces one or more signal transduction pathways. In certain embodiments, the antibody or ligand binds to a receptor on a cell and induces one or more signal transduction pathways. In other embodiments, the agonist facilitates the interaction of the native ligand with the native receptor.
  • As used herein, the term “antagonist(s)” refers to a molecule(s) that inhibits the action of another molecule without provoking a biological response itself. In a specific embodiment, an antagonist is a molecule that binds to a receptor on a cell and blocks or dampens the biological activity of an agonist. For example, an antagonist includes an antibody or ligand that binds to a receptor on a cell and blocks or dampens binding of the native ligand to the cell without inducing one or more signal transduction pathways. Another example of an antagonist includes an antibody or soluble receptor that competes with the native receptor on cells for binding to the native ligand, and thus, blocks or dampens one or more signal transduction pathways induced when the native receptor binds to the native ligand.
  • As used herein, the terms “antibody” and “antibodies” refer to molecules that contain an antigen binding site, e.g., immunoglobulins. Antibodies include, but are not limited to, monoclonal antibodies, bispecific antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, polyclonal antibodies, single domain antibodies, camelized antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked bispecific Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id and anti-anti-Id antibodies to antibodies), and epitope-binding fragments of any of the above. In particular, antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass. In a specific embodiment, an antibody is a human or humanized antibody. In another specific embodiment, an antibody is a monoclonal antibody or scFv. In certain embodiments, an antibody is a human or humanized monoclonal antibody or scFv. In other specific embodiments, the antibody is a bispecific antibody. In certain embodiments, the bispecific antibody specifically binds to a co-stimulatory receptor of an immune cell or an inhibitory receptor of an immune, and a receptor on a cancer cell. In some embodiments, the bispecific antibody specifically binds to two receptors immune cells, e.g., two co-stimulatory receptors on immune cells, two inhibitory receptors on immune cells, or one co-stimulatory receptor on immune cells and one inhibitory receptor on immune cells.
  • As used herein, the term “derivative” in the context of proteins or polypeptides refers to: (a) a polypeptide that is at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% or is 40% to 65%, 50% to 90%, 65% to 90%, 70% to 90%, 75% to 95%, 80% to 95%, or 85% to 99% identical to a native polypeptide; (b) a polypeptide encoded by a nucleic acid sequence that is at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% or is 40% to 65%, 50% to 90%, 65% to 90%, 70% to 90%, 75% to 95%, 80% to 95%, or 85% to 99% identical a nucleic acid sequence encoding a native polypeptide; (c) a polypeptide that contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more, or 2 to 5, 2 to 10, 5 to 10, 5 to 15, 5 to 20, 10 to 15, or 15 to 20 amino acid mutations (i.e., additions, deletions and/or substitutions) relative to a native polypeptide; (d) a polypeptide encoded by nucleic acid sequence that can hybridize under high, moderate or typical stringency hybridization conditions to a nucleic acid sequence encoding a native polypeptide; (e) a polypeptide encoded by a nucleic acid sequence that can hybridize under high, moderate or typical stringency hybridization conditions to a nucleic acid sequence encoding a fragment of a native polypeptide of at least 10 contiguous amino acids, at least 12 contiguous amino acids, at least 15 contiguous amino acids, at least 20 contiguous amino acids, at least 30 contiguous amino acids, at least 40 contiguous amino acids, at least 50 contiguous amino acids, at least 75 contiguous amino acids, at least 100 contiguous amino acids, at least 125 contiguous amino acids, at least 150 contiguous amino acids, or 10 to 20, 20 to 50, 25 to 75, 25 to 100, 25 to 150, 50 to 75, 50 to 100, 75 to 100, 50 to 150, 75 to 150, 100 to 150, or 100 to 200 contiguous amino acids; or (f) a fragment of a native polypeptide. Derivatives also include a polypeptide that comprises the amino acid sequence of a naturally occurring mature form of a mammalian polypeptide and a heterologous signal peptide amino acid sequence. In addition, derivatives include polypeptides that have been chemically modified by, e.g., glycosylation, acetylation, pegylation, phosphorylation, amidation, derivitization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein moiety, etc. Further, derivatives include polypeptides comprising one or more non-classical amino acids. In one embodiment, a derivative is isolated. In specific embodiments, a derivative retains one or more functions of the native polypeptide from which it was derived.
  • Percent identity can be determined using any method known to one of skill in the art. In a specific embodiment, the percent identity is determined using the “Best Fit” or “Gap” program of the Sequence Analysis Software Package (Version 10; Genetics Computer Group, Inc., University of Wisconsin Biotechnology Center, Madison, Wis.). Information regarding hybridization conditions (e.g., high, moderate, and typical stringency conditions) have been described, see, e.g., U.S. Patent Application Publication No. US 2005/0048549 (e.g., paragraphs 72-73).
  • As used herein, the term “fragment” is the context of a fragment of a proteinaceous agent (e.g., a protein) refers to a fragment that is 8 or more contiguous amino acids, 10 or more contiguous amino acids, 15 or more contiguous amino acids, 20 or more contiguous amino acids, 25 or more contiguous amino acids, 50 or more contiguous amino acids, 75 or more contiguous amino acids, 100 or more contiguous amino acids, 150 or more contiguous amino acids, 200 or more contiguous amino acids, or in the range of between 10 to 300 contiguous amino acids, 10 to 200 contiguous amino acids, 10 to 250 contiguous amino acids, 10 to 150 contiguous amino acids, 10 to 100 contiguous amino acids, 10 to 50 contiguous amino acids, 50 to 100 contiguous amino acids, 50 to 150 contiguous amino acids, 50 to 200 contiguous amino acids, 50 to 250 contiguous amino acids, 50 to 300 contiguous amino acids, 25 to 50 contiguous amino acids, 25 to 75 contiguous amino acids, 25 to 100 contiguous amino acids, or 75 to 100 contiguous amino acids of a proteinaceous agent. In a specific embodiment, a fragment of a proteinaceous agent retains one or more functions of the proteinaceous agent—in other words, it is a functional fragment. For example, a fragment of a proteinaceous agent retains the ability to interact with another protein and/or to induce, enhance or activate one or more signal transduction pathways.
  • As used herein, the term “functional fragment,” in the context of a proteinaceous agent, refers to a portion of a proteinaceous agent that retains one or more activities or functions of the proteinaceous agent. For example, a functional fragment of an inhibitory receptor may retain the ability to bind one or more of its ligands. A functional fragment of a ligand of a co-stimulatory receptor may retain the ability to bind to the receptor and/or induce, enhance or activate one or more signal transduction pathways mediated by the ligand binding to its co-stimulatory receptor.
  • As used herein, the term “heterologous” refers an entity not found in nature to be associated with (e.g., encoded by and/or expressed by the genome of) a naturally occurring NDV.
  • As used herein, the term “elderly human” refers to a human 65 years or older.
  • As used herein, the term “human adult” refers to a human that is 18 years or older.
  • As used herein, the term “human child” refers to a human that is 1 year to 18 years old.
  • As used herein, the term “human toddler” refers to a human that is 1 year to 3 years old.
  • As used herein, the term “human infant” refers to a newborn to 1 year old year human.
  • In certain embodiments, the terms “highly fusogenic” and “increased fusogenic activity”, and the like, as used herein, refers to an increase in the ability of the NDV to form syncytia involving a large number of cells. In a specific embodiment, cells infected with an NDV described herein that is engineered to express a mutated F protein have an increased ability to form syncytia relative to cells infected with the parental virus from which the virus is derived, which parental virus has an unmutated F protein. In another specific embodiment, about 10% to about 25%, about 25% to about 50%, about 25% to about 75%, about 50% to about 75%, about 50% to about 95%, or about 75% to about 99% or about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% more cells infected with an NDV described herein that is engineered to express a mutated F protein form syncytia relative to the number of cells forming syncytia that are infected with the parental virus from the chimeric virus is derived which has an unmutated F protein. In certain embodiments, the syncytia are quantitated microscopically by counting the number of nuclei per syncytium after a certain period of time (e.g., about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 36 hours, or about 36 hours to about 48 hours).
  • As used herein, the term “interferon antagonist” refers to an agent that reduces or inhibits the cellular interferon immune response. In one embodiment, an interferon antagonist is a proteinaceous agent that reduces or inhibits the cellular interferon immune response. In a specific embodiment, an interferon antagonist is a viral protein or polypeptide that reduces or inhibits the cellular interferon response.
  • In a specific embodiment, an interferon antagonist is an agent that reduces or inhibits interferon expression and/or activity. In one embodiment, the interferon antagonist reduces or inhibits the expression and/or activity of type I IFN. In another embodiment, the interferon antagonist reduces or inhibits the expression and/or activity of type II IFN. In another embodiment, the interferon antagonist reduces or inhibits the expression and/or activity of type III IFN. In a specific embodiment, the interferon antagonist reduces or inhibits the expression and/or activity of either IFN-α, IFN-β or both. In another specific embodiment, the interferon antagonist reduces or inhibits the expression and/or activity of IFN-γ. In another embodiment, the interferon antagonist reduces or inhibits the expression and/or activity of one, two or all of IFN-α, IFN-β, and IFN-γ.
  • In certain embodiments, the expression and/or activity of IFN-α, IFN-β and/or IFN-γ in an embryonated egg or cell is reduced approximately 1 to approximately 100 fold, approximately 5 to approximately 80 fold, approximately 20 to approximately 80 fold, approximately 1 to approximately 10 fold, approximately 1 to approximately 5 fold, approximately 40 to approximately 80 fold, or 1, 2, 3, 4, 5, 7, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 fold by an interferon antagonist relative to the expression and/or activity of IFN-α, IFN-β, and/or IFN-γ in a control embryonated egg or a cell not expressing or not contacted with such an interferon antagonist as measured by the techniques described herein or known to one skilled in the art. In other embodiments, the expression and/or activity of IFN-α, IFN-β and/or IFN-γ in an embryonated egg or cell is reduced by at least 20% to 25%, at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, at least 80% to 85%, at least 85% to 90%, at least 90% to 95%, at least 95% to 99% or by 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% by an interferon antagonist relative to the expression and/or activity of IFN-α, IFN-β, and/or IFN-γ in a control embryonated egg or a cell not expressing or not contacted with such an interferon antagonist as measured by the techniques described herein or known to one skilled in the art.
  • As used herein, the phrases “IFN deficient systems” or “IFN-deficient substrates” refer to systems, e.g., cells, cell lines and animals, such as mice, chickens, turkeys, rabbits, rats, horses etc., which do not produce one, two or more types of IFN, or do not produce any type of IFN, or produce low levels of one, two or more types of IFN, or produce low levels of any IFN (i.e., a reduction in any IFN expression of 5-10%, 10-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-90% or more when compared to IFN-competent systems under the same conditions), do not respond or respond less efficiently to one, two or more types of IFN, or do not respond to any type of IFN, have a delayed response to one, two or more types of IFN, and/or are deficient in the activity of antiviral genes induced by one, two or more types of IFN, or induced by any type of IFN.
  • As used herein, the terms “immunospecifically binds,” “immunospecifically recognizes,” “specifically binds,” and “specifically recognizes” are analogous terms in the context of antibodies and refer to molecules that specifically bind to an antigen (e.g., epitope or immune complex) as understood by one skilled in the art. A molecule that specifically binds to an antigen may bind to other peptides or polypeptides with lower affinity as determined by, e.g., immunoassays (e.g., ELISA), surface plasmon resonance (e.g., BIAcore®), a KinEx assay (using, e.g., a KinExA 3000 instrument (Sapidyne Instruments, Boise, Id.)), or other assays known in the art. In a specific embodiment, molecules that specifically bind to an antigen bind to the antigen with a dissociation constant (i.e., Ka) that is at least 2 logs, 2.5 logs, 3 logs, 3.5 logs, 4 logs or greater than the Ka when the molecules bind to another antigen. In a another specific embodiment, molecules that specifically bind to an antigen do not cross react with other proteins.
  • As used herein, the term “monoclonal antibody” is a term of the art and generally refers to an antibody obtained from a population of homogenous or substantially homogeneous antibodies, and each monoclonal antibody will typically recognize a single epitope (e.g., single conformation epitope) on the antigen.
  • As used herein, the phrase “multiplicity of infection” or “MOI” is the average number of virus per infected cell. The MOI is determined by dividing the number of virus added (ml added x Pfu) by the number of cells added (ml added x cells/ml).
  • As used herein, the term “native ligand” refers to any naturally occurring ligand that binds to a naturally occurring receptor. In a specific embodiment, the ligand is a mammalian ligand. In another specific embodiment, the ligand is a human ligand.
  • As used herein, the term “native polypeptide(s)” in the context of proteins or polypeptides refers to any naturally occurring amino acid sequence, including immature or precursor and mature forms of a protein. In a specific embodiment, the native polypeptide is a human protein or polypeptide.
  • As used herein, the term “native receptor” refers to any naturally occurring receptor that binds to a naturally occurring ligand. In a specific embodiment, the receptor is a mammalian receptor. In another specific embodiment, the receptor is a human receptor.
  • As used herein, the terms “subject” or “patient” are used interchangeably. As used herein, the terms “subject” and “subjects” refers to an animal. In some embodiments, the subject is a mammal including a non-primate (e.g., a camel, donkey, zebra, cow, horse, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, chimpanzee, and a human). In some embodiments, the subject is a non-human mammal. In certain embodiments, the subject is a pet (e.g., dog or cat) or farm animal (e.g., a horse, pig or cow). In other embodiments the subject is a human. In certain embodiments, the mammal (e.g., human) is 0 to 6 months old, 6 to 12 months old, 1 to 5 years old, 5 to 10 years old, 10 to 15 years old, 15 to 20 years old, 20 to 25 years old, 25 to 30 years old, 30 to 35 years old, 35 to 40 years old, 40 to 45 years old, 45 to 50 years old, 50 to 55 years old, 55 to 60 years old, 60 to 65 years old, 65 to 70 years old, 70 to 75 years old, 75 to 80 years old, 80 to 85 years old, 85 to 90 years old, 90 to 95 years old or 95 to 100 years old. In specific embodiments, the subject is an animal that is not avian.
  • As used herein, the terms “treat” and “treating” in the context of the administration of a therapy refers to a treatment/therapy from which a subject receives a beneficial effect, such as the reduction, decrease, attenuation, diminishment, stabilization, remission, suppression, inhibition or arrest of the development or progression of cancer, or a symptom thereof. In certain embodiments, the treatment/therapy that a subject receives results in at least one or more of the following effects: (i) the reduction or amelioration of the severity of cancer and/or a symptom associated therewith; (ii) the reduction in the duration of a symptom associated with cancer; (iii) the prevention in the recurrence of a symptom associated with cancer; (iv) the regression of cancer and/or a symptom associated therewith; (v) the reduction in hospitalization of a subject; (vi) the reduction in hospitalization length; (vii) the increase in the survival of a subject; (viii) the inhibition of the progression of cancer and/or a symptom associated therewith; (ix) the enhancement or improvement the therapeutic effect of another therapy; (x) a reduction or elimination in the cancer cell population; (xi) a reduction in the growth of a tumor or neoplasm; (xii) a decrease in tumor size; (xiii) a reduction in the formation of a tumor; (xiv) eradication, removal, or control of primary, regional and/or metastatic cancer; (xv) a decrease in the number or size of metastases; (xvi) a reduction in mortality; (xvii) an increase in cancer-free survival rate of patients; (xviii) an increase in relapse-free survival; (xix) an increase in the number of patients in remission; (xx) a decrease in hospitalization rate; (xxi) the size of the tumor is maintained and does not increase in size or increases the size of the tumor by less 5% or 10% after administration of a therapy as measured by conventional methods available to one of skill in the art, such as MRI, X-ray, and CAT Scan; (xxii) the prevention of the development or onset of cancer and/or a symptom associated therewith; (xxiii) an increase in the length of remission in patients; (xxiv) the reduction in the number of symptoms associated with cancer; (xxv) an increase in symptom-free survival of cancer patients; and/or (xxvi) limitation of or reduction in metastasis. In some embodiments, the treatment/therapy that a subject receives does not cure cancer, but prevents the progression or worsening of the disease. In certain embodiments, the treatment/therapy that a subject receives does not prevent the onset/development of cancer, but may prevent the onset of cancer symptoms.
  • As used herein, the term “in combination” in the context of the administration of (a) therapy(ies) to a subject, refers to the use of more than one therapy. The use of the term “in combination” does not restrict the order in which therapies are administered to a subject. A first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject.
  • As used herein, the terms “therapies” and “therapy” can refer to any protocol(s), method(s), and/or agent(s) that can be used in the treatment of cancer. In certain embodiments, the terms “therapies” and “therapy” refer to biological therapy, supportive therapy, hormonal therapy, chemotherapy, immunotherapy and/or other therapies useful in the treatment of cancer. In a specific embodiment, a therapy includes adjuvant therapy. For example, using a therapy in conjunction with a drug therapy, biological therapy, surgery, and/or supportive therapy. In certain embodiments, the term “therapy” refers to a chimeric NDV described herein. In other embodiments, the term “therapy” refers to an agent that is not a chimeric NDV.
  • 4. BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. NDV infection upregulates the expression of MHC I, MHC II, and ICAM-1 on the surface of in vitro infected B16-F10 cells (24 hours post-infection).
  • FIGS. 2A-2E. Intratumoral NDV treatment leads to infiltration with macrophages, NK cells, CD8 and CD4 effector cells and decreases the frequency of Tregs. A) Overall study scheme. B) Total CD45+ infiltrates. C) Total immune cell infiltrates. D) Representative flow cytometry dot plots of relative CD4 FoxP3+ and FoxP3-subsets. E) Teff/Treg and CD8/Treg ratios.
  • FIGS. 3A-3C. Therapy with NDV exhibits favorable effects on tumor microenvironment of distant tumors. A) Representative flow cytometry dot plots of relative CD4 FoxP3+ and FoxP3-subsets. B) Absolute numbers of CD4 effector, Treg, and CD8 cells per gram of tumor. C) Teff/Treg and CD8/Treg ratios.
  • FIGS. 4A-4C. Lymphocytes infiltrating distant tumors upregulate activation, lytic, and proliferation markers. Representative expression plots on CD4 effector cells (top) and the corresponding percentages in the CD4 effector, CD8, Tregs (bottom) are shown for A) CD44, B) Granzyme B, and C) Ki-67.
  • FIGS. 5A-5D. NDV Monotherapy delays the growth of distant tumors and provides some protection against tumor rechallenge. Bilateral flank tumors were established as described in FIG. 2A and the animals were treated and followed for survival. A) Growth of right flank (treated) tumors. B) Growth of left flank (non-treated) tumors. C) Overall survival. Numbers in boxes indicate percent of animals free of tumors. D) Survival in animals cured of B16-F10 melanoma by NDV re-challenged on day 75 with B16-F10 melanoma cells. Representative results of two different experiments with 10 mice per group.
  • FIGS. 6A-6B. Tumor-infiltrating lymphocytes from both treated and non-treated tumors upregulate CTLA-4 in response to NDV therapy. A) Representative dot plots of CTLA-4 expression in CD8, CD4 effector, and Tregs in right (treated) tumors. B) Representative dot plots of CTLA-4 expression in CD8, CD4 effector, and Tregs in left (non-treated) tumors.
  • FIG. 7A-7C. Combination therapy with NDV and CTLA-4 blockade enhances anti-tumor effect in the injected and distant tumors. Bilateral B16 flank tumors were established and the animals were treated as described in FIG. 2A with or without anti-CTLA-4 antibody 9H10. A) Growth of treated tumors. B) Growth of distant tumors. Numbers in boxes represent percentage of mice free of tumors. C) Long-term survival. Representative results of 2 different experiments with 10 mice per group.
  • FIG. 8. Combination therapy with NDV and anti-CTLA-4 is effective systemically against non-virus-permissive prostate TRAMP tumors. Right (day 12) and left (day 3) flank TRAMP tumors were established and the animals were treated with NDV as described in FIG. 2A with or without systemic anti-CTLA-4 antibody. Growth of left flank (non-injected) tumors is shown. Numbers in boxes indicate percent of animals free of tumors.
  • FIG. 9A-9C. NDV infection upregulates expression of PD-L1 in B16-F10 tumors. A) Surface PD-L1 expression on B16-F10 cells infected with NDV for 24 hours. B) Surface PD-L1 expression on B16-F10 cells treated with UV-inactivated supernatant from infected B16-F10 cells. C) Upregulation of PD-L1 on the surface of tumor cells isolated from injected and distant tumors from the animals treated as in FIG. 2A (2 left panels-representative flow cytometry plots, right panel-calculated averages of 5 mice per group).
  • FIGS. 10A-10F. Combination therapy with NDV and anti-PD-1 is effective systemically against B16 melanoma and results in increased T cell infiltration with upregulation of activation markers. A) Overall survival. Animals were treated as described in FIG. 2A with or without anti-PD-1 antibody. B) Absolute numbers of CD45, CD3, CD8, and CD4 effector cells in tumors. C) Relative percentage of regulatory T cells in tumor-infiltrating lymphocytes. D-E) Tumor-infiltrating lymphocytes from distant tumors were isolated and stained for expression of ICOS (D) and Granzyme B (E). F) Tumor infiltrating lymphocytes were re-stimulated with dendritic cells loaded with tumor lysates and assessed for expression of IFN gamma by intracellular cytokine staining.
  • FIG. 11. Combination therapy with NDV and CTLA-4 induces upregulation of ICOS and CD4 effector cells in distant tumors and tumor-draining lymph nodes (TDLN).
  • FIGS. 12A-12D. Generation and in vitro evaluation of NDV-ICOSL virus. A) Viral genomic construct scheme. B) Expression of ICOSL on the surface of B16-F10 cells infected for 24 hours (representative histogram, left and average of 3 samples per group, right). C) Cytolytic activity of NDV in the infected B16-F10 cells determined by LDH assay. D) Replication of recombinant NDV in the B16-F10 cells.
  • FIGS. 13A-13C. Combination therapy with NDV-mICOSL and anti-CTLA-4 protects mice from contralateral tumor challenge and results in long-term animal survival. Animals were challenged with a larger tumor dose and treated with NDV as described in FIG. 2A with or without systemic anti-CTLA-4 antibody. Growth of left flank (non-injected) tumors is shown. B) Long-term survival. Numbers in boxes indicate percent of animals protected from tumors. Pooled data of 3 different experiments of 5-10 mice per group. C) Mice treated with combination therapy develop vitiligo at the former tumor sites, but not systemically.
  • FIG. 14A-14B. Combination therapy with NDV-mICOSL and anti-CTLA-4 protects mice from contralateral tumor challenge and results in long-term animal survival in the CT26 colon carcinoma model. Animals were challenged with a larger tumor dose and treated with NDV as described in FIG. 2A with or without systemic anti-CTLA-4 antibody. Growth of left flank (non-injected) tumors is shown. Numbers in boxes indicate percent of animals protected from tumors. B) Long-term survival. Representative experiment with 5-10 mice per group (A) and pooled data of 2 different experiments of 5-10 mice per group (B).
  • FIGS. 15A-15C. NDV treatment leads to distant B16 tumor infiltration with macrophages, NK cells, CD8 and CD4 effector cells and decreases the frequency of Tregs. A) Total CD45+, CD3+, CD8+, CD4+ FoxP3− (Teff), and CD4+ FoxP3+ (Treg) infiltrates. B) Teff/Treg and CD8/Treg ratios. C) Total macrophage, NK, and NKT cell infiltrates.
  • FIG. 16A-16B. Lymphocytes infiltrating distant B16 tumors upregulate activation, lytic, and proliferation markers. A) Representative Ki-67, Granzyme B (GrB) and ICOS expression plots. B) the corresponding percentages in the CD4 effector and CD8 cells.
  • FIG. 17. Tumor infiltrating lymphocytes from treated animals secrete IFN-gamma in response to stimulation with DC's loaded with B16-F10 lysates. Representative dot plots are shown.
  • FIGS. 18A-18B. Animals cured by combination therapy are protected from further tumor challenge. A) B16-F10 melanoma, day 120 re-challenge with 1×105 cells. B) CT26 colon carcinoma, day 90 re-challenge with 1×106 cells. Representative results of two different experiments with 10 mice per group.
  • FIG. 19A-19B. Recombinant ICOSL-F chimeric protein is efficiently expressed on surface. A) Schematic diagram of the chimeric protein. B) Expression of the chimeric ICOSL-Ftm fusion protein on the surface of transfected cells.
  • 5. DETAILED DESCRIPTION
  • In one aspect, presented herein are chimeric Newcastle disease viruses (NDVs) engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell. In a specific embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an agonist of a co-stimulatory signal of an immune cell, wherein the agonist is expressed. In a specific embodiment, presented herein are chimeric NDVs, comprising a packaged genome which encodes an antagonist of an inhibitory signal of an immune cell, wherein the antagonist is expressed.
  • In another aspect, presented herein are methods for propagating the NDVs described herein (e.g., chimeric NDVs described herein). The NDVs described herein (e.g., chimeric NDVs described herein) can be propagated in any cell, subject, tissue, organ or animal susceptible to a NDV infection.
  • In another aspect, presented herein are compositions comprising an NDV described herein (e.g., a chimeric NDV described herein). In a specific embodiment, presented herein are pharmaceutical compositions comprising an NDV described herein (e.g., a chimeric NDV described herein) and a pharmaceutically acceptable carrier. In another embodiment, presented herein are pharmaceutical compositions comprising cancer cells infected with an NDV described herein (e.g., a chimeric NDV described herein), and a pharmaceutically acceptable carrier. In another embodiment, presented herein are pharmaceutical compositions comprising protein concentrate from lysed NDV-infected cancer cells (e.g., chimeric-NDV infected cancer cells), and a pharmaceutically acceptable carrier.
  • In another aspect, presented herein are methods for producing pharmaceutical compositions comprising an NDV described herein (e.g., a chimeric NDV described herein). In one embodiment, a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in a cell line that is susceptible to an NDV infection; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition. In another embodiment, a method for producing a pharmaceutical composition comprises: (a) propagating an NDV described herein (e.g., a chimeric NDV described herein) in an embryonated egg; and (b) collecting the progeny virus, wherein the virus is grown to sufficient quantities and under sufficient conditions that the virus is free from contamination, such that the progeny virus is suitable for formulation into a pharmaceutical composition.
  • In another aspect, presented herein are methods for treating cancer utilizing a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition comprising such a chimeric NDV. In a specific embodiment, a method for treating cancer comprises infecting a cancer cell in a subject with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof. In another embodiment, a method for treating cancer comprises administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof. In specific embodiments, an effective amount of a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or a composition comprising an effective amount of a chimeric NDV described herein is administered to a subject to treat cancer. In specific embodiments, the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell), wherein the agonist and/or antagonist are expressed by the NDV. In certain embodiments, the genome of the NDV also comprises a mutated F protein. In certain embodiments, two or more chimeric NDVs are administered to a subject to treat cancer.
  • In another embodiment, a method for treating cancer comprises administering to a subject in need thereof cancer cells infected with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra) or composition thereof. In specific embodiments, the cancer cells have been treated with gamma radiation prior to administration to the subject or incorporation into the composition. In another embodiment, a method for treating cancer comprises administering to a subject in need thereof a protein concentrate or plasma membrane fragments from cancer cells infected with a chimeric NDV (e.g., a chimeric NDV described in Section 5.2, infra) or a composition thereof. In specific embodiments, the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell), wherein the agonist and/or antagonist are expressed by the NDV. In certain embodiments, the genome of the NDV also comprises a mutated F protein, which is expressed by the NDV.
  • In another aspect, presented herein are methods for treating cancer utilizing an NDV described herein (e.g., a chimeric NDV such as described in Section 5.2, infra) or a composition comprising such the NDV in combination with one or more other therapies. In one embodiment, presented herein are methods for treating cancer comprising administering to a subject an NDV described herein (e.g., a chimeric NDV, such as described in Section 5.2, infra) and one or more other therapies. In another embodiment, presented herein are methods for treating cancer comprising administering to a subject an effective amount of an NDV described herein or a composition comprising an effective amount of an NDV described herein, and one or more other therapies. The NDV and one or more other therapies can be administered concurrently or sequentially to the subject. In certain embodiments, the NDV and one or more other therapies are administered in the same composition. In other embodiments, the NDV and one or more other therapies are administered in different compositions. The NDV and one or more other therapies can be administered by the same or different routes of administration to the subject.
  • Any NDV type or strain may be used in a combination therapy disclosed herein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses. In a specific embodiment, the NDV used in a combination with one or more other therapies is a naturally-occurring strain. In another embodiment, the NDV used in combination with one or more other therapies is a chimeric NDV. In a specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising a cytokine (e.g., IL-2, IL-7, IL-15, IL-17 or IL-21). In specific embodiments, the chimeric NDV comprises a packaged genome, the genome comprising a tumor antigen. In specific embodiments, the tumor antigen is expressed by cells infected with the chimeric NDV. In another specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising a pro-apoptotic molecule (e.g., Bax, Bak, Bad, BID, Bc1-xS, Bim, Noxa, Puma, AIF, FasL, and TRAIL) or an anti-apoptotic molecule (e.g., Bc1-2, Bc1-xL, Mc1-1, and XIAP). In specific embodiments, the pro-apoptotic molecule or anti-apoptotic molecule is expressed by cells infected with the chimeric NDV. In another specific embodiment, the chimeric NDV comprises a packaged genome, the genome comprising an agonist of a co-stimulatory signal of an immune cell (e.g., an agonist of a co-stimulatory receptor of an immune cell) and/or an antagonist of an inhibitory signal of an immune cell (e.g., an antagonist of an inhibitory receptor of an immune cell). In specific embodiments, the agonist and/or antagonist are expressed by cells infected with the chimeric NDV. In certain embodiments, the genome of the NDV also comprises a mutated F protein, a tumor antigen, a heterologous interferon antagonist, a pro-apoptotic molecule and/or an anti-apoptotic molecule. In certain embodiments, the one or more therapies used in combination with an NDV described herein is one or more other therapies described in Section 5.6.4, infra. In particular embodiments, the one or more therapies used in combination with an NDV described herein are an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell. See, e.g., Section 5.2.1, infra, for examples of agonists of a co-stimulatory signal of an immune cell and antagonists of an inhibitory signal of an immune cell. In a specific embodiment, the antagonist of an inhibitory signal of an immune cell is the anti-CTLA-4 antibody described in Section 6, infra. In another specific embodiment, the agonist of a co-stimulatory signal of an immune cell is the ICOS ligand described in Section 6, infra
  • 5.1 Newcastle Disease Virus
  • Any NDV type or strain may be used in a combination therapy disclosed herein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses. In a specific embodiment, the NDV used in a combination therapy disclosed herein is a naturally-occurring strain. In certain embodiments, the NDV is a lytic strain. In other embodiments, the NDV used in a combination therapy disclosed herein is a non-lytic strain. In certain embodiments, the NDV used in a combination therapy disclosed herein is lentogenic strain. In some embodiments, the NDV is a mesogenic strain. In other embodiments, the NDV used in a combination therapy disclosed herein is a velogenic strain. Specific examples of NDV strains include, but are not limited to, the 73-T strain, NDV HUJ strain, Ulster strain, MTH-68 strain, Italien strain, Hickman strain, PV701 strain, Hitchner B1 strain (see, e.g., Genbank No. AF309418 or NC_002617), La Sota strain (see, e.g., Genbank No. AY845400), YG97 strain, MET95 strain, Roakin strain, and F48E9 strain. In a specific embodiment, the NDV used in a combination therapy disclosed herein that is the Hitchner B1 strain. In another specific embodiment, the NDV used in a combination therapy disclosed herein is a B1 strain as identified by Genbank No. AF309418 or NC_002617. In another specific embodiment, the NDV used in a combination therapy disclosed herein is the NDV identified by ATCC No. VR2239. In another specific embodiment, the NDV used in a combination therapy disclosed herein is the La Sota strain.
  • In specific embodiments, the NDV used in a combination therapy disclosed herein is not pathogenic birds as assessed by a technique known to one of skill. In certain specific embodiments, the NDV used in a combination therapy is no pathogenic as assessed by intracranial injection of 1-day-old chicks with the virus, and disease development and death as scored for 8 days. In some embodiments, the NDV used in a combination therapy disclosed herein has an intracranial pathogenicity index of less than 0.7, less than 0.6, less than 0.5, less than 0.4, less than 0.3, less than 0.2 or less than 0.1. In certain embodiments, the NDV used in a combination therapy disclosed herein has an intracranial pathogenicity index is zero.
  • In certain embodiments, the NDV used in a combination therapy disclosed herein is a mesogenic strain that has been genetically engineered so as not be a considered pathogenic in birds as techniques known to one skilled in the art. In certain embodiments, the NDV used in a combination therapy disclosed herein is a velogenic strain that has been genetically engineered so as not be a considered pathogenic in birds as techniques known to one skilled in the art.
  • In certain embodiments, the NDV used in a combination therapy disclosed herein expresses a mutated F protein. In a specific embodiment, the NDV used in a combination therapy expresses a mutated F protein is highly fusogenic and able to form syncytia. In another specific embodiment, the mutated F protein is incorporated into the virion.
  • In one embodiment, a genome of a NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein with a mutated cleavage site. In a specific embodiment, the NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein in which the cleavage site of the F protein is mutated to produce a polybasic amino acid sequence, which allows the protein to be cleaved by intracellular proteases, which makes the virus more effective in entering cells and forming syncytia. In another specific embodiment, the NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein in which the cleavage site of the F protein is replaced with one containing one or two extra arginine residues, allowing the mutant cleavage site to be activated by ubiquitously expressed proteases of the furin family. Specific examples of NDVs that express such a mutated F protein include, but are not limited to, rNDV/F2aa and rNDV/F3aa. For a description of mutations introduced into a NDV F protein to produce a mutated F protein with a mutated cleavage site, see, e.g., Park et al. (2006) Engineered viral vaccine constructs with dual specificity: avian influenza and Newcastle disease. PNAS USA 103: 8203-2808, which is incorporated herein by reference in its entirety. In some embodiments, the NDV used in a combination therapy disclosed herein is engineered to express a mutated F protein with the amino acid mutation L289A. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein.
  • In certain embodiments, the NDV used in a combination therapy disclosed herein is attenuated such that the NDV remains, at least partially, infectious and can replicate in vivo, but only generate low titers resulting in subclinical levels of infection that are non-pathogenic (see, e.g., Khattar et al., 2009, J. Virol. 83:7779-7782). In a specific embodiment, the NDV is attenuated by deletion of the V protein. Such attenuated NDVs may be especially suited for embodiments wherein the virus is administered to a subject in order to act as an immunogen, e.g., a live vaccine. The viruses may be attenuated by any method known in the art.
  • In certain embodiments, the NDV used in a combination therapy disclosed herein does not comprise an NDV V protein encoding sequence. In other embodiments, the NDV used in a combination therapy disclosed herein expresses a mutated V protein. See, e.g., Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844, which is incorporated herein by reference, for examples of mutated V proteins. In certain embodiments, a mesogenic or velogenic NDV strain used in a combination therapy disclosed herein expresses a mutated V protein, such as disclosed by Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844.
  • In certain embodiments, the NDV used in a combination therapy disclosed herein is an NDV disclosed in U.S. Pat. No. 7,442,379, U.S. Pat. No. 6,451,323, or U.S. Pat. No. 6,146,642, which is incorporated herein by reference in its entirety. In specific embodiments, the NDV used in a combination therapy disclosed herein is genetically engineered to encode and express a heterologous peptide or protein. In certain embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV known to one of skill in the art, or a chimeric NDV disclosed herein (see, e.g., Section 5.2, infra). In some embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express a tumor antigen (see below for examples of tumor antigens). In certain embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express a heterologous interferon antagonist (see below for examples of heterologous interferon antagonists). In some embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV disclosed in U.S. patent application publication No. 2012/0058141, which is incorporated herein by reference in its entirety. In certain embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV disclosed in U.S. patent application publication No. 2012/0122185, which is incorporated herein by reference in its entirety. In some embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express a cytokine, such as, e.g., IL-2, IL-7, IL-9, IL-15, IL-17, IL-21, IL-22, IFN-gamma, GM-CSF, and TNF-alpha. In some embodiments, the NDV used in a combination therapy disclosed herein is a chimeric NDV comprising a genome engineered to express IL-2.
  • 5.2 Chimeric Newcastle Disease Virus
  • In one aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or Natural Killer (NK) cell. In a specific embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell. In another specific embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell. In other words, the NDV serves as the “backbone” that is engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or Natural Killer (NK) cell. Specific examples of agonists of co-stimulatory signals as well as specific examples of antagonists of inhibitory signal are provided below.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein. In one embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein. In another embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein. In a specific embodiment, the mutated F protein is highly fusogenic and able to form syncytia. In another specific embodiment, the mutated F protein is incorporated into the virion. In certain embodiments, the genome of a NDV engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, comprises an NDV V protein encoding sequence.
  • In one embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a mutated F protein with a mutated cleavage site. In a specific embodiment, the NDV is engineered to express a mutated F protein in which the cleavage site of the F protein is mutated to produce a polybasic amino acid sequence, which allows the protein to be cleaved by intracellular proteases, which makes the virus more effective in entering cells and forming syncytia. In another specific embodiment, the NDV is engineered to express a mutated F protein in which the cleavage site of the F protein is replaced with one containing one or two extra arginine residues, allowing the mutant cleavage site to be activated by ubiquitously expressed proteases of the furin family. Specific examples of NDVs that express such a mutated F protein include, but are not limited to, rNDV/F2aa and rNDV/F3aa. For a description of mutations introduced into a NDV F protein to produce a mutated F protein with a mutated cleavage site, see, e.g., Park et al. (2006) Engineered viral vaccine constructs with dual specificity: avian influenza and Newcastle disease. PNAS USA 103: 8203-2808, which is incorporated herein by reference in its entirety. In some embodiments, the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein. In specific embodiments, the mutated F protein is incorporated into the virion.
  • In some embodiments, the genome of a NDV engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, comprises a mutated NDV V protein encoding sequence, such as disclosed by Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844. In other embodiments, the genome of a NDV engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell does not comprise an NDV V protein encoding sequence. In certain embodiments, parental backbone of the chimeric NDV is a mesogenic or velogenic NDV strain that is engineered to express a mutated V protein, such as disclosed by Elankumaran et al., 2010, J. Virol. 84(8): 3835-3844.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a cytokine. In a specific embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a cytokine. In a specific embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a cytokine. Specific examples of cytokines include, but are not limited to, interleukin (IL)-2, IL-7, IL-9, IL-15, IL-17, IL-21, IL-22, interferon (IFN) gamma, GM-CSF, and tumor necrosis factor (TNF)-alpha.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a cytokine (e.g., IL-2, IL-7, IL-9, IL-15, IL-17, IL-21, IL-22, IFN-gamma, GM-CSF, and TNF-alpha). In a specific embodiment, the mutated F protein are highly fusogenic. In a specific embodiment, the mutated F protein has a mutant cleavage site (such as described herein). In some embodiments, the mutated F protein comprises the amino acid mutation L289A. In some embodiments, the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein. In specific embodiments, the mutated F protein is incorporated into the virion.
  • In a specific embodiment, described herein are chimeric NDVs, comprising a genome engineered to express (i) an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, and (ii) a tumor antigen. In a specific embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a tumor antigen. In a specific embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a tumor antigen.
  • Tumor antigens include tumor-associated antigens and tumor-specific antigens. Specific examples of tumor antigens include, but are not limited to, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, N-acetylglucosaminyltransferase-V, p-15, gp100, MART-1/MelanA, TRP-1 (gp75), Tyrosinase, cyclin-dependent kinase 4, β-catenin, MUM-1, CDK4, HER-2/neu, human papillomavirus-E6, human papillomavirus E7, CD20, carcinoembryonic antigen (CEA), epidermal growth factor receptor, MUC-1, caspase-8, CD5, mucin-1, Lewisx, CA-125, p185HER2, IL-2R, Fap-α, tenascin, antigens associated with a metalloproteinase, and CAMPATH-1. Other examples include, but are not limited to, KS 1/4 pan-carcinoma antigen, ovarian carcinoma antigen (CA125), prostatic acid phosphate, prostate specific antigen, melanoma-associated antigen p97, melanoma antigen gp75, high molecular weight melanoma antigen (HMW-MAA), prostate specific membrane antigen, CEA, polymorphic epithelial mucin antigen, milk fat globule antigen, colorectal tumor-associated antigens (such as: CEA, TAG-72, CO17-1A, GICA 19-9, CTA-1 and LEA), Burkitt's lymphoma antigen-38.13, CD19, B-lymphoma antigen-CD20, CD33, melanoma specific antigens (such as ganglioside GD2, ganglioside GD3, ganglioside GM2, ganglioside GM3), tumor-specific transplantation type of cell-surface antigen (TSTA) (such as virally-induced tumor antigens including T-antigen DNA tumor viruses and Envelope antigens of RNA tumor viruses), oncofetal antigen-alpha-fetoprotein such as CEA of colon, bladder tumor oncofetal antigen, differentiation antigen (such as human lung carcinoma antigen L6 and L20), antigens of fibrosarcoma, leukemia T cell antigen-Gp37, neoglycoprotein, sphingolipids, breast cancer antigens (such as EGFR (Epidermal growth factor receptor), HER2 antigen (p185.sup.HER2) and HER2 neu epitope), polymorphic epithelial mucin (PEM), malignant human lymphocyte antigen-APO-1, differentiation antigen (such as I antigen found in fetal erythrocytes, primary endoderm, I antigen found in adult erythrocytes, preimplantation embryos, I(Ma) found in gastric adenocarcinomas, M18, M39 found in breast epithelium, SSEA-1 found in myeloid cells, VEP8, VEP9, My1, VIM-D5, D.sub.156-22 found in colorectal cancer, TRA-1-85 (blood group H), C14 found in colonic adenocarcinoma, F3 found in lung adenocarcinoma, AH6 found in gastric cancer, Y hapten, Le.sup.y found in embryonal carcinoma cells, TL5 (blood group A), EGF receptor found in A431 cells, E1 series (blood group B) found in pancreatic cancer, FC10.2 found in embryonal carcinoma cells, gastric adenocarcinoma antigen, CO-514 (blood group Lea) found in Adenocarcinoma, NS-10 found in adenocarcinomas, CO-43 (blood group Leb), G49 found in EGF receptor of A431 cells, MH2 (blood group ALeb/Ley) found in colonic adenocarcinoma, 19.9 found in colon cancer, gastric cancer mucins, T5A7 found in myeloid cells, R24 found in melanoma, 4.2, GD3, D1.1, OFA-1, GM2, OFA-2, GD2, and M1:22:25:8 found in embryonal carcinoma cells, and SSEA-3 and SSEA-4 found in 4 to 8-cell stage embryos), T cell receptor derived peptide from a Cutaneous T cell Lymphoma, C-reactive protein (CRP), cancer antigen-50 (CA-50), cancer antigen 15-3 (CA15-3) associated with breast cancer, cancer antigen-19 (CA-19) and cancer antigen-242 associated with gastrointestinal cancers, carcinoma associated antigen (CAA), chromogranin A, epithelial mucin antigen (MC5), human epithelium specific antigen (E1A), Lewis(a)antigen, melanoma antigen, melanoma associated antigens 100, 25, and 150, mucin-like carcinoma-associated antigen, multidrug resistance related protein (MRPm6), multidrug resistance related protein (MRP41), Neu oncogene protein (C-erbB-2), neuron specific enolase (NSE), P-glycoprotein (mdr1 gene product), multidrug-resistance-related antigen, p170, multidrug-resistance-related antigen, prostate specific antigen (PSA), CD56, and NCAM.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a tumor antigen. In a specific embodiment, the mutated F protein are highly fusogenic. In a specific embodiment, the mutated F protein has a mutant cleavage site (such as described herein). In some embodiments, the mutated F protein comprises the amino acid mutation L289A. In some embodiments, the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein. In specific embodiments, the mutated F protein is incorporated into the virion.
  • In another specific embodiment, described herein are chimeric NDVs, comprising a genome engineered to express (i) an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, and (ii) a heterologous interferon antagonist. In a specific embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a heterologous interferon antagonist. In a specific embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a heterologous interferon antagonist. See, e.g., U.S. patent application publication No. 2012-0058141, which is incorporated herein by reference, for examples of chimeric NDV engineered to express heterologous interferon antagonists.
  • Interferon antagonists may be identified using any technique known to one of skill in the art, including, e.g., the techniques described in U.S. Pat. Nos. 6,635,416; 7,060,430; and 7,442,527; which are incorporated herein by reference in their entirety. In a specific embodiment, the heterologous interferon antagonist is a viral protein. Such viral proteins may be obtained or derived from any virus and the virus may infect any species (e.g., the virus may infect humans or non-human mammals). Exemplary heterologous interferon antagonists include, without limitation, Nipah virus W protein, Nipah V protein, Ebola virus VP35 protein, vaccinia virus E3L protein, influenza virus NS1 protein, respiratory syncytial virus (RSV) NS2 protein, herpes simplex virus (HSV) type 1 ICP34.5 protein, Hepatitis C virus NS3-4 protease, dominant-negative cellular proteins that block the induction or response to innate immunity (e.g., STAT1, MyD88, IKK and TBK), and cellular regulators of the innate immune response (e.g., SOCS proteins, PIAS proteins, CPLD proteins, IkB protein, Atg5 protein, Pin1 protein, IRAK-M protein, and UBP43). See, e.g., U.S. patent application publication No. 2012-0058141, which is incorporated herein by reference in its entirety, for additional information regarding heterologous interferon antagonist.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a heterologous interferon antagonist. In a specific embodiment, the mutated F protein are highly fusogenic. In a specific embodiment, the mutated F protein has a mutant cleavage site (such as described herein). In some embodiments, the mutated F protein comprises the amino acid mutation L289A. In some embodiments, the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein. In specific embodiments, the mutated F protein is incorporated into the virion.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a pro-apoptotic molecule. In a specific embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a pro-apoptotic molecule. In a specific embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and a pro-apoptotic molecule. Specific examples of pro-apoptotic molecules include, but are not limited to, Bax, Bak, Bad, BID, Bcl-xS, Bim, Noxa, Puma, AIF, FasL, and TRAIL.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and a pro-apoptotic molecule. In a specific embodiment, the mutated F protein are highly fusogenic. In a specific embodiment, the mutated F protein has a mutant cleavage site (such as described herein). In some embodiments, the mutated F protein comprises the amino acid mutation L289A. In some embodiments, the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein. In specific embodiments, the mutated F protein is incorporated into the virion.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and an anti-apoptotic molecule. In a specific embodiment, a genome of a NDV is engineered to express an agonist of a co-stimulatory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and an anti-apoptotic molecule. In a specific embodiment, a genome of a NDV is engineered to express an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and an anti-apoptotic molecule. Specific examples of anti-apoptotic molecules include, but are not limited to, Bcl-2, Bcl-xL, Mcl-1, and XIAP.
  • In another aspect, described herein are chimeric NDVs, comprising a genome engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, a mutated F protein, and an anti-apoptotic molecule. In a specific embodiment, the mutated F protein are highly fusogenic. In a specific embodiment, the mutated F protein has a mutant cleavage site (such as described herein). In some embodiments, the mutated F protein comprises the amino acid mutation L289A. In some embodiments, the chimeric NDV is engineered to express a mutated F protein with the amino acid mutation L289A. In certain embodiments, the mutated F protein is from a different type or strain of NDV than the backbone NDV. In specific embodiments the L289A mutated F protein possesses one, two or three arginine residues in the cleavage site. In some embodiments, the mutated F protein is in addition to the backbone NDV F protein. In specific embodiments, the mutated F protein replaces the backbone NDV F protein. In specific embodiments, the mutated F protein is incorporated into the virion.
  • Any NDV type or strain may be used as a backbone that is engineered to express an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and in certain embodiments, engineered to express a cytokine, tumor antigen, heterologous interferon antagonist and/or mutated F protein, including, but not limited to, naturally-occurring strains, variants or mutants, mutagenized viruses, reassortants and/or genetically engineered viruses. In a specific embodiment, the NDV used in a combination therapy disclosed herein is a naturally-occurring strain. In certain embodiments, the NDV that serves as the backbone for genetic engineering is a lytic strain. In other embodiments, the NDV that serves as the backbone for genetic engineering is a non-lytic strain. In certain embodiments, the NDV that serves as the backbone for genetic engineering is lentogenic strain. In some embodiments, the NDV that serves as the backbone for genetic engineering is mesogenic strain. In other embodiments, the NDV that serves as the backbone for genetic engineering is a velogenic strain. Specific examples of NDV strains include, but are not limited to, the 73-T strain, NDV HUJ strain, Ulster strain, MTH-68 strain, Italien strain, Hickman strain, PV701 strain, Hitchner B1 strain, La Sota strain (see, e.g., Genbank No. AY845400), YG97 strain, MET95 strain, Roakin strain, and F48E9 strain. In a specific embodiment, the NDV that serves as the backbone for genetic engineering is the Hitchner B1 strain. In another specific embodiment, the NDV that serves as the backbone for genetic engineering is a B1 strain as identified by Genbank No. AF309418 or NC_002617. In another specific embodiment, the NDV that serves as the backbone for genetic engineering is the NDV identified by ATCC No. VR2239. In another specific embodiment, the NDV that serves as the backbone for genetic engineering is the La Sota strain.
  • In certain embodiments, attenuation, or further attenuation, of the chimeric NDV is desired such that the chimeric NDV remains, at least partially, infectious and can replicate in vivo, but only generate low titers resulting in subclinical levels of infection that are non-pathogenic (see, e.g., Khattar et al., 2009, J. Virol. 83:7779-7782). In a specific embodiment, the NDV is attenuated by deletion of the V protein. Such attenuated chimeric NDVs may be especially suited for embodiments wherein the virus is administered to a subject in order to act as an immunogen, e.g., a live vaccine. The viruses may be attenuated by any method known in the art.
  • In specific embodiments, in addition to expressing an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and in certain embodiments, a mutated F protein and a cytokine, a chimeric NDV is engineered to express a suicide gene (e.g., thymidine kinase) or another molecule that inhibits NDV replication or function (a gene that makes NDV sensitive to an antibiotic or an anti-viral agent). In some embodiments, in addition to expressing an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte or NK cell, and in certain embodiments, a mutated F protein and a cytokine, a chimeric NDV is engineered to encode tissue-specific microRNA (miRNA) target sites (e.g., sites targeted by miR-21, miR-184, miR-133a/133b, miR-137, and/or miR-193a microRNAs).
  • In certain embodiments, the tropism of the chimeric NDV is altered. In a specific embodiment, the tropism of the virus is altered by modification of the F protein cleavage site to be recognized by tissue-specific or tumor-specific proteases such as matrix metalloproteases (MMP) and urokinase. In other embodiments, tropism of the virus is altered by introduction of tissue-specific miRNA target sites. In certain embodiments NDV HN protein is mutated to recognize tumor-specific receptor.
  • In embodiments herein, the agonist of a co-stimulatory signal and/or the antagonist of an inhibitory signal of an immune cell may be inserted into the genome of the backbone NDV between two transcription units. In a specific embodiment, the agonist of a co-stimulatory signal and/or the antagonist of an inhibitory signal of an immune cell is inserted into the genome of the backbone NDV between the M and P transcription units or between the HN and L transcription units. In accordance with other embodiments herein, the cytokine, and/or mutated F protein are inserted into the genome of the backbone NDV between two or more transcription units (e.g., between the M and P transcription units or between the HN and L transcription units).
  • 5.2.1. Immune Cell Stimulatory Agents
  • The chimeric NDVs described herein may be engineered to express any agonist of a co-stimulatory signal and/or any antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage), known to one of skill in the art. In specific embodiments, the agonist and/or antagonist is an agonist of a human co-stimulatory signal of an immune cell and/or antagoinst of a human inhibitory signal of an immune cell. In certain embodiments, the agonist of a co-stimulatory signal is an agonist of a co-stimulatory molecule (e.g., co-stimulatory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+ T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages). Specific examples of co-stimulatory molecules include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell costimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-1BB (CD137), CD40, lymphotoxin alpha (LT alpha), and LIGHT (lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes). In specific embodiments, the agonist is an agonist of a human co-stimulatory receptor of an immune cell. In certain embodiments, the agonist of a co-stimulatory receptor is not an agonist of ICOS. In some embodiments, the antagonist is an antagonist of an inhibitory molecule (e.g., inhibitory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+ T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages). Specific examples of inhibitory molecules include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD160, and adenosine A2a receptor (A2aR). In specific embodiments, the antagonist is an antagonist of a human inhibitory receptor of an immune cell.
  • In a specific embodiment, the agonist of a co-stimulatory receptor is an antibody or antigen-binding fragment thereof that specifically binds to the co-stimulatory receptor. Specific examples of co-stimulatory receptors include GITR, ICOS, OX40, CD27, CD28, 4-1BB, or CD40. In certain specific embodiments, the antibody is a monoclonal antibody. In other specific embodiments, the antibody is an sc-Fv. In a specific embodiment the antibody is a bispecific antibody that binds to two receptors on an immune cell. In other embodiments, the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell. In specific embodiments, the antibody is a human or humanized antibody. In other embodiments the ligand or an antibody is a chimeric protein with NDV F protein or NDV HN protein. See, e.g., U.S. patent application Publication No. 2012/0122185, which is incorporated herein by reference for a description regarding generation of chimeric F or chimeric HN proteins. In a specific embodiment, the chimeric protein is the chimeric F protein described in Section 6, infra. The techniques described below for generating the chimeric ICOSL-F protein can be used to generate other chimeric F proteins or chimeric HN proteins.
  • In another embodiment, the agonist of a co-stimulatory receptor is a ligand of the co-stimulatory receptor. In certain embodiments, the ligand is fragment of a native ligand. Specific examples of native ligands include B7RP1, CD137L, OX40L, CD70, herpes virus entry mediator (HVEM), CD80, and CD86. The nucleotide sequences encoding native ligands as well as the amino acid sequences of native ligands are known in the art. For example, the nucleotide and amino acid sequences of B7RP1 (GenBank human: NM_015259.4, NP_056074.1 murine: NM_015790.3, NP_056605.1), CD137L (GenBank human: NM_003811.3, NP_003802.1, murine: NM_009404.3, NP_033430.1), OX40L (GenBank human: NM_003326.3, NP_003317.1, murine: NM_009452.2, NP_033478.0, CD70 (GenBank human: NM_001252.3, NP_001243.1, murine: NM_011617.2, AAD00274.1), CD80 (GenBank human: NM_05191.3, NP_005182.1, murine: NM_009855.2, NP_033985.3), and CD86 (GenBank human: NM_005191.3, CAG46642.1, murine: NM_019388.3, NP_062261.3) can be found in GenBank. In other embodiments, the ligand is a derivative of a native ligand. In some embodiments, the ligand is a fusion protein comprising at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and a heterologous amino acid sequence. In specific embodiments, the fusion protein comprises at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and the Fc portion of an immunoglobulin or a fragment thereof. An example of a ligand fusion protein is a 4-1BB ligand fused to Fc portion of immunoglobulin (described by Meseck M et al., J Immunother. 2011 34:175-82).
  • In another embodiment, the antagonist of an inhibitory receptor is an antibody (or an antigen-binding fragment) or a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s). Specific examples of native ligands for inhibitory receptors include PDL-1, PDL-2, B7-H3, B7-H4, HVEM, Ga19 and adenosine. Specific examples of inhibitory receptors that bind to a native ligand include CTLA-4, PD-1, BTLA, KIR, LAG3, TIM3, and A2aR.
  • In specific embodiments, the antagonist of an inhibitory receptor is a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s). In certain embodiments, the soluble receptor is a fragment of a native inhibitory receptor or a fragment of a derivative of a native inhibitory receptor that specifically binds to native ligand (e.g., the extracellular domain of a native inhibitory receptor or a derivative of an inhibitory receptor). In some embodiments, the soluble receptor is a fusion protein comprising at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor (e.g., the extracellular domain of the native inhibitory receptor or a derivative of the native inhibitory receptor), and a heterologous amino acid sequence. In specific embodiments, the fusion protein comprises at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor, and the Fc portion of an immunoglobulin or a fragment thereof. An example of a soluble receptor fusion protein is a LAG3-Ig fusion protein (described by Huard B et al., Eur J Immunol. 1995 25:2718-21).
  • In specific embodiments, the antagonist of an inhibitory receptor is an antibody (or an antigen-binding fragment) that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s). In certain specific embodiments, the antibody is a monoclonal antibody. In other specific embodiments, the antibody is an scFv. In particular embodiments, the antibody is a human or humanized antibody. A specific example of an antibody to inhibitory ligand is anti-PD-L1 antibody (Iwai Y, et al. PNAS 2002; 99:12293-12297).
  • In another embodiment, the antagonist of an inhibitory receptor is an antibody (or an antigen-binding fragment) or ligand that binds to the inhibitory receptor, but does not transduce an inhibitory signal(s). Specific examples of inhibitory receptors include CTLA-4, PD1, BTLA, KIR, LAG3, TIM3, and A2aR. In certain specific embodiments, the antibody is a monoclonal antibody. In other specific embodiments, the antibody is an scFv. In particular embodiments, the antibody is a human or humanized antibody. A specific example of an antibody to inhibitory receptor is anti-CTLA-4 antibody (Leach D R, et al. Science 1996; 271: 1734-1736). Another example of an antibody to inhibitory receptor is anti-PD-1 antibody (Topalian S L, NEJM 2012; 28:3167-75).
  • In certain embodiments, a chimeric NDV described herein is engineered to an antagonist of CTLA-4, such as, e.g., Ipilimumab or Tremelimumab. In certain embodiments, a chimeric NDV described herein is engineered to an antagonist of PD1, such as, e.g., MDX-1106 (BMS-936558), MK3475, CT-011, AMP-224, or MDX-1105. In certain embodiments, a chimeric NDV described herein is engineered to express an antagonist of LAG3, such as, e.g., IMP321. In certain embodiments, a chimeric NDV described herein is engineered to express an antibody (e.g., a monoclonal antibody or an antigen-binding fragment thereof, or scFv) that binds to B7-H3, such as, e.g., MGA271. In specific embodiments, a chimeric NDV described herein is engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell described in Section 6, infra. In specific embodiments, NDV described herein is engineered to express anti-CD28 scvFv, ICOSL, CD40L, OX40L, CD137L, GITRL, and/or CD70.
  • In certain embodiments, an agonist of a co-stimulatory signal of an immune cell induces (e.g., selectively) induces one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand. In specific embodiments, an agonist of a co-stimulatory receptor induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands in the absence of the agonist. In specific embodiments, an agonist of a co-stimulatory receptor: (i) induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not induce, or induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to such one or more other ligands in the absence of the agonist.
  • In certain embodiments, an agonist of a co-stimulatory signal of an immune cell activates or enhances (e.g., selectively activates or enhances) one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand. In specific embodiments, an agonist of a co-stimulatory receptor activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of co-stimulatory receptor to one or more of its ligands in the absence of the agonist. In specific embodiments, an agonist of a co-stimulatory receptor: (i) an agonist of a co-stimulatory signal activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not activate or enhance, or activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to such one or more other ligands in the absence of the agonist.
  • In some embodiments, an antagonist of an inhibitory signal of an immune cell (e.g., selectively) inhibits or reduces one or more of the signal transduction pathways induced by the binding of an inhibitory receptor to its ligand. In specific embodiments, an antagonist of an inhibitory receptor inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands in the absence of the antagonist. In specific embodiments, an antagonist of an inhibitory receptor: (i) inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to the one particular ligand in the absence of the antagonist; and (ii) does not inhibit or reduce, or inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or more signal transduction pathways induced by the binding of inhibitory receptor to such one or more other ligands in the absence of the antagonist.
  • In specific embodiments, an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell induces, activates and/or enhances one or more immune activities, functions or responses. The one or more immune activities, functions or responses can be in the form of, e.g., an antibody response (humoral response) or a cellular immune response, e.g., cytokine secretion (e.g., interferon-gamma), helper activity or cellular cytotoxicity. In one embodiment, expression of an activation marker on immune cells (e.g., CD44, Granzyme, or Ki-67), expression of a co-stimulatory receptor on immune cells (e.g., ICOS, CD28, OX40, or CD27), expression of a ligand for a co-stimulatory receptor (e.g., B7HRP1, CD80, CD86, OX40L, or CD70), cytokine secretion, infiltration of immune cells (e.g., T-lymphocytes, B lymphocytes and/or NK cells) to a tumor, antibody production, effector function, T cell activation, T cell differentiation, T cell proliferation, B cell differentiation, B cell proliferation, and/or NK cell proliferation is induced, activated and/or enhanced following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell. In another embodiment, myeloid-derived suppressor cell (MDSC) tumor infiltration and proliferation, Treg tumor infiltration, activation and proliferation, peripheral blood MDSC and Treg counts are inhibited following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • 5.3 Construction of NDVS
  • The NDVs described herein can be generated using the reverse genetics technique. The reverse genetics technique involves the preparation of synthetic recombinant viral RNAs that contain the non-coding regions of the negative-strand, viral RNA which are essential for the recognition by viral polymerases and for packaging signals necessary to generate a mature virion. The recombinant RNAs are synthesized from a recombinant DNA template and reconstituted in vitro with purified viral polymerase complex to form recombinant ribonucleoproteins (RNPs) which can be used to transfect cells. A more efficient transfection is achieved if the viral polymerase proteins are present during transcription of the synthetic RNAs either in vitro or in vivo. The synthetic recombinant RNPs can be rescued into infectious virus particles. The foregoing techniques are described in U.S. Pat. No. 5,166,057 issued Nov. 24, 1992; in U.S. Pat. No. 5,854,037 issued Dec. 29, 1998; in U.S. Pat. No. 6,146,642 issued Nov. 14, 2000; in European Patent Publication EP 0702085A1, published Feb. 20, 1996; in U.S. patent application Ser. No. 09/152,845; in International Patent Publications PCT WO97/12032 published Apr. 3, 1997; WO96/34625 published Nov. 7, 1996; in European Patent Publication EP A780475; WO 99/02657 published Jan. 21, 1999; WO 98/53078 published Nov. 26, 1998; WO 98/02530 published Jan. 22, 1998; WO 99/15672 published Apr. 1, 1999; WO 98/13501 published Apr. 2, 1998; WO 97/06270 published Feb. 20, 1997; and EPO 780 475A1 published Jun. 25, 1997, each of which is incorporated by reference herein in its entirety.
  • The helper-free plasmid technology can also be utilized to engineer a NDV described herein. Briefly, a complete cDNA of a NDV (e.g., the Hitchner B1 strain) is constructed, inserted into a plasmid vector and engineered to contain a unique restriction site between two transcription units (e.g., the NDV P and M genes; or the NDV HN and L genes). A nucleotide sequence encoding a heterologous amino acid sequence (e.g., a nucleotide sequence encoding an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell) may be inserted into the viral genome at the unique restriction site. Alternatively, a nucleotide sequence encoding a heterologous amino acid sequence (e.g., a nucleotide sequence encoding an agonist of a co-stimulatory signal and/or an antagonist of an inhibitory signal of an immune cell) may be engineered into a NDV transcription unit so long as the insertion does not affect the ability of the virus to infect and replicate. The single segment is positioned between a T7 promoter and the hepatitis delta virus ribozyme to produce an exact negative transcript from the T7 polymerase. The plasmid vector and expression vectors comprising the necessary viral proteins are transfected into cells leading to production of recombinant viral particles (see, e.g., International Publication No. WO 01/04333; U.S. Pat. Nos. 7,442,379, 6,146,642, 6,649,372, 6,544,785 and 7,384,774; Swayne et al. (2003). Avian Dis. 47:1047-1050; and Swayne et al. (2001). J. Virol. 11868-11873, each of which is incorporated by reference in its entirety).
  • Techniques for the production of a chimeric NDV that express an antibody are known in the art. See, e.g., Puhler et al., Gene Ther. 15(5): 371-283 (2008) for the generation of a recombinant NDV expressing a full IgG from two transgenes.
  • Bicistronic techniques to produce multiple proteins from a single mRNA are known to one of skill in the art. Bicistronic techniques allow the engineering of coding sequences of multiple proteins into a single mRNA through the use of IRES sequences. IRES sequences direct the internal recruitment of ribozomes to the RNA molecule and allow downstream translation in a cap independent manner. Briefly, a coding region of one protein is inserted into the ORF of a second protein. The insertion is flanked by an IRES and any untranslated signal sequences necessary for proper expression and/or function. The insertion must not disrupt the open reading frame, polyadenylation or transcriptional promoters of the second protein (see e.g., Garcia-Sastre et al., 1994, J. Virol. 68:6254-6261 and Garcia-Sastre et al., 1994 Dev. Biol. Stand. 82:237-246, each of which are incorporated by reference herein in their entirety).
  • 5.4 Propagation of NDVS
  • The NDVs described herein (e.g., the chimeric NDVs) can be propagated in any substrate that allows the virus to grow to titers that permit the uses of the viruses described herein. In one embodiment, the substrate allows the NDVs described herein (e.g., the chimeric NDVs) to grow to titers comparable to those determined for the corresponding wild-type viruses.
  • The NDVs described herein (e.g., the chimeric NDVs) may be grown in cells (e.g., avian cells, chicken cells, etc.) that are susceptible to infection by the viruses, embryonated eggs (e.g., chicken eggs or quail eggs) or animals (e.g., birds). Such methods are well-known to those skilled in the art. In a specific embodiment, the NDVs described herein (e.g., the chimeric NDVs) may be propagated in cancer cells, e.g., carcinoma cells (e.g., breast cancer cells and prostate cancer cells), sarcoma cells, leukemia cells, lymphoma cells, and germ cell tumor cells (e.g., testicular cancer cells and ovarian cancer cells). In another specific embodiment, the NDVs described herein (e.g., the chimeric NDVs) may be propagated in cell lines, e.g., cancer cell lines such as HeLa cells, MCF7 cells, THP-1 cells, U87 cells, DU145 cells, Lncap cells, and T47D cells. In another embodiment, the NDVs described herein (e.g., the chimeric NDVs) are propagated in chicken cells or embryonated eggs. Representative chicken cells include, but are not limited to, chicken embryo fibroblasts and chicken embryo kidney cells. In a specific embodiment, the NDVs described herein (e.g., the chimeric NDVs) are propagated in Vero cells. In another specific embodiment, the NDVs described herein (e.g., the chimeric NDVs) are propagated in cancer cells in accordance with the methods described in Section 6, infra. In another specific embodiment, the NDVs described herein (e.g., the chimeric NDVs) are propagated in chicken eggs or quail eggs. In certain embodiments, a NDV virus described herein (e.g., a chimeric NDV) is first propagated in embryonated eggs and then propagated in cells (e.g., a cell line).
  • The NDVs described herein (e.g., the chimeric NDVs) may be propagated in embryonated eggs, e.g., from 6 to 14 days old, 6 to 12 days old, 6 to 10 days old, 6 to 9 days old, 6 to 8 days old, or 10 to 12 days old. Young or immature embryonated eggs can be used to propagate the NDVs described herein (e.g., the chimeric NDVs). Immature embryonated eggs encompass eggs which are less than ten day old eggs, e.g., eggs 6 to 9 days old or 6 to 8 days old that are IFN-deficient. Immature embryonated eggs also encompass eggs which artificially mimic immature eggs up to, but less than ten day old, as a result of alterations to the growth conditions, e.g., changes in incubation temperatures; treating with drugs; or any other alteration which results in an egg with a retarded development, such that the IFN system is not fully developed as compared with ten to twelve day old eggs. The NDVs described herein (e.g., the chimeric NDVs) can be propagated in different locations of the embryonated egg, e.g., the allantoic cavity. For a detailed discussion on the growth and propagation viruses, see, e.g., U.S. Pat. No. 6,852,522 and U.S. Pat. No. 7,494,808, both of which are hereby incorporated by reference in their entireties.
  • For virus isolation, the NDVs described herein (e.g., the chimeric NDVs) can be removed from cell culture and separated from cellular components, typically by well known clarification procedures, e.g., such as gradient centrifugation and column chromatography, and may be further purified as desired using procedures well known to those skilled in the art, e.g., plaque assays.
  • 5.5 Compositions & Routes of Administration
  • Encompassed herein is the use of a NDV described herein (e.g., the chimeric NDVs) in compositions. Also encompassed herein is the use of plasma membrane fragments from NDV infected cells or whole cancer cells infected with NDV in compositions. In a specific embodiment, the compositions are pharmaceutical compositions, such as immunogenic formulations (e.g., vaccine formulations). The compositions may be used in methods of treating cancer.
  • In one embodiment, a pharmaceutical composition comprises a NDV described herein (e.g., the chimeric NDVs), in an admixture with a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition further comprises one or more additional prophylactic or therapeutic agents, such as described in Section 5.6.4, infra. In a specific embodiment, a pharmaceutical composition comprises an effective amount of a NDV described herein (e.g., the chimeric NDVs), and optionally one or more additional prophylactic of therapeutic agents, in a pharmaceutically acceptable carrier. In some embodiments, the NDV (e.g., a chimeric NDV) is the only active ingredient included in the pharmaceutical composition.
  • In another embodiment, a pharmaceutical composition (e.g., an oncolysate vaccine) comprises a protein concentrate or a preparation of plasma membrane fragments from NDV infected cancer cells, in an admixture with a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition further comprises one or more additional prophylactic or therapeutic agents, such as described in Section 5.6.4, infra. In another embodiment, a pharmaceutical composition (e.g., a whole cell vaccine) comprises cancer cells infected with NDV, in an admixture with a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition further comprises one or more additional prophylactic or therapeutic agents, such as described in Section 5.6.4, infra.
  • The pharmaceutical compositions provided herein can be in any form that allows for the composition to be administered to a subject. In a specific embodiment, the pharmaceutical compositions are suitable for veterinary and/or human administration. As used herein, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeiae for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the pharmaceutical composition is administered. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin. The formulation should suit the mode of administration.
  • In a specific embodiment, the pharmaceutical compositions are formulated to be suitable for the intended route of administration to a subject. For example, the pharmaceutical composition may be formulated to be suitable for parenteral, intrapleural, inhalation, intraperitoneal, oral, intradermal, colorectal, intraperitoneal and intratumoral administration. In a specific embodiment, the pharmaceutical composition may be formulated for intravenous, oral, intraperitoneal, intranasal, intratracheal, subcutaneous, intramuscular, topical, pulmonary, or intratumoral administration.
  • 5.6 Anti-Cancer Uses and Other Uses
  • In one aspect, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) may be used in the treatment of cancer. In one embodiment, provided herein are methods for treating cancer, comprising administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof. In a specific embodiment, provided herein is a method for treating cancer, comprising administering to a subject in need thereof an effective amount of a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof.
  • In specific embodiments, a chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell, or a composition thereof is administered to a subject to treat cancer. In another specific embodiments, a chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell, or a composition thereof is administered to a subject to treat cancer. In certain embodiments, a chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell and a mutated F protein or a composition thereof is administered to a subject to treat cancer. In certain embodiments, a chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell and a mutated F protein or a composition thereof is administered to a subject to treat cancer.
  • A chimeric NDV (e.g., a chimeric NDV described in Section 5.2, supra) described herein or a composition thereof, an oncolysate vaccine, or a whole cell cancer vaccine used in a method for treating cancer may be used as any line of therapy (e.g., a first, second, third, fourth or fifth line therapy).
  • In certain embodiments, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) is the only active ingredient administered to treat cancer. In specific embodiments, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) is the only active ingredient in a composition administered to treat cancer.
  • The chimeric NDV (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof may be administered locally or systemically to a subject. For example, the chimeric NDV (e.g., a chimeric NDV described in Section 5.2, supra) or composition may be administered parenterally (e.g., intravenously or subcutanously), intratumorally, intrapleurally, intranasally, intraperitoneally, orally, by inhalation, topically or intradermally to a subject.
  • In certain embodiments, the methods described herein include the treatment of cancer for which no treatment is available. In some embodiments, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof is administered to a subject to treat cancer as an alternative to other conventional therapies.
  • In one embodiment, provided herein is a method for treating cancer, comprising administering to a subject in need thereof a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof and one or more additional therapies, such as described in Section 5.6.4, infra. In a particular embodiment, one or more therapies are administered to a subject in combination with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof to treat cancer. In a specific embodiment, the additional therapies are currently being used, have been used or are known to be useful in treating cancer. In another embodiment, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) or a composition thereof is administered to a subject in combination with a supportive therapy, a pain relief therapy, or other therapy that does not have a therapeutic effect on cancer. In a specific embodiment, the one or more additional therapies administered in combination with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) is one or more of the therapies described in Section 5.6.4.1, infra. In certain embodiments, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) and one or more additional therapies are administered in the same composition. In other embodiments, a chimeric NDV and one or more additional therapies are administered in different compositions.
  • In certain embodiments, two, three or multiple NDVs (including one, two or more chimeric NDVs described herein, such as one, two or more of the chimeric NDVs described in Section 5.2, supra) are administered to a subject to treat cancer. The second or more chimeric NDVs used in accordance with methods described herein that comprise administration of two, three or multiple NDVs to a subject to treat cancer may be naturally occurring chimeric NDVs or engineered chimeric NDVs that have been engineered to express heterologous amino acid sequence (e.g., a cytokine). The first and second chimeric NDVs may be part of the same pharmaceutical composition or different pharmaceutical compositions. In certain embodiments, the first chimeric NDV and the second chimeric NDV are administered by the same route of administration (e.g., both are administered intratumorally or intravenously). In other embodiments, the first chimeric NDV and the second chimeric NDV are administered by different routes of administration (e.g., one is administered intratumorally and the other is administered intravenously).
  • In specific embodiments, a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell is administered to a patient to treat cancer in combination with a second chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell. In other specific embodiments, a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune is administered in combination with a second chimeric NDV engineered to express one, two or more of the following: a cytokine (e.g., IL-2), a heterologous interferon antagonist, a tumor antigen, a pro-apopototic molecule, and/or anti-apoptotic molecule. In a specific embodiment, the first chimeric NDV, the second chimeric NDV, or both express a mutated F protein that increases the fusogenic activity of the chimeric NDV. In another specific embodiment, the first chimeric NDV, the second chimeric NDV or both express a mutated F protein with a mutation in the cleavage site (such as described herein).
  • In specific embodiments, a first composition (e.g., a pharmaceutical composition) comprising a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell is administered to a patient to treat cancer in combination with a second composition (e.g., a pharmaceutical composition) comprising a second chimeric NDV engineered to express an antagonist of an inhibitory signal of an immune cell. In other specific embodiments, a first composition (e.g., a pharmaceutical composition) comprising a first chimeric NDV engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune is administered in combination with a second composition (e.g., a pharmaceutical composition) comprising a second chimeric NDV engineered to express one, two or more of the following: a cytokine (e.g., IL-2), a heterologous interferon antagonist, a tumor antigen, a pro-apopototic molecule, and/or anti-apoptotic molecule. In a specific embodiment, the first chimeric NDV, the second chimeric NDV, or both express a mutated F protein that increases the fusogenic activity of the chimeric NDV. In another specific embodiment, the first chimeric NDV, the second chimeric NDV or both express a mutated F protein with a mutation in the cleavage site (such as described herein).
  • In another aspect, an NDV described herein (e.g., an NDV described in Section 5.1, supra) may be used in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra (e.g., Section 5.6.4.1, infra), in the treatment of cancer. In one embodiment, provided herein are methods for treating cancer, comprising administering to a subject in need thereof an NDV described herein (e.g., an NDV described in Section 5.1, supra) or a composition thereof and one or more additional therapies, such as described herein in Section 5.6.4, infra. (e.g., Section 5.6.4.1). In a specific embodiment, provided herein is a method for treating cancer, comprising administering to a subject in need thereof an effective amount of an NDV described herein (e.g., an NDV described in Section 5.1, supra) or a composition thereof and an effective amount of one or more additional therapies, such as described in Section 5.6.4, infra. (e.g., Section 5.6.4.1). In certain embodiments, an NDV described herein (e.g., an NDV described in Section 5.1, supra) and one or more additional therapies, such as described in Section 5.6.4, infra (e.g., Section 5.6.4.1), are administered in the same composition. In other embodiments, an NDV (e.g., an NDV described in Section 5.1, supra) and one or more additional therapies are administered in different compositions.
  • An NDV (e.g., an NDV described in Section 5.1, supra) described herein or a composition thereof, an oncolysate vaccine, or a whole cell cancer vaccine in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra, may be used as any line of therapy (e.g., a first, second, third, fourth or fifth line therapy) for treating cancer in accordance with a method described herein.
  • In another aspect, whole cancer cells infected with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) can be used to treat cancer. In a specific embodiment, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) may be contacted with a cancer cell or a population of cancer cells and the infected cancer cell or population of cancer cells may be administered to a subject to treat cancer. In one embodiment, the cancer cells are subjected to gamma radiation prior to infection with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra). In another embodiment, the cancer cells are subjected to gamma radiation after infection with a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra). In a particular embodiment, the cancer cells are treated prior to administration to a subject so that the cancer cells cannot multiply in the subject. In a specific embodiment, the cancer cells cannot multiply in the subject and the virus cannot infect the subject. In one embodiment, the cancer cells are subjected to gamma radiation prior to administration to subject. In another embodiment, the cancer cells are sonicated prior to administration to a subject. In another embodiment, the cancer cells are treated with mitomycin C prior to administration to a subject. In another embodiment, the cancer cells are treated by freezing and thawing prior to administration to a subject. In another embodiment, the cancer cells are treated with heat treatment prior to administration to a subject. The cancer cells may be administered locally or systemically to a subject. For example, the cancer cells may be administered parenterally (e.g., intravenously or subcutaneously), intratumorally, intransally, orally, by inhalation, intrapleurally, topically or intradermally to a subject. In a specific embodiment, the cancer cells are administered intratumorally or to the skin (e.g., intradermally) of a subject. The cancer cells used may be autologous or allogeneic. In a specific embodiment, the backbone of the chimeric NDV is a non-lytic strain. The cancer cells may be administered to a subject alone or in combination with an additional therapy. The cancer cells are preferably in a pharmaceutical composition. In certain embodiments, the cancer cells are administered in combination with one or more additional therapies, such as described in Section 5.6.4, infra. In certain embodiments, the cancer cells and one or more additional therapies are administered in the same composition. In other embodiments, the cancer cells and one or more additional therapies are administered in different compositions.
  • In another aspect, whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) may be used in combination with one or more additional therapies described herein in Section 5.6.4, infra, in the treatment of cancer. In one embodiment, provided herein are methods for treating cancer, comprising administering to a subject in need thereof whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) in combination with one or more additional therapies described herein in Section 5.6.4, infra. In a specific embodiment, provided herein is a method for treating cancer, comprising administering to a subject in need thereof an effective amount of whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) in combination with an effective amount of one or more additional therapies described in Section 5.6.4, infra. In certain embodiments, whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) and one or more additional therapies described in Section 5.6.4, infra, are administered in the same composition. In other embodiments, whole cancer cells infected with an NDV described herein (e.g., an NDV described in Section 5.1, supra) and one or more additional therapies are administered in different compositions.
  • In another aspect, a protein concentrate or plasma membrane preparation from lysed cancer cells infected with a chimeric NDV (e.g., a chimeric NDV described in Section 5.2, supra) can be used to treat cancer. In one embodiment, a plasma membrane preparation comprising fragments from cancer cells infected with a chimeric NDV described herein can be used to treat cancer. In another embodiment, a protein concentrate from cancer cells infected with a chimeric NDV described herein can be used to treat cancer. Techniques known to one of skill in the art may be used to produce the protein concentrate or plasma membrane preparation. In a specific embodiment, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) may be contacted with a cancer cell or a population of cancer cells and the infected cancer cell or population of cancer cells may be lysed using techniques known to one of skill in the art to obtain protein concentrate or plasma membrane fragments of the NDV-infected cancer cells, and the protein concentrate or plasma membrane fragments of the NDV-infected cancer cells may be administered to a subject to treat cancer. The protein concentrate or plasma membrane fragments may be administered locally or systemically to a subject. For example, the protein concentrate or plasma membrane fragments may be administered parenterally, intratumorally, intransally, intrapleurally, orally, by inhalation, topically or intradermally to a subject. In a specific embodiment, such a protein concentrate or plasma membrane preparation is administered intratumorally or to the skin (e.g., intradermally) of a subject. The cancer cells used to produce the protein concentrate or plasma membrane preparation may be autologous or allogeneic. In a specific embodiment, the backbone of the chimeric NDV is a lytic strain. The protein concentrate or plasma membrane preparation may be administered to a subject alone or in combination with an additional therapy. The protein concentrate or plasma membrane preparation is preferably in a pharmaceutical composition. In certain embodiments, the protein concentrate or plasma membrane preparation is administered in combination with one or more additional therapies, such as described in Section 5.6.4, infra (e.g., Section 5.6.4.1) In certain embodiments, the protein concentrate or plasma membrane preparation and one or more additional therapies are administered in the same composition. In other embodiments, the protein concentrate or plasma membrane preparation and one or more additional therapies are administered in different compositions.
  • In another aspect, a protein concentrate or plasma membrane preparation from lysed cancer cells infected with an NDV (e.g., an NDV described in Section 5.1, supra) may be used in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra (e.g., Section 5.6.4.1), in the treatment of cancer. In one embodiment, provided herein are methods for treating cancer, comprising administering to a subject in need thereof a protein concentrate or plasma membrane preparation from lysed cancer cells infected with an NDV (e.g., an NDV described in Section 5.1, supra) in combination with one or more additional therapies, such as described herein in Section 5.6.4, infra. (e.g., Section 5.6.4.1). In a specific embodiment, provided herein is a method for treating cancer, comprising administering to a subject in need thereof an effective amount of a protein concentrate or plasma membrane preparation from lysed cancer cells infected with an NDV (e.g., an NDV described in Section 5.1, supra) in combination with an effective amount of one or more additional therapies, such as described in Section 5.6.4, infra. (e.g., Section 5.6.4.1). In certain embodiments, the protein concentrate or plasma membrane preparation and one or more additional therapies, such as described in Section 5.6.4, infra, are administered in the same composition. In other embodiments, the protein concentrate or plasma membrane preparation and one or more additional therapies are administered in different compositions.
  • In another aspect, the chimeric NDVs described herein (e.g., a chimeric NDV described in Section 5.2, supra) can be used to produce antibodies which can be used in diagnostic immunoassays, passive immunotherapy, and the generation of antiidiotypic antibodies. For example, a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra) can be administered to a subject (e.g., a mouse, rat, pig, horse, donkey, bird or human) to generate antibodies which can then be isolated and used, e.g., in diagnostic assays, passive immunotherapy and generation of antiidiotypic antibodies. In certain embodiments, an NDV described herein (e.g., an NDV described in Section 5.1 or 5.2, supra) is administered to a subject (e.g., a mouse, rat, pig, horse, donkey, bird, or human) in combination with one or more additional therapies, such as described in Section 5.6.4, infra, to generated antibodies which can then be isolated and used, e.g., in diagnostic assays, passive immunotherapy and generation of antiidiotypic antibodies. The generated antibodies may be isolated by standard techniques known in the art (e.g., immunoaffinity chromatography, centrifugation, precipitation, etc.) and used in diagnostic immunoassays, passive immunotherapy and generation of antiidiotypic antibodies.
  • In certain embodiments, the antibodies isolated from subjects administered a chimeric NDV described herein (e.g., a chimeric NDV described in Section 5.2, supra), or isolated from subjects administered an NDV described herein (e.g., an NDV described in Section 5.1 or 5.2, supra) in combination with one or more additional therapies, such as described in Section 5.6.4, infra, are used to assess the expression of NDV proteins, a heterologous peptide or protein expressed by a chimeric NDV, or both. Any immunoassay system known in the art may be used for this purpose including but not limited to competitive and noncompetitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assays), “sandwich” immunoassays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays and immunoelectrophoresis assays, to name but a few.
  • 5.6.1. Patient Population
  • In some embodiments, an NDV (e.g., a chimeric NDV) described herein or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject suffering from cancer. In other embodiments, an NDV (e.g., a chimeric NDV) described herein or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject predisposed or susceptible to cancer. In some embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject diagnosed with cancer. Specific examples of the types of cancer are described herein. In an embodiment, the subject has metastatic cancer. In another embodiment, the subject has stage 1, stage 2 or stage 3 cancer. In another embodiment, the subject is in remission. In yet another embodiment, the subject has a recurrence of cancer.
  • In certain embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human that is 0 to 6 months old, 6 to 12 months old, 6 to 18 months old, 18 to 36 months old, 1 to 5 years old, 5 to 10 years old, 10 to 15 years old, 15 to 20 years old, 20 to 25 years old, 25 to 30 years old, 30 to 35 years old, 35 to 40 years old, 40 to 45 years old, 45 to 50 years old, 50 to 55 years old, 55 to 60 years old, 60 to 65 years old, 65 to 70 years old, 70 to 75 years old, 75 to 80 years old, 80 to 85 years old, 85 to 90 years old, 90 to 95 years old or 95 to 100 years old. In some embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human infant. In other embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human toddler. In other embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human child. In other embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a human adult. In yet other embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to an elderly human.
  • In certain embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject in an immunocompromised state or immunosuppressed state or at risk for becoming immunocompromised or immunosuppressed. In certain embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject receiving or recovering from immunosuppressive therapy. In certain embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject that has or is at risk of getting cancer. In certain embodiments, the subject is, will or has undergone surgery, chemotherapy and/or radiation therapy. In certain embodiments, the patient has undergone surgery to remove the tumor or neoplasm. In specific embodiments, the patient is administered an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein following surgery to remove a tumor or neoplasm. In other embodiment, the patient is administered an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein prior to undergoing surgery to remove a tumor or neoplasm. In certain embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject that has, will have or had a tissue transplant, organ transplant or transfusion.
  • In some embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a patient who has proven refractory to therapies other than the chimeric NDV or composition thereof, oncolysate, whole cell vaccine, or a combination therapy but are no longer on these therapies. In a specific embodiment, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a patient who has proven refractory to chemotherapy. In one embodiment, that a cancer is refractory to a therapy means that at least some significant portion of the cancer cells are not killed or their cell division arrested. The determination of whether the cancer cells are refractory can be made either in vivo or in vitro by any method known in the art for assaying the effect of a therapy on cancer cells, using the art-accepted meanings of “refractory” in such a context. In a certain embodiment, refractory patient is a patient refractory to a standard therapy. In certain embodiments, a patient with cancer, is refractory to a therapy when the tumor or neoplasm has not significantly been eradicated and/or the symptoms have not been significantly alleviated. The determination of whether a patient is refractory can be made either in vivo or in vitro by any method known in the art for assaying the effectiveness of a treatment of cancer, using art-accepted meanings of “refractory” in such a context.
  • In certain embodiments, the patient to be treated in accordance with the methods described herein is a patient already being treated with antibiotics, anti-virals, anti-fungals, or other biological therapy/immunotherapy or anti-cancer therapy. Among these patients are refractory patients, and patients who are too young for conventional therapies. In some embodiments, the subject being administered an NDV (e.g., a chimeric NDV), an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein has not received therapy prior to the administration of the chimeric NDV or composition, the oncolysate vaccine, or the whole cell vaccine, or the combination therapy.
  • In some embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a patient to prevent the onset of cancer in a patient at risk of developing cancer. In some embodiments, compounds are administered to a patient who are susceptible to adverse reactions to conventional therapies.
  • In some embodiments, the subject being administered an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein has not received prior therapy. In other embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein is administered to a subject who has received a therapy prior to administration of the NDV (e.g., a chimeric NDV) or composition, the oncolysate vaccine, the whole cell vaccine, or the combination therapy. In some embodiments, the subject administered an NDV (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine described herein, or a whole cell vaccine described herein, or a combination therapy described herein experienced adverse side effects to a prior therapy or a prior therapy was discontinued due to unacceptable levels of toxicity to the subject.
  • 5.6.2. Dosage & Frequency
  • The amount of an NDV or a composition thereof, an oncolysate vaccine, or a whole cell vaccine which will be effective in the treatment of cancer will depend on the nature of the cancer, the route of administration, the general health of the subject, etc. and should be decided according to the judgment of a medical practitioner. Standard clinical techniques, such as in vitro assays, may optionally be employed to help identify optimal dosage ranges. However, suitable dosage ranges of an NDV for administration are generally about 102, 5×102, 103, 5×103, 104, 5×104, 105, 5×105, 106, 5×106, 107, 5×107, 108, 5×108, 1×109, 5×109, 1×101°, 5×1010, 1×1011, 5×1011 or 1012 pfu, and most preferably about 104 to about 1012, 106 to 1012, 108 to 1012, 109 to 1012 or 109 to 1011, and can be administered to a subject once, twice, three, four or more times with intervals as often as needed. Dosage ranges of oncolysate vaccines for administration may include 0.001 mg, 0.005 mg, 0.01 mg, 0.05 mg. 0.1 mg. 0.5 mg, 1.0 mg, 2.0 mg. 3.0 mg, 4.0 mg, 5.0 mg, 10.0 mg, 0.001 mg to 10.0 mg, 0.01 mg to 1.0 mg, 0.1 mg to 1 mg, and 0.1 mg to 5.0 mg, and can be administered to a subject once, twice, three or more times with intervals as often as needed. Dosage ranges of whole cell vaccines for administration may include 102, 5×102, 103, 5×103, 104, 5×104, 105, 5×105, 106, 5×106, 107, 5×107, 108, 5×108, 1×109, 5×109, 1×1010, 5×1010, 1×1011, 5×1011 or 1012 cells, and can be administered to a subject once, twice, three or more times with intervals as often as needed. In certain embodiments, dosages similar to those currently being used in clinical trials for NDV, oncolysate vaccines or whole cell vaccines are administered to a subject. Effective doses may be extrapolated from dose response curves derived from in vitro or animal model test systems.
  • In certain embodiments, an NDV (e.g., a chimeric NDV) or a composition thereof is administered to a subject as a single dose followed by a second dose 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 1 to 3 weeks, 1 to 2 weeks later. In accordance with these embodiments, booster inoculations may be administered to the subject at 6 to 12 month intervals following the second inoculation. In certain embodiments, an oncolysate vaccine or a whole cell vaccine is administered to a subject as a single dose followed by a second dose 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 1 to 3 weeks, 1 to 2 weeks later.
  • In certain embodiments, administration of the same NDV (e.g., chimeric NDV) or a composition thereof, oncolysate vaccine, or whole cell vaccine may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 6 says, 7 days, 10 days, 14 days, 15 days, 21 days, 28 days, 30 days, 45 days, 2 months, 75 days, 3 months, or at least 6 months. In other embodiments, administration of the same NDV (e.g., a NDV) or a composition thereof, oncolysate vaccine, or whole cell vaccine may be repeated and the administrations may be separated by 1 to 14 days, 1 to 7 days, 7 to 14 days, 1 to 30 days, 15 to 30 days, 15 to 45 days, 15 to 75 days, 15 to 90 days, 1 to 3 months, 3 to 6 months, 3 to 12 months, or 6 to 12 months. In some embodiments, a first NDV (e.g., a first chimeric NDV) or a composition thereof is administered to a subject followed by the administration of a second NDV (e.g., a second chimeric NDV) or a composition thereof. In certain embodiments, the first and second NDVs (e.g., the first and second chimeric NDVs) or compositions thereof may be separated by at least 1 day, 2 days, 3 days, 5 days, 6 days, 7 days, 10 days, 14 days, 15 days, 21 days, 28 days, 30 days, 45 days, 2 months, 75 days, 3 months, or at least 6 months. In other embodiments, the first and second NDVs (e.g., the first and second chimeric NDVs) or compositions thereof may be separated by 1 to 14 days, 1 to 7 days, 7 to 14 days, 1 to 30 days, 15 to 30 days, 15 to 45 days, 15 to 75 days, 15 to 90 days, 1 to 3 months, 3 to 6 months, 3 to 12 months, or 6 to 12 months.
  • In certain embodiments, an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject in combination with one or more additional therapies, such as a therapy described in Section 5.6.4, infra. The dosage of the other one or more additional therapies will depend upon various factors including, e.g., the therapy, the nature of the cancer, the route of administration, the general health of the subject, etc. and should be decided according to the judgment of a medical practitioner. In specific embodiments, the dose of the other therapy is the dose and/or frequency of administration of the therapy recommended for the therapy for use as a single agent is used in accordance with the methods disclosed herein. In other embodiments, the dose of the other therapy is a lower dose and/or less frequent administration of the therapy than recommended for the therapy for use as a single agent is used in accordance with the methods disclosed herein. Recommended doses for approved therapies can be found in the Physician's Desk Reference.
  • In certain embodiments, an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject concurrently with the administration of one or more additional therapies. In other embodiments, an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject every 3 to 7 days, 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 2 to 4 weeks, 1 to 3 weeks, or 1 to 2 weeks and one or more additional therapies (such as described in Section 5.6.4, infra) is administered every 3 to 7 days, 1 to 6 weeks, 1 to 5 weeks, 1 to 4 weeks, 1 to 3 weeks, or 1 to 2 weeks. In certain embodiments, an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject every 1 to 2 weeks and one or more additional therapies (such as described in Section 5.6.4, infra) is administered every 2 to 4 weeks. In some embodiments, an NDV or composition thereof, or oncolysate vaccine or whole cell vaccine is administered to a subject every week and one or more additional therapies (such as described in Section 5.6.4, infra) is administered every 2 weeks.
  • 5.6.3. Types of Cancer
  • Specific examples of cancers that can be treated in accordance with the methods described herein include, but are not limited to: leukemias, such as but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias, such as, myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia leukemias and myelodysplastic syndrome; chronic leukemias, such as but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as but not limited to Hodgkin's disease, non-Hodgkin's disease; multiple myelomas such as but not limited to smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, placancer cell leukemia, solitary placancercytoma and extramedullary placancercytoma; Waldenstrom's macroglobulinemia; monoclonal gammopathy of undetermined significance; benign monoclonal gammopathy; heavy chain disease; bone and connective tissue sarcomas such as but not limited to bone sarcoma, osteosarcoma, chondrosarcoma, Ewing's sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, neurilemmoma, rhabdomyosarcoma, synovial sarcoma; brain tumors such as but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, glioblastoma multiforme, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, primary brain lymphoma; breast cancer including but not limited to ductal carcinoma, adenocarcinoma, lobular (cancer cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, Paget's disease, and inflammatory breast cancer; adrenal cancer such as but not limited to pheochromocytom and adrenocortical carcinoma; thyroid cancer such as but not limited to papillary or follicular thyroid cancer, medullary thyroid cancer and anaplastic thyroid cancer; pancreatic cancer such as but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; pituitary cancers such as but limited to Cushing's disease, prolactin-secreting tumor, acromegaly, and diabetes insipidus; eye cancers such as but not limited to ocular melanoma such as iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma; vaginal cancers such as squamous cell carcinoma, adenocarcinoma, and melanoma; vulvar cancer such as squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget's disease; cervical cancers such as but not limited to, squamous cell carcinoma, and adenocarcinoma; uterine cancers such as but not limited to endometrial carcinoma and uterine sarcoma; ovarian cancers such as but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor; esophageal cancers such as but not limited to, squamous cancer, adenocarcinoma, adenoid cystic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, placancercytoma, verrucous carcinoma, and oat cell (cancer cell) carcinoma; stomach cancers such as but not limited to, adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; colon cancers; rectal cancers; liver cancers such as but not limited to hepatocellular carcinoma and hepatoblastoma; gallbladder cancers such as adenocarcinoma; cholangiocarcinomas such as but not limited to papillary, nodular, and diffuse; lung cancers such as non-cancer cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma and cancer-cell lung cancer; testicular cancers such as but not limited to germinal tumor, seminoma, anaplastic, classic (typical), spermatocytic, nonseminoma, embryonal carcinoma, teratoma carcinoma, choriocarcinoma (yolk-sac tumor), prostate cancers such as but not limited to, prostatic intraepithelial neoplasia, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; penal cancers; oral cancers such as but not limited to squamous cell carcinoma; basal cancers; salivary gland cancers such as but not limited to adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; pharynx cancers such as but not limited to squamous cell cancer, and verrucous; skin cancers such as but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, acral lentiginous melanoma; kidney cancers such as but not limited to renal cell carcinoma, adenocarcinoma, hypernephroma, fibrosarcoma, transitional cell cancer (renal pelvis and/or uterer); Wilms' tumor; bladder cancers such as but not limited to transitional cell carcinoma, squamous cell cancer, adenocarcinoma, carcinosarcoma. In addition, cancers include myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangioendotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma and papillary adenocarcinomas (for a review of such disorders, see Fishman et al., 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al., 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).
  • In a specific embodiment, the chimeric NDVs described herein or compositions thereof, an oncolysate vaccine described herein, a whole cell vaccine herein, or a combination therapy described herein are useful in the treatment of a variety of cancers and abnormal proliferative diseases, including (but not limited to) the following: carcinoma, including that of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid and skin; including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T cell lymphoma, Burkitt's lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyoscarcoma; other tumors, including melanoma, seminoma, teratocarcinoma, neuroblastoma and glioma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; tumors of mesenchymal origin, including fibrosarcoma, rhabdomyoscarama, and osteosarcoma; and other tumors, including melanoma, xeroderma pigmentosum, keratoactanthoma, seminoma, thyroid follicular cancer and teratocarcinoma.
  • In some embodiments, cancers associated with aberrations in apoptosis are treated in accordance with the methods described herein. Such cancers may include, but are not limited to, follicular lymphomas, carcinomas with p53 mutations, hormone dependent tumors of the breast, prostate and ovary, and precancerous lesions such as familial adenomatous polyposis, and myelodysplastic syndromes. In specific embodiments, malignancy or dysproliferative changes (such as metaplasias and dysplasias), or hyperproliferative disorders of the skin, lung, liver, bone, brain, stomach, colon, breast, prostate, bladder, kidney, pancreas, ovary, and/or uterus are treated in accordance with the methods described herein. In other specific embodiments, a sarcoma or melanoma is treated in accordance with the methods described herein.
  • In a specific embodiment, the cancer being treated in accordance with the methods described herein is leukemia, lymphoma or myeloma (e.g., multiple myeloma). Specific examples of leukemias and other blood-borne cancers that can be treated in accordance with the methods described herein include, but are not limited to, acute lymphoblastic leukemia “ALL”, acute lymphoblastic B-cell leukemia, acute lymphoblastic T-cell leukemia, acute myeloblastic leukemia “AML”, acute promyelocytic leukemia “APL”, acute monoblastic leukemia, acute erythroleukemic leukemia, acute megakaryoblastic leukemia, acute myelomonocytic leukemia, acute nonlymphocyctic leukemia, acute undifferentiated leukemia, chronic myelocytic leukemia “CIVIL”, chronic lymphocytic leukemia “CLL”, and hairy cell leukemia.
  • Specific examples of lymphomas that can be treated in accordance with the methods described herein include, but are not limited to, Hodgkin's disease, non-Hodgkin's Lymphoma, Multiple myeloma, Waldenstrom's macroglobulinemia, Heavy chain disease, and Polycythemia vera.
  • In another embodiment, the cancer being treated in accordance with the methods described herein is a solid tumor. Examples of solid tumors that can be treated in accordance with the methods described herein include, but are not limited to fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, colorectal cancer, kidney cancer, pancreatic cancer, bone cancer, breast cancer, ovarian cancer, prostate cancer, esophageal cancer, stomach cancer, oral cancer, nasal cancer, throat cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, uterine cancer, testicular cancer, cancer cell lung carcinoma, bladder carcinoma, lung cancer, epithelial carcinoma, glioma, glioblastoma multiforme, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, skin cancer, melanoma, neuroblastoma, and retinoblastoma. In another embodiment, the cancer being treated in accordance with the methods described herein is a metastatic. In another embodiment, the cancer being treated in accordance with the methods described herein is malignant.
  • In a specific embodiment, the cancer being treated in accordance with the methods described herein is a cancer that has a poor prognosis and/or has a poor response to conventional therapies, such as chemotherapy and radiation. In another specific embodiment, the cancer being treated in accordance with the methods described herein is malignant melanoma, malignant glioma, renal cell carcinoma, pancreatic adenocarcinoma, malignant pleural mesothelioma, lung adenocarcinoma, lung small cell carcinoma, lung squamous cell carcinoma, anaplastic thyroid cancer, and head and neck squamous cell carcinoma.
  • 5.6.4. Additional Therapies
  • Additional therapies that can be used in a combination with an NDV described herein or a composition thereof, an oncolysate vaccine, or a whole cell vaccine for the treatment of cancer include, but are not limited to, small molecules, synthetic drugs, peptides (including cyclic peptides), polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides or peptides), antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules. In a specific embodiment, the additional therapy is a chemotherapeutic agent.
  • In some embodiments, an NDV described herein or a composition thereof, an oncolysate vaccine, or a whole cell vaccine is used in combination with radiation therapy comprising the use of x-rays, gamma rays and other sources of radiation to destroy cancer cells. In specific embodiments, the radiation therapy is administered as external beam radiation or teletherapy, wherein the radiation is directed from a remote source. In other embodiments, the radiation therapy is administered as internal therapy or brachytherapy wherein a radioactive source is placed inside the body close to cancer cells and/or a tumor mass.
  • Currently available cancer therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the Physician's Desk Reference (67th ed., 2013).
  • Specific examples of anti-cancer agents that may be used in combination with an NDV described herein or a composition thereof include: hormonal agents (e.g., aromatase inhibitor, selective estrogen receptor modulator (SERM), and estrogen receptor antagonist), chemotherapeutic agents (e.g., microtubule disassembly blocker, antimetabolite, topoisomerase inhibitor, and DNA crosslinker or damaging agent), anti-angiogenic agents (e.g., VEGF antagonist, receptor antagonist, integrin antagonist, vascular targeting agent (VTA)/vascular disrupting agent (VDA)), radiation therapy, and conventional surgery.
  • Non-limiting examples of hormonal agents that may be used in combination with an NDV described herein or a composition thereof include aromatase inhibitors, SERMs, and estrogen receptor antagonists. Hormonal agents that are aromatase inhibitors may be steroidal or nonsteroidal. Non-limiting examples of nonsteroidal hormonal agents include letrozole, anastrozole, aminoglutethimide, fadrozole, and vorozole. Non-limiting examples of steroidal hormonal agents include aromasin (exemestane), formestane, and testolactone. Non-limiting examples of hormonal agents that are SERMs include tamoxifen (branded/marketed as Nolvadex®), afimoxifene, arzoxifene, bazedoxifene, clomifene, femarelle, lasofoxifene, ormeloxifene, raloxifene, and toremifene. Non-limiting examples of hormonal agents that are estrogen receptor antagonists include fulvestrant. Other hormonal agents include but are not limited to abiraterone and lonaprisan.
  • Non-limiting examples of chemotherapeutic agents that may be used in combination with an NDV described herein or a composition thereof, an oncolysate vaccine, or a whole cell vaccine include microtubule disasssembly blocker, antimetabolite, topoisomerase inhibitor, and DNA crosslinker or damaging agent. Chemotherapeutic agents that are microtubule disassembly blockers include, but are not limited to, taxenes (e.g., paclitaxel (branded/marketed as TAXOL®), docetaxel, abraxane, larotaxel, ortataxel, and tesetaxel); epothilones (e.g., ixabepilone); and vinca alkaloids (e.g., vinorelbine, vinblastine, vindesine, and vincristine (branded/marketed as ONCOVIN®)).
  • Chemotherapeutic agents that are antimetabolites include, but are not limited to, folate antimetabolites (e.g., methotrexate, aminopterin, pemetrexed, raltitrexed); purine antimetabolites (e.g., cladribine, clofarabine, fludarabine, mercaptopurine, pentostatin, thioguanine); pyrimidine antimetabolites (e.g., 5-fluorouracil, capecitabine, gemcitabine) (GEMZAR®, cytarabine, decitabine, floxuridine, tegafur); and deoxyribonucleotide antimetabolites (e.g., hydroxyurea).
  • Chemotherapeutic agents that are topoisomerase inhibitors include, but are not limited to, class I (camptotheca) topoisomerase inhibitors (e.g., topotecan (branded/marketed as HYCAMTIN®) irinotecan, rubitecan, and belotecan); class II (podophyllum) topoisomerase inhibitors (e.g., etoposide or VP-16, and teniposide); anthracyclines (e.g., doxorubicin, epirubicin, Doxil, aclarubicin, amrubicin, daunorubicin, idarubicin, pirarubicin, valrubicin, and zorubicin); and anthracenediones (e.g., mitoxantrone, and pixantrone).
  • Chemotherapeutic agents that are DNA crosslinkers (or DNA damaging agents) include, but are not limited to, alkylating agents (e.g., cyclophosphamide, mechlorethamine, ifosfamide (branded/marketed as IFEX®), trofosfamide, chlorambucil, melphalan, prednimustine, bendamustine, uramustine, estramustine, carmustine (branded/marketed as BiCNU®), lomustine, semustine, fotemustine, nimustine, ranimustine, streptozocin, busulfan, mannosulfan, treosulfan, carboquone, N,N′N′-triethylenethiophosphoramide, triaziquone, triethylenemelamine); alkylating-like agents (e.g., carboplatin (branded/marketed as PARAPLATIN®), cisplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, satraplatin, picoplatin); nonclassical DNA crosslinkers (e.g., procarbazine, dacarbazine, temozolomide (branded/marketed as TEMODAR®), altretamine, mitobronitol); and intercalating agents (e.g., actinomycin, bleomycin, mitomycin, and plicamycin).
  • 5.6.4.1 Immune Modulators
  • In specific embodiments, an NDV described herein (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the following: any agonist of a co-stimulatory signal of an immune cell (such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage) and/or any antagonist of an inhibitory signal of an immune cell (such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage), known to one of skill in the art. In particular embodiments, an NDV described herein (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the agonists of a co-stimulatory signal of an immune cell described in Section 5.2.1, supra. In some embodiments, an NDV described herein (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the antagonists of an inhibitory signal of an immune cell described in Section 5.2.1, supra. In certain embodiments, an NDV described herein (e.g., a chimeric NDV) or a composition thereof, an oncolysate vaccine, or a whole cell vaccine are administered to a subject in combination with one or more of the agonists of a co-stimulatory signal of an immune cell and/or one or more of the antagonists of an inhibitory signal of an immune cell described in Section 6, infra (e.g., an anti-CTLA-4 antibody or an ICOS-L)
  • 5.7 Biological Assays In Vitro Viral Assays
  • Viral assays include those that measure altered viral replication (as determined, e.g., by plaque formation) or the production of viral proteins (as determined, e.g., by western blot analysis) or viral RNAs (as determined, e.g., by RT-PCR or northern blot analysis) in cultured cells in vitro using methods which are well known in the art.
  • Growth of the NDVs described herein can be assessed by any method known in the art or described herein (e.g., in cell culture (e.g., cultures of chicken embryonic kidney cells or cultures of chicken embryonic fibroblasts (CEF)). Viral titer may be determined by inoculating serial dilutions of a NDV described herein into cell cultures (e.g., CEF, MDCK, EFK-2 cells, Vero cells, primary human umbilical vein endothelial cells (HUVEC), H292 human epithelial cell line or HeLa cells), chick embryos, or live animals (e.g., avians). After incubation of the virus for a specified time, the virus is isolated using standard methods. Physical quantitation of the virus titer can be performed using PCR applied to viral supernatants (Quinn & Trevor, 1997; Morgan et al., 1990), hemagglutination assays, tissue culture infectious doses (TCID50) or egg infectious doses (EID50). An exemplary method of assessing viral titer is described in Section 6, below.
  • Incorporation of nucleotide sequences encoding a heterologous peptide or protein (e.g., a cytokine, a mutated F protein, a mutated V protein, or miRNA target site into the genome of a chimeric NDV described herein can be assessed by any method known in the art or described herein (e.g., in cell culture, an animal model or viral culture in embryonated eggs). For example, viral particles from cell culture of the allantoic fluid of embryonated eggs can be purified by centrifugation through a sucrose cushion and subsequently analyzed for fusion protein expression by Western blotting using methods well known in the art.
  • Immunofluorescence-based approaches may also be used to detect virus and assess viral growth. Such approaches are well known to those of skill in the art, e.g., fluorescence microscopy and flow cytometry (see Section 6, infra).
  • Antibody Assays
  • Antibodies generated by the NDVs described herein may be characterized in a variety of ways well-known to one of skill in the art (e.g., ELISA, Surface Plasmon resonance display (BIAcore), Western blot, immunofluorescence, immunostaining and/or microneutralization assays). In particular, antibodies generated by the chimeric NDVs described herein may be assayed for the ability to specifically bind to an antigen of the virus or a heterologous peptide or protein. Such an assay may be performed in solution (e.g., Houghten, 1992, Bio/Techniques 13:412 421), on beads (Lam, 1991, Nature 354:82 84), on chips (Fodor, 1993, Nature 364:555 556), on bacteria (U.S. Pat. No. 5,223,409), on spores (U.S. Pat. Nos. 5,571,698; 5,403,484; and 5,223,409), on plasmids (Cull et al., 1992, Proc. Natl. Acad. Sci. USA 89:1865 1869) or on phage (Scott and Smith, 1990, Science 249:386 390; Cwirla et al., 1990, Proc. Natl. Acad. Sci. USA 87:6378 6382; and Felici, 1991, J. Mol. Biol. 222:301 310) (each of these references is incorporated herein in its entirety by reference).
  • Antibodies generated by the chimeric NDVs described herein that have been identified to specifically bind to an antigen of the virus or a heterologous peptide or protein can be assayed for their specificity to said antigen of the virus or heterologous peptide or protein. The antibodies may be assayed for specific binding to an antigen of the virus or a heterologous peptide or protein and for their cross-reactivity with other antigens by any method known in the art. Immunoassays which can be used to analyze specific binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al., eds., 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety).
  • The binding affinity of an antibody to an antigen and the off-rate of an antibody-antigen interaction can be determined by competitive binding assays. Alternatively, a surface plasmon resonance assay (e.g., BIAcore kinetic analysis) or KinExA assay (Blake, et al., Analytical Biochem., 1999, 272:123-134) may be used to determine the binding on and off rates of antibodies to an antigen of the chimeric NDVs described herein.
  • IFN Assays
  • IFN induction and release by an NDV described herein may be determined using techniques known to one of skill in the art or described herein. For example, the amount of IFN induced in cells following infection with an NDV described herein may be determined using an immunoassay (e.g., an ELISA or Western blot assay) to measure IFN expression or to measure the expression of a protein whose expression is induced by IFN. Alternatively, the amount of IFN induced may be measured at the RNA level by assays, such as Northern blots and quantitative RT-PCR, known to one of skill in the art. In specific embodiments, the amount of IFN released may be measured using an ELISPOT assay. (See, e.g., the methods described in Section 6, below.)
  • Activation Marker Assays
  • Techniques for assessing the expression of activation marker, co-stimulatory molecule, ligand, or inhibitory molecule by immune cells are known to one of skill in the art. For example, the expression of an activation marker, co-stimulatory molecule, ligand, or inhibitory molecule by an immune cell (e.g., T lymphocyte or NK cell) can be assessed by flow cytometry. In a specific embodiment, techniques described in Section 6, infra, are used to assess the expression of an activation marker, co-stimulatory molecule, ligand, or inhibitory molecule by an immune cell.
  • Immune Cell Infiltration Assays
  • Techniques for assessing immune cell infiltration are known to one of skill in the art. In a specific embodiment, techniques described in Section 6, infra, are used to assess immune cell infiltration.
  • Toxicity Studies
  • In some embodiments, the NDVs described herein or compositions thereof, oncolysate vaccines described herein, whole cell vaccines described herein, or combination therapies described herein are tested for cytotoxicity in mammalian, preferably human, cell lines (see, e.g., the cytotoxicity assay described in Section 6, infra). In certain embodiments, cytotoxicity is assessed in one or more of the following non-limiting examples of cell lines: U937, a human monocyte cell line; primary peripheral blood mononuclear cells (PBMC); Huh7, a human hepatoblastoma cell line; HL60 cells, HT1080, HEK 293T and 293H, MLPC cells, human embryonic kidney cell lines; human melanoma cell lines, such as SkMe12, SkMe1-119 and SkMe1-197; THP-1, monocytic cells; a HeLa cell line; and neuroblastoma cells lines, such as MC-IXC, SK-N-MC, SK-N-MC, SK-N-DZ, SH-SY5Y, and BE(2)-C. In certain embodiments, cytotoxicity is assessed in various cancer cells. In some embodiments, the ToxLite assay is used to assess cytotoxicity.
  • Many assays well-known in the art can be used to assess viability of cells or cell lines following infection with an NDV described herein or composition thereof, or treatment with an oncolysate vaccine described herein, a whole cell vaccine described herein, or a combination therapy described herein and, thus, determine the cytotoxicity of the NDV or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy. For example, cell proliferation can be assayed by measuring Bromodeoxyuridine (BrdU) incorporation, (3H) thymidine incorporation, by direct cell count, or by detecting changes in transcription, translation or activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers (Rb, cdc2, cyclin A, D1, D2, D3, E, etc). The levels of such protein and mRNA and activity can be determined by any method well known in the art. For example, protein can be quantitated by known immunodiagnostic methods such as ELISA, Western blotting or immunoprecipitation using antibodies, including commercially available antibodies. mRNA can be quantitated using methods that are well known and routine in the art, for example, using northern analysis, RNase protection, or polymerase chain reaction in connection with reverse transcription. Cell viability can be assessed by using trypan-blue staining or other cell death or viability markers known in the art. In a specific embodiment, the level of cellular ATP is measured to determined cell viability. In preferred embodiments, an NDV described herein or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy kills cancer cells but does not kill healthy (i.e., non-cancerous) cells. In one embodiment, an NDV described herein or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy preferentially kills cancer cells but does not kill healthy (i.e., non-cancerous) cells.
  • In specific embodiments, cell viability is measured in three-day and seven-day periods using an assay standard in the art, such as the CellTiter-Glo Assay Kit (Promega) which measures levels of intracellular ATP. A reduction in cellular ATP is indicative of a cytotoxic effect. In another specific embodiment, cell viability can be measured in the neutral red uptake assay. In other embodiments, visual observation for morphological changes may include enlargement, granularity, cells with ragged edges, a filmy appearance, rounding, detachment from the surface of the well, or other changes.
  • The NDVs described herein or compositions thereof, oncolysate vaccines, whole cell vaccines or combination therapies can be tested for in vivo toxicity in animal models (see, e.g., the animal models described in Section 6, below). For example, animal models, described herein and/or others known in the art, used to test the effects of compounds on cancer can also be used to determine the in vivo toxicity of the NDVs described herein or compositions thereof, oncolysate vaccines, whole cell vaccines, or combination therapies. For example, animals are administered a range of pfu of an NDV described herein (e.g., a chimeric NDV described in Section 5.2, infra). Subsequently, the animals are monitored over time for lethality, weight loss or failure to gain weight, and/or levels of serum markers that may be indicative of tissue damage (e.g., creatine phosphokinase level as an indicator of general tissue damage, level of glutamic oxalic acid transaminase or pyruvic acid transaminase as indicators for possible liver damage). These in vivo assays may also be adapted to test the toxicity of various administration mode and/or regimen in addition to dosages.
  • The toxicity and/or efficacy of an NDV described herein or a composition thereof, an oncolysate vaccine described herein, a whole cell vaccine described herein, or a combination therapy described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Therapies that exhibits large therapeutic indices are preferred. While therapies that exhibits toxic side effects may be used, care should be taken to design a delivery system that targets such therapies to the site of affected tissue in order to minimize potential damage to noncancerous cells and, thereby, reduce side effects.
  • The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the therapies for use in subjects. The dosage of such agents lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any therapy described herein, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the chimeric NDV that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in subjects. Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • Anti-Cancer Studies
  • The NDVs described herein or compositions thereof, oncolysate vaccines described herein, whole cell vaccines described herein, or combination therapies described herein can be tested for biological activity using animal models for cancer. Such animal model systems include, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, dogs, rabbits, etc. In a specific embodiment, the anti-cancer activity of an NDV described herein or combination therapy is tested in a mouse model system. Such model systems are widely used and well-known to the skilled artisan such as the SCID mouse model or transgenic mice.
  • The anti-cancer activity of an NDV described herein or a composition thereof, oncolysate vaccine described herein, whole cell vaccine described herein, or a combination therapy described herein can be determined by administering the NDV or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy to an animal model and verifying that the NDV or composition thereof, oncolysate vaccine, whole cell vaccine, or combination therapy is effective in reducing the severity of cancer, reducing the symptoms of cancer, reducing cancer metastasis, and/or reducing the size of a tumor in said animal model (see, e.g., Section 6, below). Examples of animal models for cancer in general include, include, but are not limited to, spontaneously occurring tumors of companion animals (see, e.g., Vail & MacEwen, 2000, Cancer Invest 18(8):781-92). Examples of animal models for lung cancer include, but are not limited to, lung cancer animal models described by Zhang & Roth (1994, In-vivo 8(5):755-69) and a transgenic mouse model with disrupted p53 function (see, e.g. Morris et al., 1998, J La State Med Soc 150(4): 179-85). An example of an animal model for breast cancer includes, but is not limited to, a transgenic mouse that over expresses cyclin D1 (see, e.g., Hosokawa et al., 2001, Transgenic Res 10(5):471-8). An example of an animal model for colon cancer includes, but is not limited to, a TCR b and p53 double knockout mouse (see, e.g., Kado et al., 2001, Cancer Res. 61(6):2395-8). Examples of animal models for pancreatic cancer include, but are not limited to, a metastatic model of PancO2 murine pancreatic adenocarcinoma (see, e.g., Wang et al., 2001, Int. J. Pancreatol. 29(1):37-46) and nu-nu mice generated in subcutaneous pancreatic tumors (see, e.g., Ghaneh et al., 2001, Gene Ther. 8(3):199-208). Examples of animal models for non-Hodgkin's lymphoma include, but are not limited to, a severe combined immunodeficiency (“SCID”) mouse (see, e.g., Bryant et al., 2000, Lab Invest 80(4):553-73) and an IgHmu-HOX11 transgenic mouse (see, e.g., Hough et al., 1998, Proc. Natl. Acad. Sci. USA 95(23):13853-8). An example of an animal model for esophageal cancer includes, but is not limited to, a mouse transgenic for the human papillomavirus type 16 E7 oncogene (see, e.g., Herber et al., 1996, J. Virol. 70(3):1873-81). Examples of animal models for colorectal carcinomas include, but are not limited to, Apc mouse models (see, e.g., Fodde & Smits, 2001, Trends Mol Med 7(8):369 73 and Kuraguchi et al., 2000). In a specific embodiment, the animal models for cancer described in Section 6, infra, are used to assess efficacy of an NDV or composition thereof, an oncolysate, a whole cell vaccine, or a combination therapy.
  • 6. EXAMPLE
  • This example demonstrates the therapeutic efficacy of NDV therapy in combination with immune checkpoint modulators that are immunostimulatory in the treatment of cancer.
  • 6.1 Materials & Methods Mice
  • BALB/c mice (6-8 weeks old), and WT C57BL/6 mice were purchased from Jackson Laboratory. All mice were maintained in microisolator cages and treated in accordance with the NIH and American Association of Laboratory Animal Care regulations. All mouse procedures and experiments for this study were approved by the Memorial Sloan-Kettering Cancer Center Institutional Animal Care and Use Committee.
  • Cell Lines
  • The murine cancer cell lines for melanoma (B16-F10), and colon carcinoma (CT26 and MC38) were maintained in RPMI medium supplemented with 10% fetal calf serum and penicillin with streptomycin. The murine prostate cancer cell line TRAMP-C2 was maintained in DMEM medium supplemented with 5% fetal calf serum (FCS; Mediatech, Inc.). 5% Nu Serum IV (BD Biosciences) HEPES, 2-ME, pen/strep, L-glut, 5 μg/mL insulin (Sigma), and 10 nmol/L DHT (Sigma).
  • Antibodies
  • Therapeutic anti-CTLA-4 (clone 9H10), anti-PD-1 (clone RMP1-14), and anti-PD-L1 monoclonal antibodies were produced by BioXcell. Antibodies used for flow cytometry were purchased from eBioscience, Biolegend, Invitrogen, and BD Pharmingen.
  • Viruses and Cloning
  • Recombinant lentogenic NDV LaSota strain was used for all experiments. To generate NDV virus expressing murine ICOSL, a DNA fragment encoding the murine ICOSL flanked by the appropriate NDV-specific RNA transcriptional signals was inserted into the SacII site created between the P and M genes of pT7NDV/LS. Viruses were rescued from cDNA using methods described previously and sequenced by reverse transcription PCR for insert fidelity. Virus titers were determined by serial dilution and immunofluorescence in Vero cells. Recombinant ICOSL-F fusion construct was generated by PCR amplification of the ICOSL DNA encoding the extracellular domain (amino acids 1-277) with flanking EcoRI and MluI restriction sites, and the NDV F DNA encoding the F transmembrane and intracellular domains (amino acids 501-554) with flanking MluI and XhoI restriction sites. The resultant DNA fragments were assembled in pCAGGS vector utilizing 3-part ligation.
  • In Vitro Infection Experiments
  • For evaluation of upregulation of surface MHC-I, MHC-II, and ICAM-1 by NDV, and for evaluation of surface expression of the ICOSL transgene from the NDV-ICOSL virus, B16-F10 cells were infected in 6-well dishes at MOI 2 in triplicate. Twenty-four hours later, the cells were harvested by mechanical scraping and processed for surface labeling and quantification by flow cytometry. For virus growth curve experiments, B16-F10 cells were incubated at room temperature with the virus in 6-well culture dishes at the indicated MOIs in a total volume of 100 μl. One hour after the incubation, the infection media was aspirated and the cells were incubated at 37° C. in 1 ml of DMEM with 10% chick allantoic fluid. After 24, 48, and 72 hours, the supernatants were collected and virus titers were determined as above. For in vitro cytotoxicity experiments, the infections were carried out in a similar fashion. At 24, 48, 72, and 96 hours post infection the cells were washed and incubated with 1% Triton X-100 at 37° C. for 30 minutes. LDH activity in the lysates was determined using the Promega CytoTox 96 assay kit, according to the manufacturer's instructions.
  • Tumor Challenge Survival Experiments.
  • Bilateral flank tumor models were established to monitor for therapeutic efficacy in both injected and systemic tumors. Treatment schedules and cell doses were established for each tumor model to achieve 10-20% tumor clearance by NDV or anti-CTLA-4/anti-PD-1 as single agents. For experiments evaluating combination therapy of wild-type NDV (NDV-WT) with immune checkpoint blockade, B16F10 tumors were implanted by injection of 2×105B16F10 cells in the right flank i.d. on day 0 and 5×104 cells in the left flank on day 4. On days 7, 10, 13, and 16 the mice were treated with 4 intratumoral injections of 2×107 pfu of NDV in PBS in a total volume of 100 μl. Concurrently, on days 7, 10, 13, and 16 the mice received 4 i.p. injections of anti-CTLA-4 antibody (100 μg) or anti-PD-1 antibody (250 μg). Control groups received a corresponding dose of isotype antibody i.p. and intratumoral injection of PBS. Tumor size and incidence were monitored over time by measurement with a caliper.
  • For the TRAMP-C2 model, 5×105 cells were implanted in right flank on day 0 and 5×105 cells were implanted in the left flank on day 8. Treatment was performed on days 11, 14, 17, and 20 in the similar fashion to above.
  • For experiments evaluating recombinant NDV expressing ICOSL (NDV-ICOSL), B16F10 tumors were implanted by injection of 2×105 B16F10 cells in the right flank i.d. on day 0 and 1×105 cells in the left flank on day 4. Treatment was carried out as above.
  • For the CT26 model, tumors were implanted by injection of 1×106 CT26 cells in the right flank i.d. on day 0 and 1×106 cells in the left flank on day 2. Treatment was carried out as above on days 6, 9, and 12.
  • Isolation of Tumor-Infiltrating Lymphocytes
  • B16F10 tumors were implanted by injection of 2×105 B16F10 cells in the right flank i.d. on day 0 and 2×105 cells in the left flank on day 4. On days 7, 10, and 13 the mice were treated with 3 intratumoral injections of 2×107 pfu of NDV, and 100 μg of i.p. anti-CTLA-4 antibody or 250 μg of i.p. anti-PD-1 antibody, where specified. On day 15, mice were sacrificed by CO2 inhalation. Tumors and tumor-draining lymph nodes were removed using forceps and surgical scissors and weighed. Tumors from each group were minced with scissors prior to incubation with 1.67 Wünsch 1J/int, Liberase and 0.2 mg/mL DNase for 30 minutes at 37° C. Tumors were homogenized by repeated pipetting and filtered through a 70-μm nylon filter. Cell suspensions were washed once with complete RPMI and purified on a Ficoll gradient to eliminate dead cells. Cells from tumor draining lymph nodes were isolated by grinding the lymph nodes through a 70-μm nylon filter.
  • Flow Cytometry
  • Cells isolated from tumors or tumor-draining lymph nodes were processed for surface labeling with several antibody panels staining CD45, CD3, CD4, CD8, CD44, PD-1, ICOS, CD11c, CD19, NK1.1, CD11b, F4/80, Ly6C and Ly6G. Fixable viability dye eFluor780 (eBioscience) was used to distinguish the live cells. Cells were further permeabilized using FoxP3 fixation and permeabilization kit (eBioscience) and stained for Ki-67, FoxP3, Granzyme B, CTLA-4, and IFN gamma. Data was acquired using the LSRII Flow cytometer (BD Biosciences) and analyzed using FlowJo software (Treestar).
  • DC Purification and Loading
  • Spleens from naïve mice were isolated and digested with 1.67 Wunsch U/mL Liberase and 0.2 mg/mL DNase for 30 minutes at 37° C. The resulting cell suspensions were filtered through 70 μm nylon filter and washed once with complete RPMI. CD11c+ dendritic cells were purified by positive selection using Miltenyi magnetic beads. Isolated dendritic cells were cultured overnight with recombinant GM-CSF and B16-F10 tumor lysates and were purified on Ficoll gradient.
  • Analysis of Cytokine Production
  • Cell suspensions from tumors or tumor-draining lymph nodes were pooled and enriched for T cells using a Miltenyi T-cell purification kit. Isolated T cells were counted and co-cultured for 8 hours with dendritic cells loaded with B16-F10 tumor cell lysates in the presence of 20 U/ml IL-2 (R and D) plus Brefeldin A (BD Bioscience). After restimulation, lymphocytes were processed for flow cytometry as above.
  • Statistics.
  • Data were analyzed by 2-tailed Student's t test, and P<0.05 was considered statistically significant.
  • 6.2 Results
  • In order to characterize the anti-tumor immune response induced by Newcastle disease virus (NDV) infection, the expression of MHC I and MHC II molecules as well as ICAM-1 on the surface of in vitro infected cells was assessed. As shown in FIG. 1, NDV infection in B16 melanoma cells induces upregulation of MHC class I and II molecules as well as adhesion molecule ICAM-1, all of which are thought to be important for recruitment of tumor-specific lymphocytes and activation of anti-tumor immune response. Next, the anti-tumor immune response induced by NDV infection in vivo was assessed in a murine melanoma model and an established 2-flank model that allowed us to monitor responses both in the virus-injected tumors as well as distant tumors which do not receive the virus. As shown in FIG. 2, the virus-infected tumors show dramatic infiltration with immune cells such as NK cells, macrophages, and CD8 and CD4 cells, but not regulatory T cells. Since part of this immune response could be a response to virus, rather than tumor, the immune response with respect to contralateral tumors was assessed (FIG. 3). Interestingly, these tumors demonstrated a similar degree of increased CD8 and CD4 effector, but not T reg infiltrate. Analysis of these cells revealed that they upregulate activation, proliferation, and lytic markers (FIG. 4). NDV monotherapy was effective in controlling the treated tumors (FIG. 5A), but only marginally slowed down the growth of the contralateral tumors (FIG. 5B). Mice that cleared the tumors, however, demonstrated some degree of protection against further tumor challenge (FIG. 5D), suggesting that NDV therapy can induce a lasting immunity.
  • Next, it was assessed whether additional mechanisms could be targeted to enhance the anti-tumor effect generated by NDV. Characterization of tumor-infiltrating lymphocytes from both NDV-injected and non-injected tumors revealed upregulation of the inhibitory receptor CTLA-4 on lymphocytes (FIG. 6). It was then assessed whether inhibition of the CTLA-4 receptor could result in a better therapeutic efficacy of NDV. Strikingly, combination therapy resulted in rejection in bilateral tumors in the majority of the animals, an effect that was not seen with either treatment alone (FIG. 7). This effect was present even when the prostate adenocarcinoma TRAMP model was used, which is not susceptible to viral infection (FIG. 8), suggesting that the minimal viral replication and the resultant inflammatory response were sufficient for generation of protective anti-tumor immunity.
  • To determine whether targeting other immune checkpoints in combination with NDV therapy could be beneficial, the effect on the PD-1-PD-L1 pathway following NDV infection was assessed. As shown in FIG. 9, NDV infected tumor cells both in vitro and in vivo had upregulated the expression of the inhibitory PD-L1 ligand on the surface of the cells. This effect was not just a result of a direct virus infection, but was also seen when non-infected cells were treated with UV-inactivated supernatants from the virus infected cells (FIG. 9B) and in contralateral, noninfected, tumors (FIG. 9C). This prompted testing combination therapy with NDV and anti-PD-1 antibody. Similar to CTLA-4 blockade, NDV therapy in combination with anti-PD-1 in the aggressive B16 melanoma model resulted in cures in the majority of animals, an effect that was associated with increased tumor infiltration with activated effector lymphocytes (FIG. 10).
  • Throughout the studies conducted, the therapeutic efficacy of a combination therapy decreased when larger tumor challenge was used. Next, activation markers that could predict a better response and could be targeted for further improvement in therapeutic efficacy were assessed. Analysis of lymphocytes isolated from the tumors and tumor-draining lymph nodes identified upregulation of the co-stimulatory molecule ICOS as one of the activation markers in the treated animals (FIG. 11). ICOS upregulation has been previously been shown to be associated with more durable therapeutic responses and increased survival in patients treated with anti-CTLA-4 therapy for malignant melanoma. It was assessed whether intratumoral expression of the ICOS ligand (ICOSL) could further boost the therapeutic response of combination therapy. Using reverse-genetics system for NDV, NDV expressing murine ICOSL (NDV-ICOSL) were generated. In vitro characterization of the virus revealed that it had similar replicative and lytic properties to the parental NDV strain (FIG. 12). When tested in vivo, however, with a larger B16 tumor challenge, NDV-ICOSL demonstrated significant advantage over the parental NDV virus when used in combination with CTLA-4 blockade, with long-term survival in the majority of treated animals (FIG. 13). This effect was not limited to B16 melanoma and was demonstrated for CT26 colon carcinoma in the Balb/C mouse strain, suggesting that this therapeutic strategy could be translatable to different tumor types (FIG. 14). Analysis of B16 tumors from the treated animals demonstrated significant infiltration with different immune cell subtypes with upregulation of the activation markers (FIGS. 15 and 16). These lymphocytes were tumor-specific and demonstrated secretion of IFN gamma in response to stimulation with dendritic cells loaded with tumor lysates (FIG. 17). Finally, animals that were cured of their B16 or CT26 tumors were re-challenged with tumor cells and demonstrated complete protection against tumor re-challenge (FIG. 18).
  • To further improve the expression of the ICOSL in the tumor and to incorporate the ligand into the virion, a chimeric protein consisting of the extracellular domain of the ICOSL (amino acids 1-277) and the transmembrane and intracellular domains of the NDV F protein (amino acids 501-554) was generated (FIG. 19A). Transfection of the resultant construct into B16-F10 cells resulted in increased expression of the chimeric ICOSL-F ligand on the surface of the transfected cells, when compared to the transfected native ICOSL, suggesting that the regulatory mechanisms governing the transport of NDV F protein to the surface can be utilized to increase the surface expression of immune stimulatory ligands.
  • Overall, these studies demonstrate that 1) combination of NDV with immune checkpoint regulatory antibodies can be used as a strategy to circumvent the limitation of both oncolytic virus therapy and antibody therapy; and 2) expression of immunostimulatory ligands by NDV can further improve the therapeutic efficacy of the virus, especially when used in combination with immunoregulatory antibodies. These findings have clinical application.
  • The invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims.
  • All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

Claims (31)

1.-28. (canceled)
29. A method for treating cancer, comprising administering to a subject in need thereof a Newcastle disease virus (NDV) and an agonist of a co-stimulatory receptor of an immune cell, wherein the co-stimulatory receptor is glucocorticoid-induced tumor necrosis factor receptor (GITR), inducible T-cell costimulator (ICOS), OX40, CD27, 4-1BB, CD226, cytotoxic and regulatory T cell molecule (CRTAM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), B cell maturation protein (BCMA), LIGHT, or LT alpha.
30. A method for treating cancer, comprising administering to a subject in need thereof an NDV and an antagonist of an inhibitory receptor of an immune cell, wherein the inhibitory receptor is programmed cell death protein 1 (PD1), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD160, or adenosine A2a receptor (A2aR).
31. A method for treating cancer, comprising administering to a subject in need thereof: (1) a chimeric NDV, and (2) an antagonist of an inhibitory receptor of an immune cell or an agonist of a co-stimulatory receptor of an immune cell, wherein the chimeric NDV comprises a packaged genome encoding a cytokine which is expressed by the virus.
32. The method of claim 31, wherein the method comprises administering an antagonist of an inhibitory receptor of an immune cell.
33. (canceled)
34. (canceled)
35. The method of claim 31, wherein the cytokine is IL-2, IL-7, IL-15 or IL-21.
36. (canceled)
37. (canceled)
38. The method of claim 29, wherein the agonist is: (a) an antibody that specifically binds to the co-stimulatory receptor; or (b) a ligand that specifically binds to the co-stimulatory receptor.
39. (canceled)
40. (canceled)
41. The method of claim 38, wherein the antibody is a monoclonal antibody or single-chain Fv.
42. The method of claim 38, wherein the ligand is CD137L, OX40L, CD40L, GITRL, or CD70.
43. The method of claim 30, wherein the antagonist is an antibody that specifically binds to the inhibitory receptor.
44. (canceled)
45. The method of claim 30, wherein the antagonist is: (a) a soluble receptor of a ligand of the inhibitory receptor, or (b) an antibody that specifically binds to a ligand of the inhibitory receptor.
46. (canceled)
47. The method of claim 30, wherein the antagonist is an antibody that specifically binds to PDL1, PDL2, B7-H3, B7-H4, HVEM, or Ga19.
48. The method of claim 45, wherein the soluble receptor is the extracellular domain of PD1, BTLA, KIR, LAG3 or TIM3.
49. The method of claim 43, wherein the antibody is a monoclonal antibody or scFv.
50. The method of claim 29, wherein the cancer is melanoma, colon cancer, lung cancer, breast cancer, head and neck squamous cell carcinoma, ovarian cancer, pancreatic cancer, liver cancer, or renal cell cancer.
51. The method of claim 29, wherein the cancer is brain tumor, bladder cancer, breast cancer, cervical cancer, Hodgkin's disease, colorectal cancer, endometrial carcinoma, esophageal cancer, glioblastoma multiforme, hepatocellular carcinoma, kidney cancer, liver cancer, lung cancer, melanoma, mesothelioma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary gland cancer, sarcoma, thyroid cancer, or uterine cancer.
52. The method of claim 30, wherein the cancer is melanoma, colon cancer, lung cancer, breast cancer, head and neck squamous cell carcinoma, ovarian cancer, pancreatic cancer, liver cancer, or renal cell cancer.
53. The method of claim 30, wherein the cancer is brain tumor, bladder cancer, breast cancer, cervical cancer, Hodgkin's disease, colorectal cancer, endometrial carcinoma, esophageal cancer, glioblastoma multiforme, hepatocellular carcinoma, kidney cancer, liver cancer, lung cancer, melanoma, mesothelioma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary gland cancer, sarcoma, thyroid cancer, or uterine cancer.
54. The method of claim 31, wherein the cancer is melanoma, colon cancer, lung cancer, breast cancer, head and neck squamous cell carcinoma, ovarian cancer, pancreatic cancer, liver cancer, or renal cell cancer.
55. The method of claim 31, wherein the cancer is brain tumor, bladder cancer, breast cancer, cervical cancer, Hodgkin's disease, colorectal cancer, endometrial carcinoma, esophageal cancer, glioblastoma multiforme, hepatocellular carcinoma, kidney cancer, liver cancer, lung cancer, melanoma, mesothelioma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary gland cancer, sarcoma, thyroid cancer, or uterine cancer.
56. The method of claim 32, wherein the inhibitory receptor is CTLA-4, PD-1, BTLA, KIR, LAG3, or TIM3.
57. The method of claim 31, wherein the co-stimulatory receptor is GITR, ICOS, OX40, CD27, CD28, 41BB, CD40, LT alpha, or LIGHT.
58. The method of claim 56, wherein the antagonist is:
(a) an antibody that specifically binds to the inhibitory receptor;
(b) soluble receptor of a ligand of the inhibitory receptor, or
(c) an antibody that specifically binds to a ligand of the inhibitory receptor.
US15/789,340 2013-03-14 2017-10-20 Newcastle Disease Viruses and Uses Thereof Abandoned US20180256655A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/789,340 US20180256655A1 (en) 2013-03-14 2017-10-20 Newcastle Disease Viruses and Uses Thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361782994P 2013-03-14 2013-03-14
US14/205,776 US20140271677A1 (en) 2013-03-14 2014-03-12 Newcastle Disease Viruses and Uses Thereof
US15/789,340 US20180256655A1 (en) 2013-03-14 2017-10-20 Newcastle Disease Viruses and Uses Thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/205,776 Division US20140271677A1 (en) 2013-03-14 2014-03-12 Newcastle Disease Viruses and Uses Thereof

Publications (1)

Publication Number Publication Date
US20180256655A1 true US20180256655A1 (en) 2018-09-13

Family

ID=51527979

Family Applications (5)

Application Number Title Priority Date Filing Date
US14/774,962 Abandoned US20160015760A1 (en) 2013-03-14 2014-03-04 Newcastle disease viruses and uses thereof
US14/205,776 Abandoned US20140271677A1 (en) 2013-03-14 2014-03-12 Newcastle Disease Viruses and Uses Thereof
US15/588,251 Abandoned US20180078592A1 (en) 2013-03-14 2017-05-05 Newcastle Disease Viruses And Uses Thereof
US15/789,539 Active US10251922B2 (en) 2013-03-14 2017-10-20 Newcastle disease viruses and uses thereof
US15/789,340 Abandoned US20180256655A1 (en) 2013-03-14 2017-10-20 Newcastle Disease Viruses and Uses Thereof

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US14/774,962 Abandoned US20160015760A1 (en) 2013-03-14 2014-03-04 Newcastle disease viruses and uses thereof
US14/205,776 Abandoned US20140271677A1 (en) 2013-03-14 2014-03-12 Newcastle Disease Viruses and Uses Thereof
US15/588,251 Abandoned US20180078592A1 (en) 2013-03-14 2017-05-05 Newcastle Disease Viruses And Uses Thereof
US15/789,539 Active US10251922B2 (en) 2013-03-14 2017-10-20 Newcastle disease viruses and uses thereof

Country Status (28)

Country Link
US (5) US20160015760A1 (en)
EP (1) EP2968525A4 (en)
JP (4) JP6596411B2 (en)
KR (1) KR102222157B1 (en)
CN (3) CN111218429A (en)
AP (1) AP2015008685A0 (en)
AU (2) AU2014241843B2 (en)
BR (1) BR112015021414B1 (en)
CA (1) CA2905272A1 (en)
CL (2) CL2015002532A1 (en)
CR (1) CR20150465A (en)
DO (1) DOP2015000227A (en)
EA (1) EA038981B1 (en)
GE (1) GEP20196976B (en)
HK (1) HK1216618A1 (en)
IL (2) IL241120A0 (en)
MA (1) MA38406B1 (en)
MD (1) MD4655C1 (en)
MX (2) MX2015011886A (en)
MY (1) MY180687A (en)
NI (1) NI201500131A (en)
NZ (1) NZ711946A (en)
PE (1) PE20151921A1 (en)
PH (1) PH12015502087B1 (en)
SG (2) SG11201507412SA (en)
TN (1) TN2015000353A1 (en)
WO (1) WO2014158811A1 (en)
ZA (1) ZA201506192B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10251922B2 (en) 2013-03-14 2019-04-09 Icahn School Of Medicine At Mount Sinai Newcastle disease viruses and uses thereof
US10308913B2 (en) 2005-12-02 2019-06-04 Icahn School Of Medicine At Mount Sinai Chimeric viruses presenting non-native surface proteins and uses thereof

Families Citing this family (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2616355T3 (en) 2007-06-18 2017-06-12 Merck Sharp & Dohme B.V. Antibodies for the human programmed death receptor PD-1
ES2550179T3 (en) 2009-02-05 2015-11-05 Icahn School Of Medicine At Mount Sinai Chimeric Newcastle disease viruses and uses thereof
MX367768B (en) * 2013-09-03 2019-09-05 Medimmune Ltd Compositions featuring an attenuated newcastle disease virus and methods of use for treating neoplasia.
SI3081576T1 (en) 2013-12-12 2019-12-31 Shanghai Hengrui Pharmaceutical Co., Ltd., Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
US20170000832A1 (en) 2014-02-27 2017-01-05 Viralytics Limited Combination method for treatment of cancer
US9394365B1 (en) 2014-03-12 2016-07-19 Yeda Research And Development Co., Ltd Reducing systemic regulatory T cell levels or activity for treatment of alzheimer's disease
US10519237B2 (en) 2014-03-12 2019-12-31 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
MX2016011832A (en) 2014-03-12 2017-05-12 Yeda Res & Dev Reducing systemic regulatory t cell levels or activity for treatment of disease and injury of the cns.
US10618963B2 (en) 2014-03-12 2020-04-14 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
KR102594343B1 (en) 2014-07-21 2023-10-26 노파르티스 아게 Treatment of cancer using a cd33 chimeric antigen receptor
RU2751660C2 (en) 2014-07-21 2021-07-15 Новартис Аг Treatment of malignant neoplasm using humanized chimeric antigen receptor against bcma
EP3193915A1 (en) 2014-07-21 2017-07-26 Novartis AG Combinations of low, immune enhancing. doses of mtor inhibitors and cars
EP3174546B1 (en) 2014-07-31 2019-10-30 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
CA2958200A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using a gfr alpha-4 chimeric antigen receptor
EP3183268B1 (en) 2014-08-19 2020-02-12 Novartis AG Anti-cd123 chimeric antigen receptor (car) for use in cancer treatment
PL3194443T3 (en) 2014-09-17 2022-01-31 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
TN2017000129A1 (en) 2014-10-14 2018-10-19 Dana Farber Cancer Inst Inc Antibody molecules to pd-l1 and uses thereof
SG11201703326SA (en) * 2014-10-24 2017-05-30 Aladar Szalay Combination immunotherapy approach for treatment of cancer
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
CN105985966A (en) * 2015-03-06 2016-10-05 普莱柯生物工程股份有限公司 Gene VII-type newcastle disease virus strain, vaccine composition thereof and preparing method and application of vaccine composition
LT3280729T (en) 2015-04-08 2022-08-10 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
GB201509338D0 (en) 2015-05-29 2015-07-15 Bergenbio As Combination therapy
CA2991976A1 (en) 2015-07-13 2017-01-19 Cytomx Therapeutics, Inc. Anti-pd-1 antibodies, activatable anti-pd-1 antibodies, and methods of use thereof
CN116196426A (en) 2015-07-16 2023-06-02 百欧肯治疗有限公司 Compositions and methods for treating cancer
US20180340025A1 (en) 2015-07-29 2018-11-29 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
JP6878405B2 (en) 2015-07-29 2021-05-26 ノバルティス アーゲー Combination therapy with antibody molecule against PD-1
CN108135996B (en) 2015-08-11 2022-06-21 卡利迪生物治疗有限公司 Smallpox vaccine for cancer treatment
JP2019503349A (en) 2015-12-17 2019-02-07 ノバルティス アーゲー Antibody molecules against PD-1 and uses thereof
EP3778881A1 (en) 2016-01-08 2021-02-17 Replimune Limited Modified oncolytic virus
BR112018067679A2 (en) 2016-03-04 2019-01-15 Novartis Ag cells expressing multiple chimeric antigen receptor (car) molecules and their use
CN109641947B (en) 2016-07-20 2023-04-14 犹他大学研究基金会 CD229CAR T cells and methods of use thereof
SG10201913823VA (en) 2016-10-07 2020-03-30 Novartis Ag Chimeric antigen receptors for the treatment of cancer
CN109996809A (en) 2016-11-14 2019-07-09 诺华股份有限公司 Composition relevant to fusogenic protein MINION, method and therapeutical uses
US20200024351A1 (en) 2017-04-03 2020-01-23 Jounce Therapeutics, Inc. Compositions and Methods for the Treatment of Cancer
US20200179511A1 (en) 2017-04-28 2020-06-11 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
JOP20190256A1 (en) 2017-05-12 2019-10-28 Icahn School Med Mount Sinai Newcastle disease viruses and uses thereof
US11684637B2 (en) 2017-05-25 2023-06-27 University Of Central Florida Research Foundation, Inc. Oncolytic viruses for sensitizing tumor cells to killing by natural killer cells
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
EP3801769A1 (en) 2018-05-25 2021-04-14 Novartis AG Combination therapy with chimeric antigen receptor (car) therapies
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
US20210379153A1 (en) * 2018-08-29 2021-12-09 Shattuck Labs, Inc. Combination therapies comprising sirp alpha-based chimeric proteins
EP3856782A1 (en) 2018-09-28 2021-08-04 Novartis AG Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
US20220047633A1 (en) 2018-09-28 2022-02-17 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020075672A1 (en) * 2018-10-09 2020-04-16 バイオコモ株式会社 Anticancer agent, pharmaceutical composition for cancer treatment, and kit
CN109627336A (en) * 2018-12-20 2019-04-16 南京昂科利医药科技创新研究院有限公司 A kind of preparation method and application of newcastle disease oncolytic virus that expressing PD-L1 single-chain antibody
US20230193213A1 (en) * 2019-01-29 2023-06-22 Arno Thaller Recombinant oncolytic newcastle disease viruses with increased activity
US20220135682A1 (en) 2019-03-11 2022-05-05 Jounce Therapeutics, Inc. Anti-ICOS Antibodies for the Treatment of Cancer
BR112021019328A2 (en) 2019-03-29 2021-11-30 Myst Therapeutics Llc Ex vivo methods for producing a t-cell therapeutic product and related compositions and methods
EP3725370A1 (en) 2019-04-19 2020-10-21 ImmunoBrain Checkpoint, Inc. Modified anti-pd-l1 antibodies and methods and uses for treating a neurodegenerative disease
CN110564766A (en) * 2019-09-20 2019-12-13 华农(肇庆)生物产业技术研究院有限公司 Preparation method of whole genome expression vector pBR322-DHN3
CN110672844A (en) * 2019-10-29 2020-01-10 华中科技大学 Newcastle disease virus antibody magnetic immuno-chemiluminescence detection kit and application thereof
US20220378909A1 (en) 2019-11-05 2022-12-01 Jounce Therapeutics, Inc. Methods of Treating Cancer with Anti-PD-1 Antibodies
TW202134285A (en) 2019-11-26 2021-09-16 瑞士商諾華公司 Cd19 and cd22 chimeric antigen receptors and uses thereof
EP4065229A1 (en) 2019-11-27 2022-10-05 Myst Therapeutics, LLC Method of producing tumor-reactive t cell composition using modulatory agents
CN115427438A (en) 2020-02-27 2022-12-02 迈斯特治疗公司 Methods for ex vivo enrichment and expansion of tumor-reactive T cells and compositions related thereto
US20210386804A1 (en) * 2020-06-11 2021-12-16 Tibor Bakács Combination of viral superinfection therapy with subthreshold doses of nivolumab plus ipilimumab in chronic HBV patients
IL302700A (en) 2020-11-13 2023-07-01 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
CN115197949A (en) * 2021-04-13 2022-10-18 江苏康缘瑞翱生物医药科技有限公司 Recombinant Newcastle disease virus rNDV-OX40L, genome thereof, preparation method and application thereof
WO2022254337A1 (en) 2021-06-01 2022-12-08 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
KR102476901B1 (en) * 2021-08-06 2022-12-14 리벤텍 주식회사 Oncolytic virus constructed by based on the colorectal cancer cells-specific infection Newcastle disease virus for colorectal cancer treatment and its composition

Family Cites Families (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3806565A1 (en) 1988-03-01 1989-09-14 Deutsches Krebsforsch VIRUS-MODIFIED TUMOR VACCINES FOR THE IMMUNOTHERAPY OF TUMOR METAL KEYS
DE3922444A1 (en) 1988-03-01 1991-01-10 Deutsches Krebsforsch Virus modified tumour specific vaccine - contains immunostimulating substances, and is used in immuno-therapy of tumours
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5854037A (en) 1989-08-28 1998-12-29 The Mount Sinai School Of Medicine Of The City University Of New York Recombinant negative strand RNA virus expression systems and vaccines
US5166057A (en) 1989-08-28 1992-11-24 The Mount Sinai School Of Medicine Of The City University Of New York Recombinant negative strand rna virus expression-systems
US5786199A (en) 1989-08-28 1998-07-28 The Mount Sinai School Of Medicine Of The City University Of New York Recombinant negative strand RNA virus expression systems and vaccines
EP1314431B1 (en) 1993-04-30 2008-07-16 Wellstat Biologics Corporation Purified compositions of Newcastle disease virus
ES2210273T5 (en) 1994-07-18 2010-03-29 Conzelmann, Karl-Klaus, Prof. Dr. VIRUS WITH NEGATIVE CHAIN NON-SEGMENTED RECOMBINANT INFECTIVE.
US5891680A (en) 1995-02-08 1999-04-06 Whitehead Institute For Biomedical Research Bioactive fusion proteins comprising the p35 and p40 subunits of IL-12
US7153510B1 (en) 1995-05-04 2006-12-26 Yale University Recombinant vesiculoviruses and their uses
EP0780475B2 (en) 1995-08-09 2006-07-19 SCHWEIZ. SERUM- &amp; IMPFINSTITUT BERN Process for the production of infectious negative-strand RNA viruses
JPH11512609A (en) 1995-09-27 1999-11-02 アメリカ合衆国 Production of infectious respiratory syncytial virus from cloned nucleotide sequence
WO1997014433A1 (en) 1995-10-17 1997-04-24 Wayne State University Chicken interleukin-15 and uses thereof
CA2257823A1 (en) 1996-07-15 1998-01-22 Government Of The United States Of America As Represented By The Secreta Ry Of The Department Of Health And Human Services National Institutes Of Production of attenuated respiratory syncytial virus vaccines from cloned nucleotide sequences
JP2000517194A (en) 1996-09-27 2000-12-26 アメリカン・サイアナミド・カンパニー Mutations in the 3 'genomic promoter region and polymerase gene responsible for attenuation in the virus of the eye termed Mononegavirales
KR100702523B1 (en) 1997-05-23 2007-04-04 더 가번먼트 오브 더 유나이티드 스테이츠 오브 아메리카, 에즈 레프리젠티드 바이 더 디파트먼트 오브 헬쓰 앤드 휴먼 서비시즈 Production of attenuated parainfluenza virus vaccines from cloned nucleotide sequences
PT1012244E (en) 1997-07-11 2007-08-21 Univ Yale Rhabdoviruses with reengineered coats
JP2001517448A (en) 1997-09-19 2001-10-09 アメリカン・サイアナミド・カンパニー Attenuated respiratory syncytial virus
US20030044384A1 (en) 1997-10-09 2003-03-06 Pro-Virus, Inc. Treatment of neoplasms with viruses
US7780962B2 (en) 1997-10-09 2010-08-24 Wellstat Biologics Corporation Treatment of neoplasms with RNA viruses
CN1281336A (en) 1997-10-09 2001-01-24 病毒防御公司 Treatment of neoplasms with viruses
US7470426B1 (en) 1997-10-09 2008-12-30 Wellstat Biologics Corporation Treatment of neoplasms with viruses
CA2334895C (en) 1998-06-12 2016-01-19 Mount Sinai School Of Medicine Attenuated negative strand viruses with altered interferon antagonist activity for use as vaccines and pharmaceuticals
EP0974660A1 (en) 1998-06-19 2000-01-26 Stichting Instituut voor Dierhouderij en Diergezondheid (ID-DLO) Newcastle disease virus infectious clones, vaccines and diagnostic assays
US6146642A (en) 1998-09-14 2000-11-14 Mount Sinai School Of Medicine, Of The City University Of New York Recombinant new castle disease virus RNA expression systems and vaccines
US6544785B1 (en) 1998-09-14 2003-04-08 Mount Sinai School Of Medicine Of New York University Helper-free rescue of recombinant negative strand RNA viruses
US7052685B1 (en) 1998-10-15 2006-05-30 Trustees Of The University Of Pennsylvania Methods for treatment of cutaneous T-cell lymphoma
HUP0302278A3 (en) 1999-04-15 2011-01-28 Pro Virus Treatment of neoplasms with viruses
US20030224017A1 (en) 2002-03-06 2003-12-04 Samal Siba K. Recombinant Newcastle disease viruses useful as vaccines or vaccine vectors
AU4971500A (en) 1999-05-05 2000-11-21 University Of Maryland Production of novel newcastle disease virus strains from cdnas and improved liveattenuated newcastle disease vaccines
DK1194580T4 (en) 1999-07-14 2011-01-03 Sinai School Medicine In-vitro reconstitution of segmented negative-stranded RNA viruses
WO2001020989A1 (en) 1999-09-22 2001-03-29 Mayo Foundation For Medical Education And Research Therapeutic methods and compositions using viruses of the recombinant paramyxoviridae family
US6896881B1 (en) 1999-09-24 2005-05-24 Mayo Foundation For Medical Education And Research Therapeutic methods and compositions using viruses of the recombinant paramyxoviridae family
WO2001052874A2 (en) 2000-01-20 2001-07-26 Universität Zürich Institut für Medizinische Virologie Intra-tumoral administration of il-12 encoding nucleic acid molecules
WO2001077394A1 (en) 2000-04-10 2001-10-18 Mount Sinai School Of Medicine Of New York University Screening methods for identifying viral proteins with interferon antagonizing functions and potential antiviral agents
US6818444B2 (en) 2000-08-04 2004-11-16 Heska Corporation Canine and feline proteins, nucleic acid molecules and uses thereof
FR2823222B1 (en) 2001-04-06 2004-02-06 Merial Sas VACCINE AGAINST NILE FEVER VIRUS
WO2002102404A1 (en) 2001-06-18 2002-12-27 Institut National De La Recherche Agronomique Uses of cytokines
WO2003092579A2 (en) 2002-04-29 2003-11-13 Hadasit Medical Research Services And Development Company Ltd. Compositions and methods for treating cancer with an oncolytic viral agent
JPWO2003102183A1 (en) 2002-06-03 2005-09-29 株式会社ディナベック研究所 Paramyxovirus vectors encoding antibodies and uses thereof
EP1543418B1 (en) 2002-08-07 2016-03-16 MMagix Technology Limited Apparatus, method and system for a synchronicity independent, resource delegating, power and instruction optimizing processor
SE0203159D0 (en) 2002-10-25 2002-10-25 Electrolux Ab Handle for a motor driven handheld tool
US9068234B2 (en) 2003-01-21 2015-06-30 Ptc Therapeutics, Inc. Methods and agents for screening for compounds capable of modulating gene expression
US20040197312A1 (en) 2003-04-02 2004-10-07 Marina Moskalenko Cytokine-expressing cellular vaccine combinations
WO2005085282A1 (en) 2004-02-27 2005-09-15 Inserm (Institut National De La Sante Et De La Recherche Medicale) Il-15 binding site for il15-ralpha and specific il-15 mutants having agonists/antagonists activity
AU2005303912B2 (en) * 2004-11-12 2011-02-17 Bayer Schering Pharma Aktiengesellschaft Recombinant Newcastle Disease Virus
NZ563263A (en) 2005-05-17 2010-04-30 Univ Connecticut Composition and methods for immunomodulation in an organism
WO2007008918A2 (en) 2005-07-08 2007-01-18 Wayne State University Virus vaccines comprising envelope-bound immunomodulatory proteins and methods of use thereof
EP1777294A1 (en) 2005-10-20 2007-04-25 Institut National De La Sante Et De La Recherche Medicale (Inserm) IL-15Ralpha sushi domain as a selective and potent enhancer of IL-15 action through IL-15Rbeta/gamma, and hyperagonist (IL15Ralpha sushi -IL15) fusion proteins
GEP20135924B (en) 2005-12-02 2013-10-10 The Mount Sinai School Of Medicine Of New York Univ Chimeric viruses presenting non-native surface proteins and usage thereof
AU2007207785B2 (en) 2006-01-13 2013-11-14 The Government Of The United States, As Represented By The Secretary Of The Department Of Health And Human Services, National Institutes Of Health Codon optimized IL- 15 and IL- 15R-alpha genes for expression in mammalian cells
WO2007113648A2 (en) 2006-04-05 2007-10-11 Pfizer Products Inc. Ctla4 antibody combination therapy
US20090175826A1 (en) 2006-06-05 2009-07-09 Elankumaran Subbiah Genetically-engineered newcastle disease virus as an oncolytic agent, and methods of using same
WO2008011726A1 (en) 2006-07-27 2008-01-31 Ottawa Health Research Institute Staged immune-response modulation in oncolytic therapy
CA2723320C (en) 2007-05-04 2019-06-11 University Health Network Il-12 immunotherapy for cancer
EP3305805A1 (en) 2007-05-11 2018-04-11 Altor BioScience Corporation Fusion molecules and il-15 variants
ES2616355T3 (en) 2007-06-18 2017-06-12 Merck Sharp & Dohme B.V. Antibodies for the human programmed death receptor PD-1
NZ616254A (en) 2007-06-27 2015-04-24 Us Sec Dep Of Health And Human Services Complexes of il-15 and il-15ralpha and uses thereof
EP2085092A1 (en) 2008-01-29 2009-08-05 Bayer Schering Pharma Aktiengesellschaft Attenuated oncolytic paramyxoviruses encoding avian cytokines
US8313896B2 (en) * 2008-04-04 2012-11-20 The General Hospital Corporation Oncolytic herpes simplex virus immunotherapy in the treatment of brain cancer
EP2350129B1 (en) 2008-08-25 2015-06-10 Amplimmune, Inc. Compositions of pd-1 antagonists and methods of use
WO2010042433A1 (en) * 2008-10-06 2010-04-15 Bristol-Myers Squibb Company Combination of cd137 antibody and ctla-4 antibody for the treatment of proliferative diseases
CN101787373B (en) 2009-01-23 2013-06-19 中国人民解放军第二军医大学东方肝胆外科医院 Foreign gene-carrying recombinant virus vector efficiently produced in packaging cell and construction method and application thereof
ES2550179T3 (en) * 2009-02-05 2015-11-05 Icahn School Of Medicine At Mount Sinai Chimeric Newcastle disease viruses and uses thereof
AU2010241864B2 (en) 2009-04-30 2014-02-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Inducible interleukin-12
US20100297072A1 (en) 2009-05-19 2010-11-25 Depinho Ronald A Combinations of Immunostimulatory Agents, Oncolytic Virus, and Additional Anticancer Therapy
EP3135294B1 (en) 2009-08-14 2020-06-03 The Government of the United States of America as represented by the Secretary of the Department of Health and Human Services Use of il-15-il-15 receptor heterodimers to treat lymphopenia
SG178523A1 (en) 2009-08-21 2012-03-29 Merial Ltd Recombinant avian paramyxovirus vaccine and method for making and using thereof
CN102740887B (en) 2009-09-30 2015-04-15 斯隆凯特林防癌纪念中心 Combination immunotherapy for the treatment of cancer
WO2011119628A2 (en) 2010-03-23 2011-09-29 The Regents Of The University Of California Compositions and methods for self-adjuvanting vaccines against microbes and tumors
WO2012000188A1 (en) 2010-06-30 2012-01-05 Tot Shanghai Rd Center Co., Ltd. Recombinant tumor vaccine and method of producing such
DK3327040T3 (en) 2010-09-21 2021-09-20 Altor Bioscience Corp MULTIMERIC SOLUBLE IL-15 FUSION MOLECULES AND METHODS OF PREPARING AND USING THE SAME
US20120082687A1 (en) * 2010-10-04 2012-04-05 Alex Wah Hin Yeung Use of cell adhesion inhibitor for the mobilization of antigen presenting cells and immune cells in a cell mixture (AIM) from the peripheral blood and methods of use
CN114262690A (en) 2011-04-15 2022-04-01 吉恩勒克斯公司 Clonal strains of attenuated vaccinia virus and methods of use thereof
EP2537933A1 (en) 2011-06-24 2012-12-26 Institut National de la Santé et de la Recherche Médicale (INSERM) An IL-15 and IL-15Ralpha sushi domain based immunocytokines
JP6489353B2 (en) 2011-10-11 2019-03-27 ウニヴェルズィテート チューリッヒ Pharmaceutical composition comprising IL-12 and a T cell inhibitory molecular blocker for tumor therapy
CA2862390A1 (en) 2012-01-25 2013-08-01 Dnatrix, Inc. Biomarkers and combination therapies using oncolytic virus and immunomodulation
EP2669381A1 (en) 2012-05-30 2013-12-04 AmVac AG Method for expression of heterologous proteins using a recombinant negative-strand RNA virus vector comprising a mutated P protein
US20150250837A1 (en) 2012-09-20 2015-09-10 Morningside Technology Ventures Ltd. Oncolytic virus encoding pd-1 binding agents and uses of the same
WO2014066527A2 (en) 2012-10-24 2014-05-01 Admune Therapeutics Llc Il-15r alpha forms, cells expressing il-15r alpha forms, and therapeutic uses of il-15r alpha and il-15/il-15r alpha complexes
GEP20196976B (en) * 2013-03-14 2019-06-10 Sloan Kettering Cancer Center Memorial Newcastle disease viruses and uses thereof
CN105324124A (en) 2013-04-19 2016-02-10 赛腾制药 Cytokine derived treatment with reduced vascular leak syndrome
WO2017123981A1 (en) 2016-01-15 2017-07-20 Rfemb Holdings, Llc Immunologic treatment of cancer
AU2014304931B2 (en) 2013-08-08 2018-05-10 Assistance Publique - Hopitaux De Paris IL-15 and IL-15Raplha sushi domain based modulokines
DK3030262T3 (en) 2013-08-08 2019-12-02 Cytune Pharma COMBINED PHARMACEUTICAL COMPOSITION
MX367768B (en) 2013-09-03 2019-09-05 Medimmune Ltd Compositions featuring an attenuated newcastle disease virus and methods of use for treating neoplasia.
US20170000832A1 (en) 2014-02-27 2017-01-05 Viralytics Limited Combination method for treatment of cancer
EP2915569A1 (en) 2014-03-03 2015-09-09 Cytune Pharma IL-15/IL-15Ralpha based conjugates purification method
ES2811974T3 (en) 2014-07-29 2021-03-15 Novartis Ag Dose escalation regimens of il-15 and il-15ralpha heterodimer for treating conditions
WO2016048903A1 (en) 2014-09-22 2016-03-31 Intrexon Corporation Improved therapeutic control of heterodimeric and single chain forms of interleukin-12
AU2015360736A1 (en) 2014-12-09 2017-06-01 Merck Sharp & Dohme Corp. System and methods for deriving gene signature biomarkers of response to PD-1 antagonists
CN106166294A (en) 2015-05-18 2016-11-30 国科丹蓝生物科技(北京)有限公司 A kind of compound for preoperative intervention radiotherapy in the treatment tumor
US20180340025A1 (en) 2015-07-29 2018-11-29 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
JP6878405B2 (en) 2015-07-29 2021-05-26 ノバルティス アーゲー Combination therapy with antibody molecule against PD-1
CA3001590A1 (en) 2015-10-10 2017-04-13 Intrexon Corporation Improved therapeutic control of proteolytically sensitive, destabilized forms of interleukin-12
WO2017083291A1 (en) 2015-11-09 2017-05-18 Immune Design Corp. Compositions comprising lentiviral vectors expressing il-12 and methods of use thereof
EP3778881A1 (en) 2016-01-08 2021-02-17 Replimune Limited Modified oncolytic virus
US10344067B2 (en) 2016-02-25 2019-07-09 Deutsches Krebsforschungszentrum RNA viruses expressing IL-12 for immunovirotherapy
CN105734023B (en) 2016-03-28 2019-04-26 江苏康缘瑞翱生物医药科技有限公司 A kind of recombinant Newcastle disease virus is preparing the application in medicines resistant to liver cancer
CN115960966A (en) 2016-04-29 2023-04-14 复诺健生物科技加拿大有限公司 HSV vectors with enhanced replication in cancer cells

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10308913B2 (en) 2005-12-02 2019-06-04 Icahn School Of Medicine At Mount Sinai Chimeric viruses presenting non-native surface proteins and uses thereof
US10251922B2 (en) 2013-03-14 2019-04-09 Icahn School Of Medicine At Mount Sinai Newcastle disease viruses and uses thereof

Also Published As

Publication number Publication date
MA38406B1 (en) 2020-03-31
CA2905272A1 (en) 2014-10-02
WO2014158811A8 (en) 2015-09-17
HK1216618A1 (en) 2016-11-25
MD4655B1 (en) 2019-11-30
EP2968525A1 (en) 2016-01-20
BR112015021414B1 (en) 2020-11-10
NZ711946A (en) 2020-05-29
EP2968525A4 (en) 2016-10-26
CN111172120A (en) 2020-05-19
WO2014158811A1 (en) 2014-10-02
JP6596411B2 (en) 2019-10-23
PH12015502087A1 (en) 2016-01-18
AU2019206040A1 (en) 2019-08-01
SG11201507412SA (en) 2015-10-29
JP2023002553A (en) 2023-01-10
AP2015008685A0 (en) 2015-08-31
KR20150127164A (en) 2015-11-16
US20160015760A1 (en) 2016-01-21
KR102222157B1 (en) 2021-03-03
US10251922B2 (en) 2019-04-09
IL264385B (en) 2021-09-30
AU2014241843A1 (en) 2015-09-10
SG10201802982WA (en) 2018-06-28
CN105188746A (en) 2015-12-23
PE20151921A1 (en) 2015-12-26
IL241120A0 (en) 2015-11-30
US20180280455A1 (en) 2018-10-04
MX2015011886A (en) 2016-05-31
CR20150465A (en) 2016-01-25
JP2018198621A (en) 2018-12-20
EA038981B1 (en) 2021-11-17
TN2015000353A1 (en) 2017-01-03
MX2019008086A (en) 2019-09-10
JP2016517269A (en) 2016-06-16
MA38406A1 (en) 2017-12-29
DOP2015000227A (en) 2015-11-15
CL2015002532A1 (en) 2016-07-15
MY180687A (en) 2020-12-07
GEP20196976B (en) 2019-06-10
AU2014241843B2 (en) 2019-05-02
CN111218429A (en) 2020-06-02
CL2018000515A1 (en) 2018-08-03
MD20150100A2 (en) 2016-02-29
CN105188746B (en) 2020-03-17
EA201591740A1 (en) 2016-06-30
JP2020202850A (en) 2020-12-24
IL264385A (en) 2019-02-28
US20140271677A1 (en) 2014-09-18
MD4655C1 (en) 2020-06-30
PH12015502087B1 (en) 2016-01-18
ZA201506192B (en) 2021-01-27
BR112015021414A2 (en) 2017-07-18
NI201500131A (en) 2015-10-19
US20180078592A1 (en) 2018-03-22

Similar Documents

Publication Publication Date Title
US10251922B2 (en) Newcastle disease viruses and uses thereof
US10035984B2 (en) Chimeric newcastle disease viruses and uses thereof
US20200061184A1 (en) Newcastle disease viruses and uses thereof
US20220241358A1 (en) Apmv and uses thereof for the treatment of cancer
US20230151070A1 (en) Vegfr-3-activating agents and oncolytic viruses and uses thereof for the treatment of cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: MEMORIAL SLOAN KETTERING CANCER CENTER, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZAMARIN, DMITRIY;ALLISON, JAMES;WOLCHOK, JEDD;SIGNING DATES FROM 20140224 TO 20140304;REEL/FRAME:047697/0248

Owner name: ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PALESE, PETER;GARCIA-SASTRE, ADOLFO;REEL/FRAME:047697/0128

Effective date: 20140129

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION