WO2022147480A1 - Virus oncolytique codant pour la sialidase et agent de ciblage de cellule immunitaire multispécifique - Google Patents

Virus oncolytique codant pour la sialidase et agent de ciblage de cellule immunitaire multispécifique Download PDF

Info

Publication number
WO2022147480A1
WO2022147480A1 PCT/US2021/073203 US2021073203W WO2022147480A1 WO 2022147480 A1 WO2022147480 A1 WO 2022147480A1 US 2021073203 W US2021073203 W US 2021073203W WO 2022147480 A1 WO2022147480 A1 WO 2022147480A1
Authority
WO
WIPO (PCT)
Prior art keywords
sialidase
cells
recombinant oncolytic
virus
amino acid
Prior art date
Application number
PCT/US2021/073203
Other languages
English (en)
Inventor
Nancy Chang
Original Assignee
Ansun Biopharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ansun Biopharma, Inc. filed Critical Ansun Biopharma, Inc.
Publication of WO2022147480A1 publication Critical patent/WO2022147480A1/fr

Links

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
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001129Molecules with a "CD" designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/275Poxviridae, e.g. avipoxvirus
    • A61K39/285Vaccinia virus or variola virus
    • 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
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01018Exo-alpha-sialidase (3.2.1.18), i.e. trans-sialidase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y402/00Carbon-oxygen lyases (4.2)
    • C12Y402/02Carbon-oxygen lyases (4.2) acting on polysaccharides (4.2.2)
    • C12Y402/02015Anhydrosialidase (4.2.2.15)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present application relates to methods and compositions for treating cancer with an oncolytic virus (e.g. , vaccinia virus) encoding a sialidase and a multispecific immune cell engager.
  • an oncolytic virus e.g. , vaccinia virus
  • Cancer is the second leading cause of death in the United States.
  • great progress has been made in cancer immunotherapy, including immune checkpoint inhibitors, T cells with chimeric antigen receptors, and oncolytic viruses.
  • Oncolytic viruses are naturally occurring or genetically modified viruses that infect, replicate in, and eventually kill cancer cells while leaving healthy cells unharmed.
  • a recently completed Phase III clinical trial of the oncolytic herpes simplex virus T-VEC in 436 patients with unresectable stage IIIB, IIIC or IV melanoma was reported to meet its primary end point, with a durable response rate of 16.3% in patients receiving T-VEC compared to 2.1% in patients receiving GM-CSF. Based on the results from this trial, FDA approved T-VEC in 2015.
  • Oncolytic virus constructs from at least eight different species have been tested in various phases of clinical trials, including adenovirus, herpes simplex virus- 1, Newcastle disease virus, reovirus, measles virus, coxsackievirus, Seneca Valley virus, and vaccinia virus. It has become clear that oncolytic viruses are well tolerated in patients with cancer. The clinical benefits of oncolytic viruses as stand-alone treatments, however, remain limited. Due to concerns on the safety of oncolytic viruses, only highly attenuated oncolytic viruses (either naturally avirulent or attenuated through genetic engineering) have been used in both preclinical and clinical studies.
  • Oncolytic viruses with a robust oncolytic effect will release abundant tumor antigens to prime or activate immune cells including T and NK cells, resulting in a strong immunotherapeutic effect.
  • the present application provides methods and compositions for delivery of an oncolytic virus expressing a heterologous protein or nucleic acid to cancer cells.
  • One aspect of the present application provides a recombinant oncolytic virus comprising a nucleotide sequence encoding one or more human or bacterial sialidases or a protein containing a sialidase catalytic domain thereof.
  • the recombinant oncolytic viruses can be derived from a poxvirus, an adenovirus, a herpes virus or any other suitable oncolytic virus.
  • Suitable recombinant oncolytic viruses can be created by inserting an expression cassette that includes a sequence encoding a sialidase or a portion thereof with sialidase activity into an oncolytic virus.
  • the nucleotide sequence encoding the sialidase is operably linked to a promoter.
  • the recombinant oncolytic viruses described herein are capable of delivering sialidase to tumor cells and the tumor microenvironment.
  • the delivered sialidase can reduce sialic acid present on tumor cells and render the tumor cells more vulnerable to killing by immune cells, immune cell-based therapies and other therapeutic agents whose effectiveness is diminished by hypersialylation of cancer cells.
  • a group of receptors called Siglec Sialic acid-binding immunoglobulin like lectins
  • Siglec sialic acid-binding immunoglobulin like lectins
  • the removal of sialic acid prevents binding of such ligands to Siglec on immune cells and thus abolishes the suppression of immunity against tumor cells.
  • a recombinant oncolytic virus comprising a first nucleotide sequence encoding a sialidase and a second nucleotide sequence encoding a multispecific immune cell engager, wherein the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the multispecific immune cell engager is a bispecific immune cell engager.
  • the multispecific immune cell engager comprises a first antigen-binding domain capable of specifically recognizing a tumor antigen and a second antigen-binding domain capable of specifically recognizing a cell surface molecule of an immune effector cell.
  • the tumor antigen is selected from the group consisting of fibroblast activation protein (FAP), epithelial cellular adhesion molecule (EpCAM), and epidermal growth factor receptor (EGFR).
  • FAP fibroblast activation protein
  • EpCAM epithelial cellular adhesion molecule
  • EGFR epidermal growth factor receptor
  • the tumor antigen is FAP.
  • the cell surface molecule on the effector cell is CD3 or 41-BB.
  • the cell surface marker on the effector cell is CD3s.
  • the first antigen-binding domain is an scFv
  • the second antigen binding domain is an scFv
  • the tumor antigen is FAP and the first antigenbinding domain comprises: (i) a first light chain complementarity -determining region (CDR- Ll) having the amino acid sequence of SEQ ID NO: 86, (i) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 87, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 88, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 89, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 90, and
  • the tumor antigen is FAP and the first antigen-binding domain comprises an amino acid sequence having at least 85% identity to the amino acid sequence of SEQ ID NO: 98. In some embodiments, the first antigenbinding domain comprises the amino acid sequence of SEQ ID NO: 98.
  • the cell surface molecule on the effector cell is CD3, and the second antigenbinding domain comprises: (i) a first light chain complementarity-determining region (CDR- Ll) having the amino acid sequence of SEQ ID NO: 92, (ii) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 93, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 94, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 95, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 96, and (vi) a third heavy chain complementarity-determining region (CDR-H3) having the amino acid sequence of SEQ ID NO:
  • the cell surface molecule on the effector cell is CD3 and the second antigen-binding domain comprises an amino acid sequence having at least 85% identity to the amino acid sequence of SEQ ID NO: 99. In some embodiments, the cell surface molecule on the effector cell is CD3 and the second antigenbinding domain comprises the amino acid sequence of SEQ ID NO: 99.
  • the multispecific immune cell engager comprises an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID N O: 100. In some embodiments according to any of the recombinant oncolytic viruses described above, the multispecific immune cell engager comprises the amino acid sequence of SEQ ID N O: 100.
  • the recombinant oncolytic virus is a virus selected from the group consisting of: vaccinia virus, reovirus, Seneca Valley virus (SVV), vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), herpes simplex virus (HSV), morbillivirus virus, retrovirus, influenza virus, Sinbis virus, poxvirus, measles virus, cytomegalovirus (CMV), lentivirus, adenovirus, and derivatives thereof.
  • the virus is Talimogene Laherparepvec.
  • the virus is a reovirus.
  • the virus is an adenovirus having an E1ACR2 deletion.
  • the recombinant oncolytic virus is a poxvirus.
  • the poxvirus is a vaccinia virus.
  • the vaccinia virus is of a strain selected from the group consisting of Dryvax, Lister, M63, LIVP, Tian Tan, Modified Vaccinia Ankara, New York City Board of Health (NYCBOH), Dairen, Ikeda, LC16M8, Tashkent, IHD-J, Brighton, Dairen I, Connaught, Elstree, Wyeth, Copenhagen, Western Reserve, Elstree, CL, Lederle-Chorioallantoic, AS, and derivatives thereof.
  • the virus is vaccinia virus Western Reserve.
  • the recombinant oncolytic virus comprises one or more mutations that reduce immunogenicity of the virus compared to a corresponding wild-type strain.
  • the virus is a vaccinia virus, and the one or more mutations are in one or more proteins selected from the group consisting of A14, A17, A13, LI, H3, D8, A33, B5, A56, F13, A28, and A27.
  • the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R.
  • the virus is a vaccinia virus, and the virus comprises one or more proteins selected from the group consisting of: (a) a variant vaccinia virus (VV) H3L protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to any one of SEQ ID NOS: 66-69; (b) a variant vaccinia virus (VV) D8L protein that comprises an amino acid seqOuence having at least 90% amino acid sequence identity to any one of SEQ ID NOS: 70-72 or 85; (c) a variant vaccinia virus (VV) A27L protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to SEQ ID NO: 73; and (d) a variant vaccinia virus (VV) L1R protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to SEQ ID NO: 74.
  • VV variant vaccinia virus
  • VV H3L protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to any one of SEQ ID NOS
  • the recombinant oncolytic virus is a vaccinia virus, and the recombinant oncolytic virus comprises a disruption of a thymidine kinase (TK) gene.
  • TK thymidine kinase
  • the first and second nucleotide sequences are inserted into the TK gene.
  • the recombinant oncolytic virus comprises a disruption of a vaccinia growth factor (VGF) gene.
  • the sialidase is a Neu5Ac alpha(2,6)-Gal sialidase, a Neu5Ac alpha(2,3)-Gal sialidase, or aNeu5Ac alpha(2,8)-Gal sialidase.
  • the sialidase is any protein having exo-sialidase activity (Enzyme Commission EC 3.2.1.18) including bacterial, human, fungal, viral sialidase and derivatives thereof.
  • the bacterial sialidase is selected from the group consisting of: Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase and Vibrio cholera sialidase.
  • the sialidase is a human sialidase or a derivative thereof.
  • the sialidase is NEU1, NEU2, NEU3, or NEU4.
  • the sialidase is a naturally occurring sialidase.
  • the sialidase comprises an anchoring domain.
  • the sialidase is a fusion protein comprising a sialidase catalytic domain fused to an anchoring domain.
  • the anchoring domain is positively charged at physiologic pH.
  • the anchoring domain is a glycosaminoglycan (GAG)-binding domain.
  • the sialidase is a protein having exo-sialidase activity as defined by Enzyme Comission EC 3.2.1.18.
  • the sialidase is an anhydrosialidase as defined by Enzyme Commission EC 4.2.2.15.
  • the sialidase comprises an amino acid sequence having at least about 80% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-33, 53-54, and 105. In some embodiments, the sialidase comprises an amino acid sequence having at least about 80% sequence identity to the amino acid sequence of SEQ ID NO: 1. In some embodiments, the sialidase is DAS181 or a derivative thereof.
  • the nucleotide sequence encoding the sialidase further encodes a secretion sequence operably linked to the sialidase.
  • the secretion sequence comprises the amino acid sequence of any one of SEQ ID NOs: 40, 101 and 102.
  • the sialidase comprises a transmembrane domain.
  • the sialidase comprises from the N-terminus to the C-terminus: a sialidase catalytic domain, a hinge region, and a transmembrane domain.
  • the sialidase comprises from the N-terminus to the C-terminus: a sialidase catalytic domain, a hinge region, an IgG Fc region, and a transmembrane domain.
  • the hinge region is an IgGl hinge region.
  • the IgG Fc domain comprises the amino acid sequence of SEQ ID NO: 104.
  • the sialidase comprises an immunoglobulin G (IgG) Fc (fragment, crystallizable) domain.
  • the sialidase comprises from the N-terminus to the C-terminus: a sialidase catalytic domain, an IgG Fc domain, and a transmembrane domain.
  • the IgG Fc domain comprises the amino acid sequence of SEQ ID NO: 104.
  • the sialidase comprises an anchoring domain or a transmembrane domain located at the carboxy terminus of the sialidase.
  • the one or more promotors comprise a viral promoter that can be an early promoter, an intermediate promoter, or a late promoter, or an early/late hybrid promoter.
  • the recombinant oncolytic virus is a poxvirus and the promoter is a poxvirus early promoter, a late promoter or a hybrid early/late promoter.
  • the one or more promoters comprise a viral late promoter.
  • the promoter is an F17R late promoter (SEQ ID NO: 61).
  • the one or more promoters comprise a hybrid early-late promoter.
  • the one or more promoters comprise a promoter comprising the nucleotide sequence of SEQ ID NO: 107.
  • the promoter comprises a partial or complete nucleotide sequence of a human promoter.
  • the human promoter is a tissue or tumor-specific promoter.
  • the one or more promotors comprise a first promoter that is operably linked to the first nucleotide sequence and a second promoter that is operably linked to the second nucleotide sequence.
  • the first promoter is an Fl 7R promoter and the second promoter is apE/L promoter.
  • the F17R promoter comprised the nucleic acid sequence of SEQ ID NO: 61.
  • the pE/L promoter comprises the nucleic acid sequence of SEQ ID NO: 107.
  • the recombinant oncolytic virus further comprises an additional nucleotide sequence encoding a heterologous protein or nucleic acid.
  • the additional nucleotide sequence encodes a heterologous protein.
  • the heterologous protein is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is an inhibitor of CTLA-4, PD-1, PD-L1, TIGIT, LAG3, TIM-3, VISTA, B7-H4, or HLA-G.
  • the immune checkpoint inhibitor is an antibody.
  • the heterologous protein is an inhibitor of an immune suppressive receptor.
  • the immune suppressive receptor is LILRB, TYRO3, AXL, or MERTK.
  • the inhibitor of an immune suppressive receptor is an anti-LILRB antibody.
  • the heterologous protein is a multi-specific immune cell engager.
  • the heterologous protein is a bispecific T cell engager (BiTE).
  • the heterologous protein is selected from the group consisting of cytokines, costimulatory molecules, tumor antigen presenting proteins, anti-angiogenic factors, tumor- associated antigens, foreign antigens, and matrix metalloproteases (MMP).
  • the heterologous protein is an inhibitor of CD55 or CD59.
  • the heterologous protein is IL-15, IL-12, IL2, modified IL-2 with reduced toxicity or better function, IL 18, modified IL- 18 with less or no binding to the IL- 18 binding protein, Llt3L, CCL5, CXCL10, or CCL4 and any modified forms of such cytokines that still have the anti-tumor immunity, or an inhibitor of any binding proteins that can block and neutralize these cytokine function and activities.
  • the heterologous protein is a bacterial polypeptide.
  • the heterologous protein is a tumor-associated antigen selected from the group consisting of carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGER (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, R0R1, WT1, NY-ESO- 1, Fibulin-3, CDH17, and other tumor antigens with clinical significance
  • the virus comprises two or more additional nucleotide sequences, wherein each nucleotide sequence encodes a heterologous protein.
  • a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager having the amino acid sequence of SEQ ID NO: 100; wherein the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters; and wherein the recombinant vaccinia virus comprises a disruption or deletion of a thymidine kinase (TK) gene and a disruption or deletion of a vaccinia growth factor (VGF) gene.
  • TK thymidine kinase
  • VVF vaccinia growth factor
  • One aspect of the present application provides a pharmaceutical composition comprising the recombinant oncolytic virus of any one of the preceding claims and a pharmaceutically acceptable carrier.
  • the carrier cell comprising any one of the recombinant oncolytic viruses described above.
  • the carrier cell is an engineered immune cell or a stem cell (e.g., a mesenchymal stem cell) or B cells or leukocytes.
  • the engineered immune cell is a Chimeric Antigen Receptor (CAR)-T, CAR-NK, or CAR-NKT cell.
  • One aspect of the present application provides a method of treating a cancer in an individual in need thereof, comprising administering to the individual an effective amount of any one of the recombinant oncolytic viruses, pharmaceutical compositions, or carrier cells described above.
  • the method comprises administering to the individual an effective amount of any one of the recombinant oncolytic viruses described above.
  • the recombinant oncolytic virus is administered via a carrier cell (e.g., an immune cell or a stem cell, such as a mesenchymal stem cell).
  • the cancer is an FAP positive cancer, or a cancer associated with FAP positive stromal cells (e.g., tumor associated fibroblasts).
  • the cancer is selected from the group consisting of lung cancer, colon cancer, and breast cancer.
  • administering the recombinant oncolytic virus activates and/or expands CD4 + and/or CD8 + T-cells in the individual.
  • administering the recombinant oncolytic virus increases tumor-infiltrating lymphocytes in the individual.
  • the recombinant oncolytic virus is administered as a naked virus. In some embodiments, the recombinant oncolytic virus is administered via direct intratumoral injection. In some embodiments, the method further comprises administering to the individual an effective amount of an immunotherapeutic agent.
  • the immunotherapeutic agent is selected from the group consisting of chemotherapy, radiotherapy, radioimmunotherapy, a mono or multi-specific antibody, a cell therapy, a cancer vaccine (e.g., a dendritic cell-based cancer vaccine), a cytokine, PI3Kgamma inhibitor, a TLR9 ligand, an HDAC inhibitor, a LILRB2 inhibitor, a MARCO inhibitor, and an immune checkpoint inhibitor.
  • a cancer vaccine e.g., a dendritic cell-based cancer vaccine
  • cytokine e.g., a dendritic cell-based cancer vaccine
  • a cytokine e.g., a dendritic cell-based cancer vaccine
  • a cytokine e.g., a dendritic cell-based cancer vaccine
  • a cytokine e.g., a dendritic cell-based cancer vaccine
  • a cytokine e.g., a dendriti
  • the immunotherapeutic agent is a cell therapy.
  • the cell therapy comprises administering to the individual an effective amount of engineered immune cells expressing a chimeric receptor.
  • One aspect of the present application provides a method of treating a cancer in an individual in need thereof, comprising administering to the individual an effective amount of engineered immune cells comprising any one of the recombinant oncolytic viruses described above and expressing a chimeric receptor.
  • One aspect of the present application provides a method of treating a tumor in an individual in need thereof comprising administering to the individual: (a) an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a foreign antigen; and (b) an effective amount of an engineered immune cell expressing a chimeric receptor specifically recognizing said foreign antigen.
  • One aspect of the present application provides a method of sensitizing a tumor to an immunotherapy, comprising administering to the individual an effective amount of any one of the recombinant oncolytic viruses, pharmaceutical compositions, or engineered immune cells described above.
  • One aspect of the present application provides a method of reducing sialylation of cancer cells in an individual, comprising administering to the individual an effective amount of any one of the recombinant oncolytic viruses, pharmaceutical compositions, or engineered immune cells described above.
  • the chimeric receptor is a Chimeric Antigen Receptor (CAR).
  • the engineered immune cells expressing the CAR are T cells, Natural Killer (NK) cells, or NKT cells.
  • the engineered immune cells express a chimeric receptor, wherein the chimeric receptor specifically recognizes one or more tumor antigens selected from the group consisting of carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR1, WT1, NY-ESO-1, Fibulin-3, CDH17, and other tumor antigens with clinical significance
  • the engineered immune cells express a chimeric receptor, wherein the chimeric receptor specifically recognizes the sialidase.
  • the sialidase is DAS 181 or a derivative thereof, and the chimeric receptor comprises an anti-DAS 181 antibody that is not cross-reactive with human native neuraminidase.
  • the engineered immune cells and the recombinant oncolytic virus are administered simultaneously.
  • the recombinant oncolytic virus is administered prior to administration of the engineered immune cells.
  • the method comprises administering any one of the recombinant oncolytic viruses describe above in combination with an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is an inhibitor of CTLA-4, PD-1, PD-L1, B7-H4, TIGIT, LAG3, TIM-3, VISTA, or HLA-G.
  • compositions, kits and articles of manufacture for use in any one the methods described above.
  • Fig. 1 Detection of 2,6 sialic acid (by FITC-SNA) on A549 and MCF cells by fluorescence microscopy. A549 and MCF cells were fixed and incubated with FITC-SNA for one hour at 37°C before imaged under fluorescence microscope to show the FITC-SNA labeled cells (left) and overlay with brightfield cells (right)
  • Fig. 2 Effective removal of 2,6 sialic acid, 2,3 sialic acid, and exposure of galactose on A549 cells by DAS181 treatment.
  • A549 were treated with DAS181 for two hours at 37°C and incubated with staining reagents one hour before imaged under fluorescence microscope to show effective removal of sialic acids on tumor cells.
  • Fig. 3 Effective removal of 2,6 sialic acid on A549 cells by DAS 181 but not DAS 185 treatment. A549 were treated with DAS181 for 30 minutes or two hours at 37°C and incubated with FITC-SNA for one hour before examined using flow cytometry to show effective removal of 2,6 sialic acids on tumor cells.
  • Fig. 4 Effective removal of 2,3 sialic acid on A549 cells by DAS 181 but not DAS 185 treatment. A549 were treated with DAS 181 for 30 minutes or two hours at 37°C and incubated with FITC-MALII for one hour before examined using flow cytometry to show effective removal of 2,3 sialic acids on tumor cells
  • FIG. 5 Effective exposure of galactose on A549 cells by DAS181 but not DAS 185 treatment.
  • A549 were treated with DAS181 for 30 minutes or two hours at 37°C and incubated with FITC-PNA for one hour before examined using flow cytometry to show effective exposure of galactose on tumor cells
  • Fig. 6 DAS 181 treatment and PBMC stimulation regimen do not affect A549-red cell proliferation.
  • A549-Red cells were seeded at 2k/well overnight, followed by replacement of medium containing reagents listed on the left. Scan by IncuCyte was initiated immediately after the reagents were added (0 hr) and scheduled for every 3 hr.
  • A549-red cell proliferation is monitored by analyzing the nuclear (red) counts.
  • Kinetic readouts reveal no effect on A549 cell proliferation by vehicle, DAS 181, or various stimulation reagents, without the presence of PBMCs.
  • Fig. 7 Detection of cytotoxicity in A549-red cells following co-culturing with PBMCs from Donor 1 with or without DAS 181 treatment. These results showed that DAS 181 treatment significantly boost anti-tumor cytotoxicity by PBMCs from Donor 1.
  • Fig. 8 Detection of cytotoxicity in A549-red cells following co-culturing with PBMCs from Donor 2 with or without DAS 181 treatment. These results showed that DAS 181 treatment significantly boost anti-tumor cytotoxicity by PBMCs from Donor 2.
  • Figs. 9A-9C Detection of cytotoxicity in A549-red cells following co-culturing with PBMCs from Donor 1 with or without DAS 181 treatment. These results showed that DAS 181 treatment significantly boost anti -tumor cytotoxicity by PBMCs from Donor 1.
  • A549-red tumor cells were seeded at 2k cells/well in 96-well plate. After overnight incubation, PBMCs from Donor 1 mixed with (A) medium (B) CD3/CD28/IL-2, or (C) CD3/CD28/IL-2/IL-15/IL- 21 were added into each well as indicated E:T ratio. At mean time, DAS 181 (100 nM) was added. Plates were scanned by IncuCyte every 3hr for total 72hrs. Proliferation is monitored by analyzing RFP cell counts.
  • Figs. 10A-10C Detection of cytotoxicity in A549-red cells following co-culturing with PBMCs from Donor 2 with or without DAS 181 treatment. These results showed that DAS 181 treatment significantly boost anti-tumor cytotoxicity by PBMCs from Donor 2.
  • A549- red tumor cells were seeded at 2k cells/well in 96-well plate. After overnight incubation, PBMCs from Donor 2 mixed with (A) medium, (B) CD3/CD28/IL-2, or (C) CD3/CD28/IL- 2/IL-l 5/IL-21 were added into each well as indicated E:T ratio. At mean time, DAS 181 (100 nM) was added. Plates were scanned by IncuCyte every 3hr for total 72hrs. Proliferation is monitored by analyzing RFP cell counts.
  • Fig. 11 DAS 181 enhances NK-mediated tumor lysis by vaccinia virus, measured by MTS assay.
  • ® T-test P value ⁇ 0.05, suggesting that DAS181 alone boosts NK cell-mediated U87 tumor killing in vitro, compared to enzyme-dead DAS 185.
  • * T-Test P value ⁇ 0.05.
  • Fig. 15 DAS181 treatment promotes oncolytic adenovirus-mediated tumor cell killing and growth prohibition. A549-red tumor cells were seeded at 2K cells/well in 96-well plates. After overnight incubation, DAS 181 vehicle, oncolytic adenovirus, and DAS181 were added as indicated. CD3/CD28/IL-2 were also added into each well with the amount described previously. Graph showed that DAS 181 plus oncolytic adenovirus effectively reduced tumor cell proliferation.
  • Figs. 16A-16B DAS 181 treatment enhances PBMC -mediated tumor cell killing by oncolytic virus. A549-red tumor cells were seeded at 2K cells/well in 96-well plate. After overnight incubation, fresh PBMCs were added at densities of lOK/well (A) or 40K/well (B). CD3, CD28, IL-2, DAS181, and oncolytic adenovirus were added as indicated in the graph following with the timed scans by IncuCyte. Graph showed that DAS 181 plus oncolytic adenovirus dramatically enhanced human PBMC-mediated tumor cell eradication.
  • Fig. 17 Schematic of a portion of a vaccinia vims construct encoding a sialidase.
  • Figs. 18A-18B DAS181 expressed by Sialidase-VV has in vitro activity towards sialic acid-containing substrates.
  • A Standard curve of DAS181 activity at 0.5 nM, 1 nM, and 2 nM.
  • B IxlO 6 cells infected with Sialidase-VV express DAS 181 equivalent to 0.78nM - 1.21 nM DAS181 in 1ml medium in vitro.
  • Fig. 19 Sialidase-VV enhances Dendritic cell maturation.
  • Fig. 20 Sialidase-VV induced IFN-gamma and IL2 expression by T cells.
  • CD3 antibody-activated human T cells were co-cultured with A594 tumor cells in the presence of Sial-VV- or VV-infected tumor cells lysate for 24 hours, and cytokine IFNr or IL-2 expression was measured by ELISA. The results suggested that Sial -VV-infected tumor cell lysate induced IFNr and IL2 expression by human T cells.
  • * T-test P value ⁇ 0.05
  • FIGS. 22A-22C Impact of DAS 181 and secreted sialidase Constructs 1, 2, and 3 on cell surface a2,3 sialic acid (FIG. 22A); a2,6 sialic acid (FIG. 22B) and galactose (FIG. 22C).
  • FIG. 22A Impact of DAS 181 and secreted sialidase Constructs 1, 2, and 3 on cell surface a2,3 sialic acid (FIG. 22A); a2,6 sialic acid (FIG. 22B) and galactose (FIG. 22C).
  • FIG. 22A A549-red cells were transfected by Construct- 1, 2 or 3. After overnight incubation, transfected cells were lifted and re-seeded in 24-well plate. After additional 24hrs, 48hrs and 72hrs, cells were fixed and stained with MALII-FITC for Jackpot before performing flow. Treat non-transfected cells with 1 OOnM DAS 181 for 2hrs before fixed. Vehicle prepared for DAS 181 was used to treat another set of non-transfected cells as control.
  • FIG. 22B A549-red cells were transfected by Construct- 1, 2 and 3. After overnight incubation, transfected cells were lifted and re-seeded in 24-well plate.
  • FIG. 22C A549-red cells were transfected by Construct- 1, 2 and 3. After overnight incubation, transfected cells were lifted and re-seeded in 24-well plate. After additional 24hrs, 48hrs and 72hrs, cells were fixed and stained with PNA-FITC for Jackpot before performing flow. Treat non-transfected cells with lOOnM DAS181 for 2hrs before fixed. Vehicle prepared for DAS 181 was used to treat another set of non-transfected cells as control.
  • FIGS. 23A-23C Impact of DAS 181 and transmembrane sialidase Constructs 1, 4, 5 and 6 on cell surface a2,3 sialic acid (FIG. 23A); a2,6 sialic acid (FIG. 23B); and galactose (FIG. 23C).
  • FIG. 23A A549-red cells were transfected by Construct-1, 4, 5, and 6. After overnight incubation, transfected cells were lifted and re-seeded in 24-well plate. After additional 24hrs, 48hrs and 72hrs, cells were fixed and stained with MALII-Biotinylated for Ice followed by FITC-streptavidin for an additional Bit.
  • FIG. 23B A549-red cells were transfected by Construct-1, 4, 5, and 6. After overnight incubation, transfected cells were lifted and re-seeded in 24-well plate. In additional 24hrs, 48hrs and 72hrs, cells were fixed and stained with SNA-FITC for Jackpot. The 2, 6-sialic acid level was detected by flow cytometry.
  • FIG. 23C A549-red cells were transfected by Construct- 1, 4, 5, and 6. After overnight incubation, transfected cells were lifted and reseeded in 24-well plate. After additional 24hrs, 48hrs and 72hrs, cells were fixed and stained with PNA-FITC for Jackpot. The galactose level was detected by flow cytometry.
  • FIG. 24 Stable expression of Construct 1 increases oncolytic virus and PBMC- mediated A549 cell killing. Freshly isolated PBMCs were incubated with A549-red parental cells only or with cells stable expressing Construct- 1 or cells stable expressing Construct- 1 with 1MOI or 5MOI on two separated plates (Plate 2 and 4).
  • FIG. 25 Stable expression of Construct 4 increases oncolytic virus and PBMC- mediated A549 cell killing. Fresh isolated PBMCs were activated and incubated with A549- red cells only or with cells stable expressing Construct-4 or cells stable expressing Construct- 4 with 1MOI or 5MOI OL in two separated plates (Plate 2 and 4).
  • FIG. 26 Design of exemplary sialidase expression constructs for recombination into the TK gene of Western Reserve VV to generate oncolytic virus encoding a sialidase. Exemplary constructs are shown for endocellular sialidase, secreted sialidase with an anchoring domain, and cell surface expressed sialidase with a transmembrane domain.
  • FIG. 27 PCR detection of Sialidase expression: CV-1 cells were infected with Sialidase-VV at an MOI of 0.2. After 48 hours, CV-1 cells were collected, and DNA were extracted using Wizard® SV Genomic DNA Purification System and used as template for Sialidase PCR amplification. PCR was conducted using standard PCR protocol. Expected PCR product size is 125 Ibp. [0095] FIG. 28: U87 or CV-1 cells were infected with control VV, SP-, Endo- or TM-Sial- VVs at MOI 1. The cells were collected at 24, 48, 72, or 96 hours. Virus titers were determined by plaque assay.
  • FIG. 29 U87 tumor cells were infected with control VV, SP-, Endo- or TM-Sial- VVs at MOI 0.1, 1, or 5. Tumor killing was measured by MTS assay.
  • FIG. 30 The expression of DC maturation marker HLA-ABC is enhanced by culture with oncolytic virus encoding secreted or transmembrane sialidase.
  • FIG. 31 The expression of DC maturation marker HLA-DR is enhanced by culture with oncolytic virus encoding secreted or transmembrane sialidase.
  • FIG. 32 The expression of DC maturation marker CD80 is enhanced by culture with oncolytic virus encoding secreted or transmembrane sialidase.
  • FIG. 33 The expression of DC maturation marker CD86 is enhanced by culture with oncolytic virus encoding secreted or transmembrane sialidase.
  • FIG. 34 Sial-VV enhances NK-mediated tumor lysis in vitro. Negative selected human NK cells (Astarte, WA) and VV-U87 cells (ATCC, VA) were co-cultured, and tumor killing efficacy was measured by LDH assay (Abeam, MA). The results suggested that Sial- VVs enhanced NK cell-mediated U87 tumor killing in vitro. (* P value, the Sial-VV vs Mock VV in U87 and NK culture).
  • FIG. 35 Results indicate that TM-sial-VV significantly inhibited tumor growth compared to control VV in vivo (tumor cells inoculated in right flank of mouse).
  • FIG. 36 Results indicate that TM-sial-VV significantly inhibited tumor growth compared to control VV in vivo (tumor cells inoculated in left flank of mouse).
  • FIG. 37 Mouse body weight was unaffected by treatment with Sial-VV or VV The results didn’t show the difference on the mouse body weight.
  • FIGS. 38A-38B Sialidase armed oncolytic vaccinia virus significantly enhanced CD8+ and CD4+ T cell infiltration within tumor. * p value: treatment group vs control VV group.
  • FIG. 38A shows quantification of the results.
  • FIG. 38B shows the FACS plots.
  • FIG. 39 TM-Sial-VV decreased the ratio of Treg/CD4+ T cells within the tumor, compared to control VV. * p value: treatment group vs control VV group.
  • FIG. 40 Sialidase armed oncolytic vaccinia virus significantly enhanced NK and NKT cell infiltration within tumor. * p value: treatment group vs control VV group.
  • FIG. 41 TM-Sial-VV significantly increased PD-L1 expression within tumor cells (p ⁇ 0.05).
  • FIGS. 42A-42B Results indicating that an exemplary oncolytic vims comprising a nucleotide sequence encoding a sialidase and a bispecific immune cell engager (vvDD-Sial- FAP/CD3) reduced the level of a-2,6-sialic acid linkages on cell surface.
  • FIG. 43 Results indicating that vvDD-Sial-FAP/CD3 induced antibody-dependent cellular cytotoxicity (ADCC) for A549 in the presence of Jurkat effector T cells.
  • FIGS. 44A-44B Conditioned media from A549 cells infected with vvDD-Sial- FAP/CD3 induced increased T-cell activation in the presence of Jurkat cells for FAP-positive COLO829 colon cancer cells (FIG 44A) compared to FAP -negative A549 cells (FIG. 44B).
  • FIG. 45 Infection of HCT116 human colon cancer cells mixed with FAP- expressing normal human dermal fibroblasts with vvDD-Sial-FAP/CD3 resulted in significantly higher LDH compared to mock infected or vvDD infected cells in the presence ofPBMCs.
  • FIGS. 46A-46C A549 human lung adenocarcinoma cells were co-cultured with normal human dermal fibroblasts (NhDF) were mock-infected or infected with vvDD or wDD-Sial-FAP/CD3 at 0.1 pfii/cell. The next day, peripheral blood mononuclear cells (PBMCs) were added at 10: 1 (EffcctorTargct). One day later, cells were harvested and analyzed for the expression of CD25 activation marker on CD4+ (FIG. 46A) and CD8+ (FIG. 46B) T cells using flow cytometry. Supernatants were analyzed for granzyme B release using ELISA (FIG. 46C).
  • PBMCs peripheral blood mononuclear cells
  • FIGS. 47A-47B HCT116 human colon cancer cells co-cultured with normal human dermal fibroblasts (NhDF) or HCC1143 human breast cancer cells were mock- infected or infected with vvDD or vvDD-Sial-FAP/CD3 at 0.1 pfii/cell. The next day, peripheral blood mononuclear cells (PBMCs) were added at 10: 1 (Effector: Target). Two days later, cells were harvested and analyzed for the expression of activation markers, CD69 and CD25, on CD4+ (FIG. 47A) and CD8+ (FIG. 47B) T cells using flow cytometry [0115] FIG.
  • PBMCs peripheral blood mononuclear cells
  • A549 human lung adenocarcinoma cells co-cultured with cancer- associated fibroblasts (CAFs) were mock-infected or infected with vvDD or wDD-Sial- FAP/CD3 expressing green or yellow fluorescent protein, respectively, at 0.3 pfii/cell.
  • CAFs cancer- associated fibroblasts
  • the expression of either GFP or YFP was monitor by imaging. Increase in the intensity of the fluorescent proteins indicates the spread of the virus within tumor spheroids.
  • FIG. 49 A549 human lung adenocarcinoma cells co-cultured with cancer- associated fibroblasts (CAFs) were mock-infected or infected with vvDD or wDD-Sial- FAP/CD3 at 0.3 pfii/cell. The next day, peripheral blood mononuclear cells (PBMCs) labeled by CellTracker DeepRed were added at 10: 1 (Effector: Target). Images taken two days after addition of PBMCs are shown. Infection with vvDD-Sial-FAP/CD3 resulted in the increase of red fluorescence in the core of the tumor spheroid, indicating enhanced tumor-infiltrating lymphocytes.
  • CAFs cancer- associated fibroblasts
  • the present application provides compositions and methods for treating cancers with an oncolytic virus (e.g., vaccinia virus) encoding a sialidase.
  • an oncolytic virus e.g., vaccinia virus
  • the recombinant oncolytic viruses described herein are capable of delivering sialidase to tumor cells and/or the tumor cell environment.
  • the delivered sialidase can reduce sialic acid present on tumor cells or immune cells and render the tumor cells more vulnerable to killing by immune cells, immune cell-based therapies and/or other therapeutic agents whose effectiveness is diminished by hypersialylation of cancer cells.
  • the delivered sialidase reduces or prevents binding of Siglects on immune cells with their inhibitory receptor ligands (sialylated glycoconjugates).
  • the delivered sialidase reduces or abolishes suppression of immunity against tumor cells.
  • the delivered sialidase e.g., a bacterial sialidase
  • the recombinant oncolytic virus is delivered via carrier cells (e.g., engineered immune cells or stem cells) expressing the virus.
  • the method further comprises administering engineered immune cells that enhance the anti-tumor effect of the recombinant oncolytic virus (e.g., by expressing a chimeric receptor targeting a foreign antigen, such as a sialidase, delivered by the recombinant oncolytic virus).
  • engineered immune cells that enhance the anti-tumor effect of the recombinant oncolytic virus (e.g., by expressing a chimeric receptor targeting a foreign antigen, such as a sialidase, delivered by the recombinant oncolytic virus).
  • treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: decreasing one more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), preventing or delaying the spread of the disease, preventing or delaying the occurrence or recurrence of the disease, delay or slowing the progression of the disease, ameliorating the disease state, providing a remission (whether partial or total) of the disease, decreasing the dose of one or more other medications required to treat the disease, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival.
  • treatment is a reduction of pathological consequence of the disease. The methods of the present application contemplate any one or more of these aspects of treatment
  • the terms “individual,” “subject” and “patient” are used interchangeably herein to describe a mammal, including humans.
  • the individual is human.
  • an individual suffers from a cancer.
  • the individual is in need of treatment.
  • an “effective amount” refers to an amount of a composition sufficient to produce a desired therapeutic outcome (e.g., reducing the severity or duration of, stabilizing the severity of, or eliminating one or more symptoms of cancer).
  • beneficial or desired results include, e.g., decreasing one or more symptoms resulting from the disease (biochemical, histologic and/or behavioral), including its complications and intermediate pathological phenotypes presented during development of the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, and/or prolonging survival of patients.
  • an effective amount of the therapeutic agent may extend survival (including overall survival and progression free survival); result in an objective response (including a complete response or a partial response); relieve to some extent one or more signs or symptoms of the disease or condition; and/or improve the quality of life of the subject.
  • wild type is a term of the art understood by skilled persons and means the typical form of an organism, strain, gene or characteristic as it occurs in nature as distinguished from mutant or variant forms.
  • nucleic acid molecules or polypeptides mean that the nucleic acid molecule or the polypeptide is at least substantially free from at least one other component with which they are naturally associated in nature and as found in nature.
  • sialidase refers to a naturally occurring or engineered sialidase that is capable of catalyzing the cleavage of terminal sialic acids from carbohydrates on glycoproteins or glycolipids.
  • sialidase can refer to a domain of a naturally occurring or non-naturally occurring sialidase that is capable of catalyzing cleavage of terminal sialic acids from carbohydrates on glycoproteins or glycolipids.
  • sialidase also encompasses fusion proteins comprising a naturally occurring or non-naturally occurring sialidase protein or an enzymatically active fragment or domain thereof and another polypeptide, fragment or domain thereof, e.g., an anchoring domain or a transmembrane domain.
  • sialidase as used herein encompasses sialidase catalytic domain proteins.
  • a "sialidase catalytic domain protein” is a protein that comprises the catalytic domain of a sialidase, or an amino acid sequence that is substantially homologous to the catalytic domain of a sialidase, but does not comprise the entire amino acid sequence of the sialidase.
  • the catalytic domain is derived from, wherein the sialidase catalytic domain protein retains substantially the functional activity as the intact sialidase the catalytic domain is derived from.
  • a sialidase catalytic domain protein can comprise amino acid sequences that are not derived from a sialidase.
  • a sialidase catalytic domain protein can comprise amino acid sequences that are derived from or substantially homologous to amino acid sequences of one or more other known proteins, or can comprise one or more amino acids that are not derived from or substantially homologous to amino acid sequences of other known proteins.
  • expression refers to the process by which a polynucleotide is transcribed from a DNA template (such as into an mRNA or other RNA transcript) and/or the process by which a transcribed mRNA is subsequently translated into peptides, polypeptides, or proteins.
  • Transcripts and encoded polypeptides may be collectively referred to as “gene product.” If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell.
  • antibody is used in its broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies, trispecific antibodies, etc ), humanized antibodies, chimeric antibodies, full-length antibodies and antigen-binding fragments, single chain Fv, nanobodies, Fc fusion proteins, thereof, so long as they exhibit the desired antigenbinding activity.
  • Antibodies and/or antibody fragments may be derived from murine antibodies, rabbit antibodies, chicken antibodies, human antibodies, fully humanized antibodies, camelid antibody variable domains and humanized versions, shark antibody variable domains and humanized versions, and camelized antibody variable domains.
  • virus or “virus particle” are used according to its plain ordinary meaning within Virology and refers to a virion including the viral genome (e.g. DNA, RNA, single strand, double strand), viral capsid and associated proteins, and in the case of enveloped viruses (e.g. herpesvirus, poxvirus), an envelope including lipids and optionally components of host cell membranes, and/or viral proteins.
  • viral genome e.g. DNA, RNA, single strand, double strand
  • enveloped viruses e.g. herpesvirus, poxvirus
  • oncolytic viruses refer to viruses that selectively replicate in and selectively kill tumor cells in subjects having a tumor. These include viruses that naturally preferentially replicate and accumulate in tumor cells, such as poxviruses, and viruses that have been engineered to do so. Some oncolytic viruses can kill a tumor cell following infection of the tumor cell. For example, an oncolytic virus can cause death of the tumor cell by lysing the tumor cell or inducing cell death of the tumor cell.
  • Exemplary oncolytic viruses include, but are not limited to, poxviruses, herpesviruses, adenoviruses, adeno-associated viruses, lentiviruses, retroviruses, rhabdoviruses, papillomaviruses, vesicular stomatitis virus, measles virus, Newcastle disease virus, picomavirus, Sindbis virus, papillomavirus, parvovirus, reovirus, and coxsackievirus.
  • poxvirus is used according to its plain ordinary meaning within Virology and refers to a member of Poxviridae family capable of infecting vertebrates and invertebrates which replicate in the cytoplasm of their host.
  • poxvirus virions have a size of about 200 nm in diameter and about 300 nm in length and possess a genome in a single, linear, double-stranded segment of DNA, typically 130-375 kilobase.
  • poxvirus includes, without limitation, all genera of poxviridae (e.g., betaentomopoxvirus, yatapoxvirus, cervidpoxvirus, gammaentomopoxvirus, leporipoxvirus, suipoxvirus, molluscipoxvirus, crocodylidpoxvirus, alphaentomopoxvirus, capripoxvirus, orthopoxvirus, avipoxvirus, and parapoxvirus).
  • poxviridae e.g., betaentomopoxvirus, yatapoxvirus, cervidpoxvirus, gammaentomopoxvirus, leporipoxvirus, suipoxvirus, molluscipoxvirus, crocodylidpoxvirus, alphaentomopoxvirus, capripoxvirus, orthopoxvirus, avipoxvirus, and parapoxvirus).
  • the poxvirus is an orthopoxvirus (e.g., smallpox virus, vaccinia virus, cowpox virus, monkeypox virus), parapoxvirus (e.g., orf virus, pseudocowpox virus, bovine popular stomatitis virus), yatapoxvirus (e.g., tanapox virus, yaba monkey tumor virus) or molluscipoxvirus (e.g., molluscum contagiosum virus).
  • orthopoxvirus e.g., smallpox virus, vaccinia virus, cowpox virus, monkeypox virus
  • parapoxvirus e.g., orf virus, pseudocowpox virus, bovine popular stomatitis virus
  • yatapoxvirus e.g., tanapox virus, yaba monkey tumor virus
  • molluscipoxvirus e.g., molluscum contagiosum virus
  • the poxvirus is an orthopoxvirus (e.g., cowpox virus strain Brighton, raccoonpox virus strain Herman, rabbitpox virus strain Utrecht, vaccinia virus strain WR, vaccinia virus strain IHD, vaccinia virus strain Elstree, vaccinia virus strain CL, vaccinia virus strain Lederle-Chorioallantoic, or vaccinia virus strain AS).
  • the poxvirus is a parapoxvirus (e.g., orf virus strain NZ2 or pseudocowpox virus strain TJS).
  • a “modified virus” or a “recombinant virus” refers to a virus that is altered in its genome compared to a parental strain of the virus.
  • modified viruses have one or more truncations, substitutions (replacement), mutations, insertions (addition) or deletions (truncation) of nucleotides in the genome of a parental strain of virus.
  • a modified virus can have one or more endogenous viral genes modified and/or one or more intergenic regions modified.
  • Exemplary modified viruses can have one or more heterologous nucleotide sequences inserted into the genome of the virus.
  • Modified viruses can contain one or more heterologous nucleotide sequences in the form of a gene expression cassette for the expression of a heterologous gene. Modifications can be made using any method known to one of skill in the art, including as provided herein, such as genetic engineering and recombinant DNA methods.
  • Percent (%) amino acid sequence identity with respect to the polypeptide and antibody sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the polypeptide being compared, after aligning the sequences considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, Megalign (DNASTAR), or MUSCLE software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program MUSCLE (Edgar, R.C., Nucleic Acids Research 32(5): 1792-1797, 2004; Edgar, R.C., BMC Bioinformatics 5(1): 113, 2004, each of which are incorporated herein by reference in their entirety for all purposes).
  • epitope refers to the specific group of atoms or amino acids on an antigen to which an antibody or diabody binds. Two antibodies or antibody moieties may bind the same epitope within an antigen if they exhibit competitive binding for the antigen.
  • polypeptide or “peptide” are used herein to encompass all kinds of naturally occurring and synthetic proteins, including protein fragments of all lengths, fusion proteins and modified proteins, including without limitation, glycoproteins, as well as all other types of modified proteins (e.g., proteins resulting from phosphorylation, acetylation, myristoylation, palmitoylation, glycosylation, oxidation, formylation, amidation, polyglutamylation, ADP-ribosylation, pegylation, biotinylation, etc.).
  • modified proteins e.g., proteins resulting from phosphorylation, acetylation, myristoylation, palmitoylation, glycosylation, oxidation, formylation, amidation, polyglutamylation, ADP-ribosylation, pegylation, biotinylation, etc.
  • the terms “specifically binds,” “specifically recognizing,” and “is specific for” refer to measurable and reproducible interactions, such as binding between a target and an antibody (such as a diabody).
  • specific binding is determinative of the presence of the target in the presence of a heterogeneous population of molecules, including biological molecules (e.g., cell surface receptors).
  • an antibody that specifically recognizes a target is an antibody (such as a diabody) that binds this target with greater affinity, avidity, more readily, and/or with greater duration than its bindings to other molecules.
  • the extent of binding of an antibody to an unrelated molecule is less than about 10% of the binding of the antibody to the target as measured, e.g., by a radioimmunoassay (RIA).
  • an antibody that specifically binds atarget has a dissociation constant (KD) of ⁇ 10' 5 M, ⁇ 10' 6 M, ⁇ 10' 7 M, ⁇ 10‘ 8 M, ⁇ 10' 9 M, ⁇ 1O' 10 M, ⁇ 10' n M, or ⁇ 10' 12 M.
  • KD dissociation constant
  • an antibody specifically binds an epitope on a protein that is conserved among the protein from different species.
  • specific binding can include, but does not require exclusive binding.
  • Binding specificity of the antibody or antigen-binding domain can be determined experimentally by methods known in the art. Such methods comprise, but are not limited to Western blots, ELISA, RIA, ECL, IRMA, EIA, BIACORETM and peptide scans.
  • the term “simultaneous administration,” as used herein, means that a first therapy and second therapy in a combination therapy are administered with a time separation of no more than about 15 minutes, such as no more than about any of 10, 5, or 1 minutes.
  • the first and second therapies may be contained in the same composition (e.g., a composition comprising both a first and second therapy) or in separate compositions (e.g., a first therapy in one composition and a second therapy is contained in another composition).
  • the term “sequential administration” means that the first therapy and second therapy in a combination therapy are administered with a time separation of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, 60, or more minutes. Either the first therapy or the second therapy may be administered first.
  • the first and second therapies are contained in separate compositions, which may be contained in the same or different packages or kits.
  • the term “concurrent administration” means that the administration of the first therapy and that of a second therapy in a combination therapy overlap with each other.
  • composition refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to one or more ingredients in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, cryoprotectant, tonicity agent, preservative, and combinations thereof.
  • Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration or other state/federal government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in mammals, and more particularly in humans.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • An “article of manufacture” is any manufacture (e.g., a package or container) or kit comprising at least one reagent, e.g., a medicament fortreatment of a disease or condition (e.g., cancer), or a probe for specifically detecting a biomarker described herein.
  • the manufacture or kit is promoted, distributed, or sold as a unit for performing the methods described herein.
  • Reference to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
  • reference to “not” a value or parameter generally means and describes "other than” a value or parameter.
  • the method is not used to treat disease of type X means the method is used to treat disease of types other than X.
  • the term “and/or” as used herein a phrase such as “A and/or B” is intended to include both A and B; A or B; A (alone); and B (alone).
  • the term “and/or” as used herein a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
  • the present application provides recombinant oncolytic viruses for treating a cancer in an individual in need thereof.
  • the present application provides a recombinant oncolytic virus comprising a nucleotide sequence encoding a sialidase.
  • the nucleotide sequence encoding the sialidase is operably linked to a promoter.
  • the recombinant oncolytic virus further comprises a second nucleotide sequence encoding a heterologous protein or nucleic acid.
  • the present application provides a recombinant oncolytic virus comprising a first nucleotide sequence encoding a sialidase and a second nucleotide sequence encoding a heterologous protein or nucleic acid, wherein the first nucleotide sequence is operably linked to a promoter and the second nucleotide sequence is operably linked to a promoter.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to the same promoter.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to different promoters.
  • the recombinant oncolytic virus comprises two or more nucleotide sequences, wherein each nucleotide sequence encodes a heterologous protein or nucleic acid.
  • the second nucleotide sequence encodes a heterologous protein selected from the group consisting of immune checkpoint inhibitors, inhibitors of immune suppressive receptors, multi-specific immune cell engager (e.g., a BiTE), cytokines, costimulatory molecules, tumor antigen presenting proteins, anti-angiogenic factors, tumor-associated antigens, foreign antigens, and matrix metalloproteases (MMP), Regulatory molecules of Macrophage or monocyte functions (antibodies to LILRBs), antibodies to folate receptor beta, tumor cell specific antigens (CD 19, CDH17, etc) or antibodies to tumor scaffold (FAP, fibulin- 3, etc).
  • the oncolytic virus is a virus selected from the group consisting of: vaccinia virus, reovirus, Seneca Valley virus (SVV), vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), herpes simplex virus (HSV), morbillivirus virus, retrovirus, influenza virus, Sinbis virus, poxvirus, measles virus, cytomegalovirus (CMV), lentivirus, adenovirus (Ad), and derivatives thereof.
  • the oncolytic virus is modified to reduce immunogenicity of the virus. Suitable oncolytic viruses and derivatives thereof are described in the ''Oncolytic Viruses" subsection below.
  • a recombinant vaccinia virus comprising a first nucleotide sequence encoding a sialidase, wherein the first nucleotide sequence is operably linked to a promoter.
  • the vaccinia virus further comprises a second nucleotide encoding a heterologous protein, e.g., an immune checkpoint inhibitor, an inhibitor of an immune suppressive receptor, a cytokine, a costimulatory molecule, a tumor antigen presenting protein, an anti-angiogenic factor, a tumor-associated antigen, a foreign antigen, or a matrix metalloprotease (MMP), Regulatory molecules of Macrophage or monocyte functions (antibodies to LILRBs), antibodies to folate receptor beta, tumor cell specific antigens (CD 19, CDH17, etc) or antibodies to tumor scaffold (FAP, fibulin-3, etc) wherein the second nucleotide sequence is operably linked to the same or a different promoter.
  • a heterologous protein e.g., an immune checkpoint inhibitor, an inhibitor of an immune suppressive receptor, a cytokine, a costimulatory molecule, a tumor antigen presenting protein, an anti-angiogenic factor, a
  • the virus is vaccinia virus Western Reserve.
  • the virus is a vaccinia virus
  • the one or more mutations are in one or more proteins selected from the group consisting of A14, A17, A13, LI, H3, D8, A33, B5, A56, F13, A28, and A27.
  • the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R.
  • a recombinant vaccinia virus comprising a first nucleotide sequence encoding a sialidase, wherein the first nucleotide sequence is operably linked to a promoter.
  • the vaccinia virus further comprises a second nucleotide encoding a heterologous protein, wherein the heterologous protein is a membranebound complement activation modulator such as CD55, CD59, CD46, CD35, factor H, C4- binding protein, or other identified complement activation modulators, and wherein the second nucleotide sequence is operably linked to the same or a different promoter.
  • the virus is vaccinia virus Western Reserve.
  • the virus is a vaccinia virus
  • the one or more mutations are in one or more proteins selected from the group consisting of A14, A17, A13, LI, H3, D8, A33, B5, A56, F13, A28, and A27.
  • the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R.
  • the present application provides recombinant oncolytic viruses (e.g., vaccinia virus) encoding heterologous proteins or nucleic acids as described below.
  • the recombinant oncolytic virus encodes a sialidase.
  • the sialidase is human or bacterial sialidase.
  • the sialidase is a secreted sialidase.
  • the sialidase comprises a membrane anchoring moiety or a transmembrane domain. Suitable sialidases and derivatives or variants thereof are described in the “Sialidase” subsection below.
  • the recombinant oncolytic virus encodes one or more heterologous proteins or nucleic acids that promote an immune response or inhibit an immune suppressive protein, as described in the "Other heterologous proteins or nucleic acids" subsection below.
  • the recombinant oncolytic virus encodes a multispecific immune cell engager. Suitable multispecific immune cell engagers are described in the “Multispecific immune cell engager” subsection below. In some embodiments, the multispecific immune cell engager recognizes FAP and CD3.
  • a recombinant oncolytic viruses comprising a first nucleotide sequence encoding a Actinomyces viscosus sialidase or a derivative thereof, wherein the first nucleotide sequence is operably linked to a promoter.
  • the oncolytic virus further comprises a second nucleotide sequence encoding a multispecific immune cell engager.
  • the oncolytic virus further comprises an additional nucleotide sequence encoding a heterologous protein (e.g, an immune checkpoint inhibitor, an inhibitor of an immune suppressive receptor, a cytokine, a costimulatory molecule, a tumor antigen presenting protein, an anti-angiogenic factor, a tumor-associated antigen, a foreign antigen, or a matrix metalloprotease (MMP)).
  • a heterologous protein e.g, an immune checkpoint inhibitor, an inhibitor of an immune suppressive receptor, a cytokine, a costimulatory molecule, a tumor antigen presenting protein, an anti-angiogenic factor, a tumor-associated antigen, a foreign antigen, or a matrix metalloprotease (MMP)
  • MMP matrix metalloprotease
  • the recombinant oncolytic virus is an enveloped virus (e.g., vaccinia virus) and the heterologous protein is a membrane-bound complement activation modulator such as CD55, CD59, CD46, CD35, factor H, C4-binding protein, or other identified complement activation modulators.
  • the sialidase comprises an amino acid sequence having at least about 80% sequence identity to the amino acid sequence of SEQ ID NO: 1 or 26.
  • a recombinant oncolytic viruses e.g., vaccinia virus
  • a sialidase comprising an anchoring domain (e.g., DAS 181).
  • the oncolytic virus further comprises a second nucleotide sequence encoding a heterologous protein or nucleic acid.
  • the anchoring domain is a glycosaminoglycan (GAG)-binding domain.
  • the anchoring domain is positively charged at physiologic pH.
  • the anchoring domain is located at the carboxy terminus of the sialidase.
  • the sialidase is derived from a Actinomyces viscosus sialidase. In some embodiments, the sialidase is DAS181. In some embodiments, the nucleotide sequence encoding the sialidase further encodes a secretion sequence operably linked to the sialidase. In some embodiments, the secretion sequence is operably linked to the amino terminus of the sialidase. [0158] In some embodiments, there is provided a recombinant oncolytic viruses (e.g., vaccinia virus) encoding a sialidase comprising a transmembrane domain.
  • vaccinia virus e.g., vaccinia virus
  • the transmembrane domain comprises an amino acid sequence selected from SEQ ID NOs: 45-52.
  • the oncolytic virus further comprises a second nucleotide sequence encoding a heterologous protein or nucleic acid.
  • the sialidase is derived from a Actinomyces viscosus sialidase.
  • the nucleotide sequence encoding the sialidase further encodes a secretion sequence operably linked to the sialidase.
  • the nucleotide sequence encoding the heterologous protein or nucleic acid is operably linked to a promoter.
  • the promoter is a viral promoter, such as an early, late, or early/late viral promoter.
  • the promoter is a hybrid promoter.
  • the promoter is comprises a promoter sequence of a human promoter (e.g. , a tissue- or tumor- specific promoter) . Suitable promoters are described in the "Promoters for expression of heterologous proteins or nucleic acids" subsection below.
  • the present application further provides engineered immune cells for treatment of a cancer in an individual in need thereof.
  • the engineered immune cells comprise chimeric receptors that specifically recognize a tumor antigen.
  • the engineered immune cells comprise chimeric receptors that specifically recognize a foreign antigen (e.g., a bacterial sialidase) encoded by any one of the recombinant oncolytic viruses described herein. Suitable engineered immune cells are described in the “Engineered immune cells” subsection below.
  • composition comprising an engineered immune cell comprising a recombinant oncolytic virus encoding a sialidase and a multispecific immune cell engager (e.g., a multispecific immune cell engager that recognizes FAP and CD3).
  • a recombinant oncolytic virus is a vaccinia virus.
  • the vaccinia virus is a Western Reserve strain.
  • the vaccinia virus is a modified vaccinia virus (e.g., a vaccinia virus comprising one or more mutations, wherein the mutations are in one or more proteins such as A 14, A 17, A 13, LI, H3, D8, A33, B5, A56, F13, or A28).
  • the sialidase is derived from a Actinomyces viscosus sialidase.
  • the sialidase is DAS181.
  • the nucleotide sequence encoding the sialidase further encodes a secretion sequence operably linked to the sialidase.
  • the sialidase further comprises a transmembrane domain.
  • the engineered immune cell encodes a chimeric receptor. In some embodiments, the chimeric receptor is a chimeric antigen receptor. In some embodiments, the engineered immune cell is a cytotoxic T cell, a helper T cell, a suppressor T cell, an NK cell, and an NK-T cell. In some embodiments, the engineered immune cell is an autologous cell of a patient or an allogeneic cell.
  • composition comprising a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having at least any one of 85%, 90%, 95%, or 100% identity to the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager having at least any one of 85%, 90%, 95%, or 100% identity the amino acid sequence of SEQ ID NO: 100.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • composition comprising a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having at least any one of 85%, 90%, 95%, or 100% identity to the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager comprising a first scFv that recognizes FAP and a second scFv that recognizes CD3.
  • the first scFv comprises: (i) a first light chain complementaritydetermining region (CDR-L1) having the amino acid sequence of SEQ ID NO: 86, (i) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 87, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 88, (iv) a first heavy chain complementaritydetermining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 89, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 90, and (vi) a third heavy chain complementarity-determining region (CDR- H3) having the amino acid sequence of SEQ ID NO: 91.
  • CDR-L1 first light chain complementaritydetermining region having the amino acid sequence of SEQ ID NO: 86
  • CDR-L2 having the amino acid sequence
  • the first scFv comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 98. In some embodiments, the first scFv comprises the amino acid sequence of SEQ ID NO: 98.
  • the second scFv comprises: (i) a first light chain complementarity-determining region (CDR-L1) having the amino acid sequence of SEQ ID NO: 92, (ii) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 93, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 94, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 95, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 96, and (vi) a third heavy chain complementarity-determining region (CDR-H3) having the amino acid sequence of SEQ ID NO: 97.
  • CDR-L1 having the amino acid sequence of SEQ ID NO: 92
  • CDR-L2 having the amino acid sequence of SEQ ID NO
  • the second scFv comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 99. In some embodiments, the second scFv comprises the amino acid sequence of SEQ ID NO: 99. In some embodiments, the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters. In some embodiments, the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • TK thymidine kinase
  • VVF vaccinia growth factor
  • composition comprising a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having at least any one of 85%, 90%, 95%, or 100% identity to the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager comprising a first scFv that recognizes FAP and a second scFv that recognizes CD3.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • composition comprising a recombinant vaccinia virus of a Western Reserve strain comprising a nucleic acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 99%, or 100% identity to the nucleic acid sequence of SEQ ID NO: 108.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • the nucleic acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 99%, or 100% identity to the nucleic acid sequence of SEQ ID NO: 108 is integrated into the TK gene of the vaccinia virus.
  • composition comprising (a) a recombinant oncolytic virus comprising a nucleotide sequence encoding a foreign antigen; and (b) an engineered immune cell expressing a chimeric receptor specifically recognizing said foreign antigen.
  • the foreign antigen is abacterial antigen.
  • the foreign antigen is a sialidase.
  • the present application further provides immune cells comprising any one of the recombinant oncolytic viruses provided herein.
  • the immune cells comprising a recombinant oncolytic virus are prepared by incubating the immune cells with the recombinant oncolytic virus.
  • the immune cells comprising a recombinant oncolytic virus are prepared by engineering a nucleotide sequence encoding the recombinant oncolytic virus into the cells (e.g., by transducing or transfecting the cells with the construct). Suitable immune cells expressing recombinant oncolytic virus and methods of preparation thereof are described in the ''Oncolytic virus and engineered immune cells” subsection below.
  • the present application provides recombinant oncolytic viruses for use in treating a cancer, comprising at least one nucleotide sequence encoding a heterologous protein.
  • the heterologous protein is operably linked to a promoter.
  • the heterologous protein is a sialidase.
  • oncolytic viruses including Vaccinia virus, Coxsackie virus, Adenovirus, Measles, Newcastle disease virus, Seneca Valley virus, Coxsackie A21, Vesicular stomatitis virus, Parvovirus Hl, Reovirus, Herpes virus, Lentivirus, and Poliovirus, and Parvovirus.
  • Vaccinia Virus Western Reserve, GLV-lh68, ACAM2000, and OncoVEX GFP are available.
  • the genomes of these oncolytic virus can be genetically modified to insert a nucleotide sequence encoding a protein that includes all or a catalytic portion of a sialidase.
  • the nucleotide sequence encoding a protein that includes all or a catalytically active portion of a sialidase is placed under the control of a viral expression cassette so that the sialidase is expressed by infected cells.
  • Oncolytic viruses have the ability to preferentially accumulate in and replicate in and kill tumor cells, relative to normal cells. This ability can be a native feature of the virus (e.g., pox virus, reovirus, Newcastle disease virus and mumps virus), or the viruses can be modified or selected for this property.
  • Viruses can be genetically attenuated or modified so that they can circumvent antiviral immune and other defenses in the subject (e.g., vesicular stomatitis virus, herpes simplex virus, adenovirus) so that they preferentially accumulate in tumor cells or the tumor microenvironment, and/or the preference for tumor cells can be selected for or engineered into the virus using, for example, tumor-specific cell surface molecules, transcription factors and tissue-specific microRNAs (see, e.g, Cattaneo el al, Nat. Rev. Microbiol., 6(7):529-540 (2008); Dorer et al, Adv. Drug Deliv. Rev., 61(7- 8):554-571 (2009); Kelly et al. , Mol. Then, 17(3):409-416 (2009); and Naik et al. , Expert Opin. Biol. Then, 9(9): 1163-1176 (2009)).
  • antiviral immune and other defenses in the subject
  • oncolytic viruses can be achieved via direct intratumoral injection. While direct intratumoral delivery can minimize the exposure of normal cells to the virus, there often are limitations due to, e.g., inaccessibility of the tumor site (e.g., brain tumors) or for tumors that are in the form of several small nodules spread out over a large area or for metastatic disease. Viruses can be delivered via systemic or local delivery, such as by intravenous administration, or intraperitoneal administration, and other such routes. Systemic delivery can deliver virus not only to the primary tumor site, but also to disseminated metastases.
  • direct intratumoral delivery can minimize the exposure of normal cells to the virus, there often are limitations due to, e.g., inaccessibility of the tumor site (e.g., brain tumors) or for tumors that are in the form of several small nodules spread out over a large area or for metastatic disease.
  • Viruses can be delivered via systemic or local delivery, such as by intravenous administration, or intraperitoneal
  • oncolytic viruses including Vaccinia virus, Coxsackie virus, Adenovirus, Measles, Newcastle disease virus, Seneca Valley virus, Coxsackie A21, Vesicular stomatitis virus, Parvovirus Hl, Reovirus, Herpes virus, Lentivirus, and Poliovirus, and Parvovirus.
  • Vaccinia Virus Western Reserve, GLV-lh68, ACAM2000, and OncoVEX GFP are available.
  • the genomes of these oncolytic virus can be genetically modified to insert a nucleotide sequence encoding a protein that includes all or a catalytic portion of a sialidase.
  • the nucleotide sequence encoding a protein that includes all or a catalytically active portion of a sialidase is placed under the control of a viral expression cassette so that the sialidase is expressed by infected cells.
  • unmodified oncolytic viruses include any known to those of skill in the art, including those selected from among viruses designated GLV-lh68, JX-594, JX-954, ColoAdl, MV-CEA, MV-NIS, ONYX-015, B18R, H101, OncoVEX GM-CSF, Reolysin, NTX-010, CCTG-102, Cavatak, Oncorine, and TNFerade.
  • Oncolytic viruses have been described, for example, in W02020097269, which is incorporated herein by reference in its entirety.
  • Oncolytic viruses described herein include for example, vesicular stomatitis virus, see, e.g., U.S. Patent Nos. 7,731,974, 7,153,510, 6,653,103 and U.S. Pat. Pub. Nos. 2010/0178684, 2010/0172877, 2010/0113567, 2007/0098743, 20050260601, 20050220818 and EP Pat. Nos. 1385466, 1606411 and 1520175; herpes simplex virus, see, e.g., U.S. Patent Nos.
  • the oncolytic virus is a vesicular stomatitis virus (VSV).
  • VSV has been used in multiple oncolytic virus applications.
  • VSV has been engineered to express an antigenic protein of human papilloma virus (HPV) as a method to treat HPV positive cervical cancers via vaccination (REF 18337377, 29998190) and to express pro- inflammatory factors to increase the immune reaction to tumors (REF 12885903).
  • HPV human papilloma virus
  • Various methods for engineering VSV to encode an additional gene have been described (REF 7753828).
  • VSV RNA genome is reverse transcribed to a complementary, doubled stranded-DNA with an upstream T7 RNA polymerase promoter and an appropriate location within the VSV genome for gene insertion is identified (e.g., within the noncoding 5’ or 3’ regions flanking VSV glycoprotein (G) (REF 12885903). Restriction enzyme digestion can be accomplished, e.g., with Mlu I and Nhe I, yielding a linearized DNA molecule. An insert consisting of a DNA molecule encoding the gene of interest flanked by appropriate restriction sites can be ligated into the linearized VSV genomic DNA.
  • G VSV glycoprotein
  • the resulting DNA can be transcribed with T7 polymerase, yielding a complete VSV genomic RNA containing the inserted gene of interest.
  • Introduction of this RNA molecule to a mammalian cell, e.g., via transfection and incubation results in viral progeny expressing the protein encoded by the gene of interest.
  • the recombinant oncolytic virus is an adenovirus.
  • the adenovirus is an adenovirus serotype 5 virus (Ad5).
  • Ad5 contains a human E2F-1 promoter, which is a retinoblastoma (Rb) pathway-defective tumor specific transcription regulatory element that drives expression of the essential Ela viral genes, restricting viral replication and cytotoxicity to Rb pathway-defective tumor cells (REF 16397056).
  • Rb pathway-defective tumor cells Rb pathway defects.
  • Engineering a gene of interest into Ad5 is accomplished through ligation into Ad5 genome.
  • a plasmid containing the gene of interest is generated via and digested, e.g.
  • Ad5 DNA plasmid e.g. , PSF-AD5 (REF Sigma OGS268) is digested with AsiSI and Pacl and ligated with recombinant bacterial ligase or co-transformed with RE digested gene of interest into permissive E.coli as has been reported for the generation of human granulocyte macrophage colony stimulating factor (GM-CSF) expressing Ad5 (REF 16397056).
  • GM-CSF granulocyte macrophage colony stimulating factor
  • the recombinant oncolytic virus is a modified oncolytic virus (e.g., a derivative of any one of the viruses described herein).
  • the recombinant oncolytic virus comprises one or more mutations that reduce immunogenicity of the virus compared to a corresponding wild-type strain.
  • the recombinant oncolytic virus is a vaccinia virus (VV).
  • VV vaccinia virus
  • TK viral thymidine kinase
  • WR Western Reserve
  • Production of VV’s with a gene of interest inserted in the genome may be accomplished with homologous recombination utilizing lox sites.
  • the virus is a modified vaccinia virus. In some embodiments, the virus is a modified vaccinia virus comprising one or more mutations. In some embodiments, the one or more mutations are in one or more proteins such as A14, A17, A13, LI, H3, D8, A33, B5, A56, F13, A28, and A27. In some embodiments, the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R. Exemplary mutations have been described, for example, in international patent publication W02020086423, which is incorporated herein by reference in its entirety.
  • VVs as cancer treatment delivery vectors
  • Nab neutralizing antibody
  • the NAbs recognize and bind viral glycoproteins embedded in the VV envelope, thus preventing virus interaction with host cell receptors.
  • a number of VV glycoproteins involved in host cell receptor recognition have been identified. Among them, proteins H3L, L1R, A27L, D8L, A33R, and B5R have been shown to be targeted by NAbs, with A27L, H3L, D8L and L1R being the main NAb antigens presented on the surface of mature viral particles.
  • A27L, H3L, and D8L are the adhesion molecules that bind to host glycosaminoglycans (GAGs) heparan sulfate (HS) (A27L and H3L) and chondroitin sulfate (CS) (D8L) and mediate endocytosis of the virus into the host cell.
  • GAGs host glycosaminoglycans
  • HS heparan sulfate
  • CS chondroitin sulfate
  • D8L mediate endocytosis of the virus into the host cell.
  • L1R protein is involved in virus maturation. Modified vaccinia viruses comprising mutations in one or more of these proteins have been described in international patent publication W02020086423, which is herein incorporated by reference in its entirety.
  • the modified vaccinia virus comprises one or more proteins selected from the group consisting of: (a) a variant vaccinia virus (VV) H3L protein that comprises an amino acid sequence having at least 90% (e.g., at least 91%, 92 %, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) amino acid sequence identity to any one of SEQ ID NOS: 66-69; (b) a variant vaccinia virus (VV) D8L protein that comprises an amino acid sequence having at least 90% (e.g., at least 91%, 92 %, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) amino acid sequence identity to any one of SEQ ID NOS: 70-72 or 85; (c) a variant vaccinia virus (VV) A27L protein that comprises an amino acid sequence having at least 90% (e.g., at least 91%, 92 %, 93%, 9
  • the variant VV H3L protein comprises amino acid substitution or deletion at one or more of the following amino acid residues: 14, 15, 16, 33, 34, 35, 38, 40, 44, 45, 52, 131, 134, 135, 136, 137, 154, 155, 156, 161, 166, 167, 168, 198, 227, 250, 253, 254, 255, and 256, wherein the amino acid numbering is based on SEQ ID NO: 66.
  • the variant VV H3L comprises one or more amino acid mutations selected from the group consisting of I14A, D15A, R16A, K38A, P44A, E45A, V52A, E131A, T134A, L136A, R137A, R154A, E155A, I156A, M168A, I198A, E250A, K253A, P254A, N255A, and F256A, wherein the amino acid numbering is based on SEQ ID NO: 66.
  • the variant VV D8L protein comprises amino acid substitution or deletion at one or more of the following amino acid residues: 44, 48, 98, 108, 117, and 220, wherein the amino acid numbering is based on SEQ ID NO: 70.
  • the variant VV D8L construct comprises one or more amino acid mutations selected from the group consisting of R44A, K48A, K98A, K108A, K117A, and R220A, wherein the amino acid numbering is based on SEQ ID NO: 70.
  • the variant VV A27L protein comprises amino acid substitution or deletion at one or more of the following amino acid residues: 27, 30, 32, 33, 34, 35, 36, 37, 39, 40, 107, 108, and 109, wherein the amino acid numbering is based on SEQ ID NO: 73.
  • the variant A27L construct comprises one or more amino acid mutations selected from the group consisting of K27A, A30D, R32A, E33A, A34D, 135A, V36A, K37A, D39A, E40A, R107A, P108A, and Y109A, wherein the amino acid numbering is based on SEQ ID NO: 73.
  • the variant VV L1R protein comprises amino acid substitution or deletion at one or more of the following amino acid residues: 25, 27, 31, 32, 33, 35, 58, 60, 62, 125, and 127, wherein the amino acid numbering is based on SEQ ID NO: 74.
  • the variant L1R construct comprises one or more amino acid mutations selected from the group consisting of E25A, N27A, Q31A, T32A, K33A, D35A, S58A, D60A, D62A, K125A, and K127A, wherein the amino acid numbering is based on SEQ ID NO: 74.
  • the variant VV H3L protein comprises amino acid substitution or deletion at one or more of the following amino acid residues: 14, 15, 16, 33, 34, 35, 38, 40, 44, 45, 52, 131, 132, 134, 135, 136, 137, 154, 155, 156, 161, 166, 167, 168, 195, 198, 199, 227, 250, 251, 252, 253, 254, 255, 256, 258, 262, 264, 266, 268, 272, 273, 275, and 277, wherein the amino acid numbering is based on SEQ ID NO: 68.
  • the variant H3L construct comprises one or more amino acid mutations selected from the group consisting of I14A, D15A, R16A, K33A, F34A, D35A, K38A, N40A, P44A, E45A, V52A, E131A, D132A, T134A, F135A, L136A, R137A, R154A, E155A, I156A, K161A, L166A, VI 67 A, M168A, E195A, I198A, V199A, R227A, E250A, N251A, M252A, K253A, P254A, N255A, F256A, S258A, T262P, A264T, K266I, Y268C, M272K, Y273N, F275N, and T277A, wherein the amino acid numbering is based on SEQ ID NO: 68.
  • the variant VV D8L protein comprises amino acid substitution or deletion at one or more of the following amino acid residues: 43, 44, 48, 53, 54, 55, 98, 108, 109, 144, 168, 177, 196, 199, 203, 207, 212, 218, 220, 222, and 227, wherein the amino acid numbering is based on SEQ ID NO: 72.
  • the variant VV D8L construct comprises one or more amino acid mutations selected from the group consisting of V43A, R44A, K48A, S53A, G54A, G55A, K98A, K108A, K109A, A144G, T168A, S177A, L196A, F199A, L203A, N207A, P212A, N218A, R220A, P222A, and D227A, wherein the amino acid numbering is based on SEQ ID NO: 72.
  • the recombinant oncolytic virus is a vaccinia virus derived from vvDD.
  • the vvDD virus has been described (see McCart JA, et al. Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes. Cancer Res 2001;61:8751-8757, the content of which is herein incorporated by reference in its entirety).
  • the vvDD virus is a Western Reserve strain virus and comprises a disruption of the vaccinia growth factor (VGF) gene and of the viral thymidine kinase (IK) gene.
  • the recombinant oncolytic virus is a vvDD derivative comprising a nucleotide sequence encoding a sialidase and a second nucleotide sequence encoding a multispecific immune cell engager integrated into the TK gene of the vaccinia virus.
  • the recombinant oncolytic virus encodes a heterologous protein that includes all or a catalytic portion of a sialidase that is capable of removing sialic acid (N- acetylneuraminic acid (Neu5Ac)), e.g. , from a glycan on a human cell.
  • a sialidase that is capable of removing sialic acid (N- acetylneuraminic acid (Neu5Ac)
  • Neu5Ac is linked via an alpha 2,3, an alpha 2,6 or alpha 2,8 linkage to the penultimate sugar in glycan on a protein by any of a variety of sialyl transferases.
  • the heterologous protein in addition to a naturally occuring sialidase or catalytic portion thereof can, optionally, include peptide or protein sequences that contribute to the therapeutic activity of the protein.
  • the protein can include an anchoring domain that promotes interaction between the protein and a cell surface.
  • the anchoring domain and sialidase domain can be arranged in any appropriate way that allows the protein to bind at or near a target cell membrane such that the therapeutic sialidase can exhibit an extracellular activity that removes sialic acid residues.
  • the protein can have more than one anchoring domains. In cases in which the polypeptide has more than one anchoring domain, the anchoring domains can be the same or different.
  • the protein can comprise one or more transmembrane domains (e.g.
  • the protein can have more than one sialidase domain. In cases in which a compound has more than one sialidase domain, the sialidase domains can be the same or different. Where the protein comprises multiple anchoring domains, the anchoring domains can be arranged in tandem (with or without linkers) or on alternate sides of other domains, such as sialidase domains. Where a compound comprises multiple sialidase domains, the sialidase domains can be arranged in tandem (with or without linkers) or on alternate sides of other domains. In some embodiments, the sialidase comprises an immunoglobulin Fc domain (e.g., an IgG Fc domain). In some embodiments, the Fc domain is positioned between the sialidase catalytic domain and a transmembrane domain.
  • an immunoglobulin Fc domain e.g., an IgG Fc domain
  • the recombinant oncolytic virus comprises a first nucleotide sequence encoding a sialidase, wherein the sialidase comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 105.
  • the sialidase comprises the amino acid sequence of SEQ ID NO: 105.
  • the first nucleotide sequence encoding the sialidase further encodes a signal peptide sequence operably linked to the sialidase.
  • the signal peptide sequence comprises the amino acid sequence of SEQ ID NO: 102.
  • the first nucleotide sequence encodes an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 106.
  • the first nucleotide sequence encodes the amino acid sequence of SEQ ID NO: 106.
  • the first nucleotide sequence comprises a sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the nucleic acid sequence of SEQ ID NO: 110. In some embodiments, the first nucleotide sequence comprises the nucleic acid sequence of SEQ ID NO: 110.
  • the sialidase has exo-sialidase activity as defined by Enzyme Commission EC 3.2. 1.18. In some embodiments, the sialidase is an anhydrosialidase as defined by Enzyme Commission EC 4.2.2.15.
  • the sialidase expressed by the oncolytic virus can be specific for Neu5Ac linked via alpha 2,3 linkage, specific for Neu5Ac linked via an alpha 2,6 specific for Neu5Ac linked via alpha 2,8 linkage, or can cleave Neu5Ac linked via an alpha 2,3 linkage or an alpha 2,6 linkage.
  • the sialidase can cleave Neu5Ac linked via an alpha 2,3 linkage, an alpha 2,6 linkage, or an alpha 2,8 linkage.
  • a variety of sialidases are described in Tables 2-5.
  • a sialidase that can cleave more than one type of linkage between a sialic acid residue and the remainder of a substrate molecule in particular, a sialidase that can cleave both alpha(2,6)-Gal and alpha(2,3)-Gal linkages or both alpha(2,6)-Gal and alpha(2,3)-Gal linkages and alpha(2,8)-Gal linkages can be used in the compounds of the disclosure.
  • Sialidases included are the large bacterial sialidases that can degrade the receptor sialic acids Neu5Ac alpha(2,6)-Gal and Neu5Ac alpha(2,3)-Gal.
  • the bacterial sialidase enzymes from Clostridium perfringens (Genbank Accession Number X87369), Actinomyces viscosus (GenBankX62276), Arthrobacter ureafaciens GenBank (AY934539), or Micromonospora viridifaciens (Genbank Accession Number DO 1045) can be used.
  • the sialidase comprises all or a portion of the amino acid sequence of a large bacterial sialidase or can comprise amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to all or a portion of the amino acid sequence of a large bacterial sialidase.
  • the sialidase domain comprises SEQ ID NO: 2 or 27, or a sialidase sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 12.
  • a sialidase domain comprises the catalytic domain of the Actinomyces viscosus sialidase extending from amino acids 274-666 of SEQ ID NO: 26, having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to amino acids 274-666 of SEQ ID NO: 26.
  • Additional sialidases include the human sialidases such as those encoded by the genes NEU2 (SEQ ID NO: 4; Genbank Accession Number Y16535; Monti, E, Preti, Rossi, E., Ballabio, A and Borsani G. (1999) Genomics 57: 137-143) andNEU4 (SEQ ID NO: 6; Genbank Accession Number NM080741; Monti et al. (2002) Neurochem Res 27:646-663).
  • Sialidase domains of compounds of the present disclosure can comprise all or a portion of the amino acid sequences of a sialidase or can comprise amino acid sequences having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to all or a portion of the amino acid sequences of a sialidase.
  • a sialidase domain comprises a portion of the amino acid sequences of a naturally occurring sialidase, or sequences having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to a portion of the amino acid sequences of a naturally occurring sialidase
  • the portion comprises essentially the same activity as the intact sialidase.
  • the sialidase expressed by the recombinant oncolytic virus is a sialidase catalytic domain protein.
  • a "sialidase catalytic domain protein” comprises a catalytic domain of a sialidase but does not comprise the entire amino acid sequence of the sialidase from which the catalytic domain is derived.
  • a “sialidase catalytic domain protein” has sialidase activity, and the term as used herein is interchangeable with a “sialidase”.
  • a sialidase catalytic domain protein comprises at least 10%, at least 20%, at least 50%, at least 70% of the activity of the sialidase from which the catalytic domain sequence is derived.
  • a sialidase catalytic domain protein comprises at least 90% of the activity of the sialidase from which the catalytic domain sequence is derived.
  • a sialidase catalytic domain protein can include other amino acid sequences, such as but not limited to additional sialidase sequences, sequences derived from other proteins, or sequences that are not derived from sequences of naturally occurring proteins. Additional amino acid sequences can perform any of a number of functions, including contributing other activities to the catalytic domain protein, enhancing the expression, processing, folding, or stability of the sialidase catalytic domain protein, or even providing a desirable size or spacing of the protein.
  • the sialidase catalytic domain protein is a protein that comprises the catalytic domain of the A. viscosus sialidase.
  • an A. viscosus sialidase catalytic domain protein comprises amino acids 270-666 of the A. viscosus sialidase sequence (SEQ ID NO: 26; GenBank WP_003789074).
  • an A. Viscosus sialidase catalytic domain protein comprises an amino acid sequence that begins at any of the amino acids from amino acid 270 to amino acid 290 of the A. viscosus sialidase sequence (SEQ ID NO: 26) and ends at any of the amino acids from amino acid 665 to amino acid 901 of said A. viscosus sialidase sequence (SEQ ID NO: 26), and lacks any A. viscosus sialidase protein sequence extending from amino acid 1 to amino acid 269.
  • an A. viscosus sialidase catalytic domain protein comprises amino acids 274-681 of the A. viscosus sialidase sequence (SEQ ID NO: 26) and lacks other viscosus sialidase sequence.
  • an A. viscosus sialidase catalytic domain protein comprises amino acids 274-666 of the A. viscosus sialidase sequence (SEQ ID NO: 26) and lacks any other A. viscosus sialidase sequence.
  • an A. viscosus sialidase catalytic domain protein comprises amino acids 290-666 of the A. viscosus sialidase sequence (SEQ ID NO: 26) and lacks any other A.
  • an A. viscosus sialidase catalytic domain protein comprises amino acids 290- 681 of the A. viscosus sialidase sequence (SEQ ID NO: 26) and lacks any other A. viscosus sialidase sequence.
  • useful sialidase polypeptides for expression by an oncolytic virus include polypeptides comprising a sequence that is 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 27 or comprises 375, 376, 377, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, or 392 contiguous amino acids of SEQ ID NO: 27.
  • the sialidase is DAS 181, a functional derivative thereof (e.g., a fragment thereof), or a biosimilar thereof.
  • the sialidase comprises an amino acid sequence that is at least about 80% (e.g., at least about any one of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) or 100% identical to SEQ ID NO: 2.
  • the sialidase comprises 414, 413, 412, 411, or 410 contiguous amino acids of SEQ ID NO: 2.
  • the sialidase comprises a fragment of DAS 181 without the anchoring domain (AR domain).
  • the sialidase comprises an amino acid sequence that is at least about 80% (e.g., at least about any one of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) or 100% identical to SEQ ID NO: 27.
  • DAS 181 is a recombinant sialidase fusion protein with a heparin-binding anchoring domain. DAS 181 and methods for preparing and formulating DAS 181 are described in US 7,645,448; US 9,700,602 and US 10,351,828, each of which is herein incorporated by reference in their entirety for any and all purposes.
  • the sialidase is a secreted form of DAS 181, a functional derivative thereof, or a biosimilar thereof.
  • the nucleotide sequence encoding a secreted form of DAS 181 encodes a secretion sequence operably linked to DAS 181, wherein the secretion sequence is enables secretion of the protein from eukaryotic cells.
  • the sialidase comprises an amino acid sequence that is at least about 80% (e.g, at least about any one of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) or 100% identical to SEQ ID NO: 28.
  • the sialidase comprises an amino acid sequence that is at least about 80% (e.g., at least about any one of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) or 100% identical to SEQ ID NO: 28.
  • the sialidase comprises 414, 413, 412, 411, or 410 contiguous amino acids of SEQ ID NO: 28.
  • An exemplary secreted form of DAS 181 is described in Example 11.
  • the sialidase is a transmembrane form of DAS 181 , a functional derivative thereof, or a biosimilar thereof.
  • the sialidase comprises an amino acid sequence that is at least about 80% (e.g., at least about any one of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) or 100% identical to SEQ ID NO: 31.
  • the sialidase comprises an amino acid sequence that is at least about 80% (e.g., at least about any one of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) or 100% identical to SEQ ID NO: 31.
  • the sialidase comprises 414, 413, 412, 411, or 410 contiguous amino acids of SEQ ID NO: 31.
  • An exemplary transmembrane form of DAS 181 is described in Example 11.
  • amino acids that differ from those from the reference sequences described above are conservative substitutions or highly conservative substitutions.
  • Conservative substitutions and highly conservative substitutions can be as defined in the “Sialidase” section above.
  • Conservative substitutions may be defined as exchanges within one of the following five groups:
  • substitutions are considered to be “highly conservative”: Asp/Glu, His/Arg/Lys, Phe/Tyr/Trp, and Met/Leu/Ile/Val.
  • Semi-conservative substitutions are defined to be exchanges between two of groups (I)-(IV) above which are limited to supergroup (A), comprising (I), (II), and (III) above, or to supergroup (B), comprising (IV) and (V) above.
  • hydrophobic amino acids refer to the amino acids Ala, Gly, Pro, Met, Leu, He, Vai, Cys, Phe, and Trp
  • hydrophilic amino acids refer to Ser, Thr, Asp, Asn, Glu, Gin, His, Arg, Lys, and Tyr.
  • the sialidase comprises an anchoring domain.
  • an "extracellular anchoring domain” or “anchoring domain” is any moiety that interacts with an entity that is at or on the exterior surface of a target cell or is in close proximity to the exterior surface of a target cell.
  • An anchoring domain can serve to retain a sialidase of the present disclosure at or near the external surface of a target cell.
  • An extracellular anchoring domain may bind 1) a molecule expressed on the surface of a cancer cell, or a moiety, domain, or epitope of a molecule expressed on the surface of a cancer cell, 2) a chemical entity attached to a molecule expressed on the surface of a cancer cell, or 3) a molecule of the extracellular matrix surrounding a cancer cell.
  • An exemplary anchoring domain binds to heparin/sulfate, a type of GAG that is ubiquitously present on cell membranes. Many proteins specifically bind to heparin/heparan sulfate, and the GAG-binding sequences in these proteins have been identified (Meyer, F A, King, M and Gelman, R A.
  • the anchoring domain is a non-protein anchoring moiety, such as a phosphatidylinositol (GPI) linker.
  • GPI phosphatidylinositol
  • the sialidase comprises a transmembrane domain.
  • the sialidase domain can be joined to a mammalian (preferably human) transmembrane (TM) domain. This arrangement permits the sialidase to be expressed on the cell surface.
  • Suitable transmembrane domain include, but are not limited to a sequence comprising human CD28 TM domain (NM_006139; FWVLVVVGGVLACYSLLVTVAFIIFWV (SEQ ID NO: 46), human CD4 TM domain (M35160; MALIVLGGVAGLLLFIGLGIFF (SEQ ID NO: 47); human CD8 TM1 domain (NM 001768; IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO: 48); human CD8 TM2 domain (NM 001768; IYIWAPLAGTCGVLLLSLVITLY (SEQ ID NO: 49); human CD8 TM3 domain (NM 001768; IYIWAPLAGTCGVLLLSLVITLYC (SEQ ID NO: 50); human 41BB TM domain (NM 001561; IISFFLALTSTALLFLLFF LTLRFSVV (SEQ ID NO: 51); human PDGFR TM1 domain (VVISAILA LVVLTIISLIILI; SEQ ID
  • the nucleotide sequence encoding a sialidase encodes a protein comprising, from amino terminus to carboxy terminus, a secretion sequence (e.g., SEQ ID NO: 40), a sialidase (e.g., a sialidase comprising an amino acid sequence selected from SEQ ID NOs: 1-27, and a transmembrane domain (e.g., a transmembrane domain selected from SEQ ID NOs: 45-52).
  • a secretion sequence e.g., SEQ ID NO: 40
  • a sialidase e.g., a sialidase comprising an amino acid sequence selected from SEQ ID NOs: 1-27
  • a transmembrane domain e.g., a transmembrane domain selected from SEQ ID NOs: 45-52
  • any suitable secretion sequence, sialidase domain sequence, or transmembrane domain may be used.
  • the nucleotide sequence encoding a sialidase encodes a protein comprising, from amino terminus to carboxy terminus, a secretion sequence (e.g., SEQ ID NO: 40), the sialidase of SEQ ID NO: 27, and a transmembrane domain (e.g, a transmembrane domain selected from SEQ ID NOs: 45-52).
  • the sialidase has at least 50%, at least 60%, at least 65%, 80% (e.g. , at least about any one of 85%, 86%, 87%, 88%, 89%) or at least 90% (e.g. , at least about any one of 91%, 92%, 94%, 96%, 98%, or 99%) sequence identity to a sequence selected from SEQ ID NOs: 31.
  • the sialidase comprises a sequence selected from SEQ ID NOs: 31.
  • the sialidase comprises the amino acid sequence of SEQ ID NO: 31.
  • the sialidase comprises an immunoglobulin G (IgG) Fc (fragment, crystallizable) domain.
  • the sialidase comprises from the N- terminus to the C-terminus: a sialidase catalytic domain, an IgG Fc domain, and a transmembrane domain.
  • the IgG Fc domain comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, or 99% identity to the amino acid sequence of SEQ ID NO: 104.
  • the IgG Fc domain comprises the amino acid sequence of SEQ ID NO: 104.
  • a protein that includes a sialidase or a catalytic domain thereof can optionally include one or more polypeptide linkers that can join various domains of the sialidase.
  • Linkers can be used to provide optimal spacing or folding of the domains of a protein.
  • the domains of a protein joined by linkers can be sialidase domains, anchoring domains, transmembrane domains, or any other domains or moieties of the compound that provide additional functions such as enhancing protein stability, facilitating purification, etc.
  • Some preferred linkers include the amino acid glycine. For example, linkers having the sequence: (GGGGS (SEQ ID NO: 55))n, where n is 1-20.
  • the linker is a hinge region of an immunoglobulin. Any hinge or linker sequence capable of keeping the catalytic domain free of steric hindrance can be used to link a domain of a sialidase to another domain (e.g., a transmembrane domain or an anchoring domain).
  • the linker is a hinge domain comprising the sequence of SEQ ID NO: 62.
  • the nucleotide sequence encoding the sialidase further encodes a secretion sequence (e.g. , a signal sequence or signal peptide) operably linked to the sialidase.
  • a secretion sequence e.g. , a signal sequence or signal peptide
  • secretion sequence is a signal peptide operably linked to the N-terminus of the protein.
  • the length of the secretion sequence ranges between 10 and 30 amino acids (e.g. , between 15 and 25 amino acids, between 15 and 22 amino acids, or between 20 and 25 amino acids).
  • the secretion sequence enables secretion of the protein from eukaryotic cells.
  • the secretion sequence is usually cleaved off and the protein enters the secretory pathway.
  • the nucleotide sequence encodes, from N- terminus to C-terminus, a secretion sequence, a sialidase, and a transmembrane domain, wherein the sialidase is operably linked to the secretion sequence and the transmembrane domain.
  • the N-terminal secretion sequence is cleaved resulting in a protein with an N-terminal extracellular domain.
  • Exemplary secretion sequences are provided in SEQ ID NOS: 40, 102, and 103.
  • the recombinant oncolytic virus encodes a multispecific immune cell engager.
  • the multi-specific immune cell engager is a bispecific immune cell engager.
  • the heterologous protein is a bispecific T cell engager (BiTE). Exemplary bispecific immune cell engagers have been described, for example, in international patent publication WO2018049261, herein incorporated by reference in its entirety.
  • the bispecific immune cell engager comprises a first antigen-binding domain (such as scFv) specifically recognizing a tumor antigen (such as EpCAM, FAP, or EGFR, etc) and a second antigen-binding domain (such as scFv) specifically recognizing a cell surface molecule on an effector cell (such as CD3 or 4- IBB on T lymphocytes).
  • Tumor antigens can be a tumor-associated antigen (TAA) or a tumor-specific antigen (TSA).
  • TAA or TSA is expressed on a cell of a solid tumor.
  • Tumor antigens include, but are not limited to, EpCAM, FAP, EphA2, HER2, GD2, EGFR, VEGFR2, and Glypican-3 (GPC3), CDH17, Fibulin-3, HHLA2, Folate receptors, etc.
  • the tumor antigen is EpCAM.
  • the tumor antigen is FAP.
  • the tumor antigen is EGFR.
  • the tumor antigen is FAP.
  • the cell surface molecule on the effector cell is CD3 or 41-BB.
  • the cell surface marker on the effector cell is CD3s.
  • the first antigen-binding domain is an scFv
  • the second antigen binding domain is an scFv.
  • the multispecific immune cell engager comprises a first scFv that recognizes FAP, and a second scFv that recognizes CD3/ CD3a.
  • the tumor antigen is FAP and the first antigen-binding domain comprises: (i) a first light chain complementarity-determining region (CDR-L1) having the amino acid sequence of SEQ ID NO: 86, (i) a second light chain complementaritydetermining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 87, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 88, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 89, (v) a second heavy chain complementaritydetermining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 90, and (vi) a third heavy chain complementarity-determining region (CDR-H3) having the amino acid sequence of SEQ ID NO: 91.
  • CDR-L1 first light chain complementarity-determining region having the amino acid sequence of SEQ ID NO: 86
  • the tumor antigen is FAP and the first antigen-binding domain comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 98.
  • the first antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 98.
  • the cell surface molecule on the effector cell is CD3, and the second antigen-binding domain comprises: (i) a first light chain complementarity -determining region (CDR-L1) having the amino acid sequence of SEQ ID NO: 92, (ii) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 93, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 94, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 95, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 96, and (vi) a third heavy chain complementarity-determining region (CDR-H3) having the amino acid sequence of SEQ ID NO: 97.
  • CDR-L1 having the amino acid sequence of SEQ ID NO: 92
  • the cell surface molecule on the effector cell is CD3 and the second antigen-binding domain comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 99.
  • the cell surface molecule on the effector cell is CD3 and the second antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 99.
  • recombinant oncolytic virus comprises a second nucleotide sequence encoding a multispecific immune cell engager comprising an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 100.
  • the multispecific immune cell engager comprises the amino acid sequence of SEQ ID NO: 100.
  • amino acids that differ from those from the reference sequences described above are conservative substitutions or highly conservative substitutions.
  • Conservative substitutions and highly conservative substitutions can be as defined in the “Sialidase” section above.
  • the second nucleotide sequence further encodes a signal peptide sequence operably linked to the multispecific immune cell engager.
  • the signal peptide sequence comprises the amino acid sequence of SEQ ID NO: 103.
  • the second nucleotide sequence encodes an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to SEQ ID NO: 101.
  • the second nucleotide sequence encodes the amino acid sequence of SEQ ID NO: 101.
  • the second nucleotide sequence comprises a nucleic acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the nucleic acid sequence of SEQ ID NO: 109. In some embodiments, the second nucleotide sequence comprises the nucleic acid sequence of SEQ ID NO: 109.
  • the oncolytic virus further comprises an additional nucleotide sequence encoding a heterologous protein or nucleic acid.
  • the additional nucleotide sequence encodes a heterologous protein.
  • the heterologous protein is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is an inhibitor of CTLA-4, PD-1, PD-L1, TIGIT, LAG3, TIM-3, VISTA, B7-H4, or HLA-G.
  • the immune checkpoint inhibitor is an antibody.
  • the immune checkpoint modulator is an immune checkpoint inhibitor, such as an inhibitor or an antagonist antibody or a decoy ligand of PD-1, PD-L1, PD-L2, CD47, CXCR4, CSF1R, LAG-3, TIM-3, HHLA2, BTLA, CD160, CD73, CTLA-4, B7-H4, TIGIT, VISTA, or 2B4
  • the immune checkpoint modulator is an inhibitor of PD-1.
  • the immune checkpoint inhibitor is an antibody against an immune checkpoint molecule, such as an anti-PD-1 antibody.
  • the immune checkpoint inhibitor is a ligand that binds to the immune checkpoint molecule, such as soluble or free PD-L1/PD-L2.
  • the immune checkpoint inhibitor is an extracellular domain of PD-1 fused to an Fc fragment of an immunoglobulin (such as IgG4 Fc)) that can block PDL-1 on tumor cell surface binding to the immune check point PD-1 on immune cells.
  • the immune checkpoint inhibitor is a ligand that binds to HHLA2.
  • the immune checkpoint inhibitor is an extracellular domain of TMIGD2 fused to an Fc fragment of an immunoglobulin, such as IgG4 Fc.
  • the immune checkpoint inhibitor is a ligand that binds to at least two different inhibitory immune checkpoint molecules (e.g. bispecific), such as a ligand that binds to both CD47 and CXCR4.
  • the immune checkpoint inhibitor comprises an extracellular domain of SIRPa and a CXCL12 fragment fused to an Fc fragment of an immunoglobulin, such as IgG4 Fc. These molecules can bind to CD47 on cancer cell, thus stopping its interaction with SIRPalpha to block the “don’t eat me” signal to macrophages and dendritic cells.
  • the heterologous protein is an inhibitor of an immune suppressive receptor.
  • the immune suppressive receptor can be any receptor expressed by an immune effector cell that inhibits or reduces an immune response to tumor cells. Exemplary effector cell includes without limitation a T lymphocyte, a B lymphocyte, a natural killer (NK) cell, a dendritic cell (DC), a macrophage, a monocyte, a neutrophil, an NKT-cell, or the like.
  • the immune suppressive receptor is LILRB, TYRO3, AXL, Folate receptor beta or MERTK.
  • the inhibitor of an immune suppressive receptor is an anti-LILRB antibody.
  • the heterologous protein is a multi-specific immune cell engager.
  • effector cells include, but are not limited to T lymphocyte, B lymphocyte, natural killer (NK) cell, dendritic cell (DC), macrophage, monocyte, neutrophil, NKT-cell, or the like.
  • the effector cell is a T lymphocyte.
  • the effector cell is a cytotoxic T lymphocyte.
  • Cell surface molecules on an effector cell include, but are not limited to CD3, CD4, CD5, CD8, CD16, CD28, CD40, CD64, CD89, CD134, CD137, NKp46, NKG2D, or the like.
  • the cell surface molecule is CD3.
  • a cell surface molecule on an effector cell of the present application is a molecule found on the external cell wall or plasma membrane of a specific cell type or a limited number of cell types.
  • Examples of cell surface molecules include, but are not limited to, membrane proteins such as receptors, transporters, ion channels, proton pumps, and G protein-coupled receptors; extracellular matrix molecules such as adhesion molecules (e.g., integrins, cadherins, selectins, orNCAMS); see, e.g., U.S. Pat. No. 7,556,928, which is incorporated herein by reference in its entirety.
  • Cell surface molecules on an effector cell include but not limited to CD3, CD4, CD5, CD8, CD16, CD27, CD28, CD38, CD64, CD89, CD134, CD137, CD154, CD226, CD278, NKp46, NKp44, NKp30, NKG2D, and an invariant TCR.
  • the cell surface molecule-binding domain of an engager molecule can provide activation to immune effector cells.
  • immune cells have different cell surface molecules.
  • CD3 is a cell surface molecule on T-cells
  • CD 16, NKG2D, or NKp30 are cell surface molecules on NK cells
  • CD3 or an invariant TCR are the cell surface molecules on NKT-cells.
  • Engager molecules that activate T-cells may therefore have a different cell surface molecule-binding domain than engager molecules that activate NK cells.
  • the activation molecule is one or more of CD3, e.g., CD3y, CD35 or CD3a; or CD27, CD28, CD40, CD134, CD137, and CD278.
  • the cell surface molecule is CD 16, NKG2D, or NKp30, or wherein the immune cell is aNKT-cell, the cell surface molecule is CD3 or an invariant TCR.
  • CD3 comprises three different polypeptide chains (a, 5 and y chains), is an antigen expressed by T cells.
  • the three CD3 polypeptide chains associate with the T-cell receptor (TCR) and the ⁇ -chain to form the TCR complex, which has the function of activating signaling cascades in T cells.
  • TCR T-cell receptor
  • the CD3 specific antibody OKT3 is the first monoclonal antibody approved for human therapeutic use, and is clinically used as an immunomodulator for the treatment of allogenic transplant rejections.
  • the heterologous protein reduces neutralization of the recombinant oncolytic virus by the immune system of the individual.
  • the recombinant oncolytic virus is an enveloped virus (e.g., vaccinia virus), and the heterologous protein is a complement activation modulator (e. g. , CD55 or CD59) .
  • Complement is a key component of the innate immune system, targeting the virus for neutralization and clearance from the circulatory system. Complement activation results in cleavage and activation of C3 and deposition of opsonic C3 fragments on surfaces. Subsequent cleavage of C5 leads to assembly of the membrane attack complex (C5b, 6, 7, 8, 9), which disrupts lipid bilayers.
  • recombinant oncolytic virus is an enveloped virus (e.g., vaccinia virus), and the heterologous protein is a complement activation modulator such as CD55, CD59, CD46, CD35, factor H, C4-binding protein, or other identified complement activation modulators.
  • a complement activation modulator such as CD55, CD59, CD46, CD35, factor H, C4-binding protein, or other identified complement activation modulators.
  • expression of the complement activation modulators on the virus envelope surface results in a virus having the ability to modulate complement activation and reduce complement- mediated virus neutralization as compared to the wild-type virus.
  • the heterologous nucleotide sequence encodes a domain of human CD55, CD59, CD46, CD35, factor H, C4-binding protein, or other identified complement activation modulators.
  • the heterologous nucleic acid encodes a CD55 protein that comprises an amino acid sequence having the sequence of SEQ ID NO: 58.
  • complement activation modulators e.g. CD59, CD46, CD35, factor H, C4-binding protein etc
  • enveloped recombinant oncolytic viruses e.g., vaccinia virus
  • the heterologous protein is a cytokine.
  • the heterologous protein is IL- 15, IL-12, IL-2, IL- 18, CXCL10, or CCL4, or a modified protein (e.g., a fusion protein) derived from of any of the aforementioned proteins.
  • the heterologous protein is a derivative of IL-2 that is modified to have reduced side effects.
  • the heterologous protein is modified IL- 18 that lacks binding to IL18-BP.
  • the heterologous protein is a fusion protein comprising an inflammatory cytokine and a stabilizing domain.
  • the stabilizing domain can be any suitable domain that stabilizes the inhibitory polypeptide.
  • the stabilizing domain extends the half-life of the inhibitory polypeptide in vivo.
  • the stabilizing domain is an Pc domain.
  • the stabilizing domain is an albumin domain.
  • the Fc domain is selected from the group consisting of Fc fragments of IgG, IgA, IgD, IgE, IgM, and combinations and hybrids thereof.
  • the Fc domain is derived from a human IgG.
  • the Fc domain comprises the Fc domain of human IgGl, IgG2, IgG3, IgG4, or a combination or hybrid IgG.
  • the Fc domain has a reduced effector function as compared to corresponding wildtype Fc domain (such as at least about 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, or 95% reduced effector function as measured by the level of antibody-dependent cellular cytotoxicity (ADCC)).
  • ADCC antibody-dependent cellular cytotoxicity
  • the inflammatory cytokine and the stabilization domain are fused to each other via a linker, such as a peptide linker.
  • a peptide linker may have a naturally occurring sequence, or a non-naturally occurring sequence. For example, a sequence derived from the hinge region of heavy chain only antibodies may be used as the linker.
  • the peptide linker can be of any suitable length.
  • the peptide linker tends not to adopt a rigid three-dimensional structure, but rather provide flexibility to a polypeptide.
  • the peptide linker is a flexible linker.
  • Exemplary flexible linkers include glycine polymers, glycine-serine polymers, glycine-alanine polymers, alanine -serine polymers, and other flexible linkers known in the art.
  • the heterologous protein is a bacterial or a viral polypeptide.
  • the heterologous protein is a tumor-associated antigen selected from carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR1, WT1, NY-ESO-1, CDH17, and other tumor antigens with clinical significance.
  • the recombinant oncolytic virus comprises two or more additional nucleotide sequences, wherein each nucleotide sequence encodes any one of the heterologous proteins or nucleic acids described herein.
  • Antagonist as used herein, is interchangeable with inhibitor.
  • the heterologous protein is an inhibitor (i.e., an antagonist) of a target protein, wherein the target protein is an immune suppressive protein (e.g., a checkpoint inhibitor or other inhibitor of immune cell activation).
  • the target protein is an immune checkpoint protein.
  • the target protein is PD-1, PD-L1, PD-L2, CD47, CXCR4, CSF1R, LAG-3, TIM-3, HHLA2, BTLA, CD160, CD73, CTLA-4, B7-H4, TIGIT, VISTA, or 2B4.
  • the target protein is CTLA-4, PD-1, PD-L1, B7-H4, or HLA-G.
  • the target protein is an immune suppressive receptor selected from LILRB, TYRO3, AXL, or MERTK.
  • the antagonist inhibits the expression and/or activity of the target protein (e.g., an immune suppressive receptor or an immune checkpoint protein).
  • the antagonist inhibits expression of the target protein (e.g., mRNA or protein level) by at least about any one of 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more.
  • Expression levels of a target protein can be determined using known methods in the art, including, for example, quantitative Polymerase Chain Reaction (qPCR), microarray, and RNA sequencing for determining RNA levels; and Western blots and enzyme-linked immunosorbent assays (ELISA) for determining protein levels.
  • the antagonist inhibits activity (e.g., binding to a ligand or receptor of the target protein, or enzymatic activity) of the target protein by at least about any one of 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more. Binding can be assessed using known methods in the art, including, for example, Surface Plasmon Resonance (SPR) assays, and gel shift assays.
  • SPR Surface Plasmon Resonance
  • the antagonist may be of any suitable molecular modalities, including, but are not limited to, small molecule inhibitors, oligopeptides, peptidomimetics, RNAi molecules (e.g., small interfering RNAs (siRNA), short hairpin RNAs (shRNA), microRNAs (miRNA)), antisense oligonucleotides, ribozymes, proteins (e.g., antibodies, inhibitory polypeptides, fusion proteins, etc.), and gene editing systems.
  • RNAi molecules e.g., small interfering RNAs (siRNA), short hairpin RNAs (shRNA), microRNAs (miRNA)
  • antisense oligonucleotides e.g., proteins, inhibitory polypeptides, fusion proteins, etc.
  • proteins e.g., antibodies, inhibitory polypeptides, fusion proteins, etc.
  • the antagonist inhibits binding of the target protein (e.g., an immune checkpoint protein or immune suppressive protein) to a ligand or a receptor.
  • the antagonist is an antibody that specifically binds to the target protein (e.g., CTLA-4, PD-1, PD-L1, B7-H4, HLA-G, LILRB, TYRO3, AXL, or MERTK, Folate receptor beta, etc ), or an antigen-binding fragment thereof.
  • the antagonist is a polyclonal antibody.
  • the antagonist is a monoclonal antibody.
  • the antagonist is a full-length antibody, or an immunoglobulin derivative.
  • the antagonist is an antigen-binding fragment.
  • antigen-binding fragments include, but are not limited to, a single-chain Fv (scFv), a Fab, a Fab’, a F(ab’)2, a Fv, a disulfide stabilized Fv fragment (dsFv), a (dsFv)2, a single-domain antibody (e.g., VHH), a Fv-Fc fusion, a scFv-Fc fusion, a scFv-Fv fusion, a diabody, a tribody, and a tetrabody.
  • the antagonist is a scFv.
  • the antagonist is a Fab or Fab’. In some embodiments, the antagonist is a chimeric, human, partially humanized, fully humanized, or semi-synthetic antibody. Antibodies and/or antibody fragments may be derived from murine antibodies, rabbit antibodies, human antibodies, fully humanized antibodies, camelid antibody variable domains and humanized versions, shark antibody variable domains and humanized versions, and camelized antibody variable domains.
  • the antagonist is a bi-specific molecule (e.g., a bi-specific antibody or bi-specific Fab, bi-specific scFv, antibody-Fc fusion protein Fv, etc) or a tri-specific molecule (e.g., a tri-specific antibody comprised of Fab, scFv, VH or Fc fusion proteins etc.).
  • a bi-specific molecule e.g., a bi-specific antibody or bi-specific Fab, bi-specific scFv, antibody-Fc fusion protein Fv, etc
  • a tri-specific molecule e.g., a tri-specific antibody comprised of Fab, scFv, VH or Fc fusion proteins etc.
  • the antibody comprises one or more antibody constant regions, such as human antibody constant regions.
  • the heavy chain constant region is of an isotype selected from IgA, IgG, IgD, IgE, and IgM.
  • the human light chain constant region is of an isotype selected from K and X.
  • the antibody comprises an IgG constant region, such as a human IgGl, IgG2, IgG3, or IgG4 constant region.
  • an antibody comprising a human IgGl heavy chain constant region or a human IgG3 heavy chain constant region may be selected.
  • an antibody comprising a human IgG4 or IgG2 heavy chain constant region, or IgGl heavy chain with mutations, such as N297A/Q, negatively impacting FcyR bindings may be selected.
  • the antibody comprises a human IgG4 heavy chain constant region.
  • the antibody comprises an S241P mutation in the human IgG4 constant region.
  • the antibody comprises an Fc domain.
  • Fc region refers to a C-terminal non-antigen binding region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226 to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present, without affecting the structure or stability of the Fc region.
  • the antibody comprises a variant Fc region has at least one amino acid substitution compared to the Fc region of a wild type IgG or a wild-type antibody.
  • the antibody is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • Antibodies that specifically bind to a target protein can be obtained using methods known in the art, such as by immunizing a non-human mammal and obtaining hybridomas therefrom, or by cloning a library of antibodies using molecular biology techniques known in the art and subsequence selection or by using phage display. ii. Nucleic acid agents
  • the heterologous nucleic acid is a nucleic acid agent that downregulates the target protein.
  • the antagonist inhibits expression (e.g. , mRNA or protein expression) of the target protein.
  • the antagonist is a siRNA, a shRNA, a miRNA, an antisense oligonucleotide, or a gene editing system.
  • the antagonist is an RNAi molecule. In some embodiments, the antagonist is a siRNA. In some embodiments, the antagonist is a shRNA. In some embodiments, the antagonist is a miRNA.
  • RNAi refers to biological process in which RNA molecules inhibit gene expression or translation by specific binding to a target mRNA molecule. See for example Zamore et al., 2000, Cell, 101, 25-33; Bass, 2001, Nature, 411, 428-429; Elbashir et al., 2001, Nature, 411, 494-498; and Kreutzer et al., International PCT Publication No. WO 00/44895; Zemicka-Goetz et al., International PCT Publication No.
  • RNAi molecules include siRNA, miRNA and shRNA.
  • a siRNA can be a double-stranded polynucleotide molecule comprising self- complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleotide sequence or a portion thereof.
  • the siRNA comprises one or more hairpin or asymmetric hairpin secondary structures.
  • the siRNA may be constructed in a scaffold of a naturally occurring miRNA.
  • the siRNA molecules need not be limited to those molecules containing only RNA, but further encompasses chemically modified nucleotides and non-nucleotides.
  • RNAi may be designed using known methods in the art.
  • siRNA may be designed by classifying RNAi sequences, for example 1000 sequences, based on functionality, with a functional group being classified as having greater than 85% knockdown activity and a non-functional group with less than 85% knockdown activity.
  • the distribution of base composition was calculated for entire the entire RNAi target sequence for both the functional group and the non-functional group.
  • the ratio of base distribution of functional and nonfunctional group may then be used to build a score matrix for each position of RNAi sequence. For a given target sequence, the base for each position is scored, and then the log ratio of the multiplication of all the positions is taken as a final score .
  • the target sequence may be filtered through both fast NCBI blast and slow Smith Waterman algorithm search against the Unigene database to identify the gene-specific RNAi or siRNA. Sequences with at least one mismatch in the last 12 bases may be selected.
  • the antagonist is an antisense oligonucleotide, e.g., antisense RNA, DNA or PNA.
  • the antagonist is a ribozyme.
  • An “antisense” nucleic acid refers to a nucleotide sequence complementary to a “sense” nucleic acid encoding a target protein or fragment (e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence).
  • the antisense nucleic acid can be complementary to an entire coding strand, or to a portion thereof or a substantially identical sequence thereof.
  • the antisense oligonucleotide can be complementary to the region surrounding the translation start site of the mRNA, e.g., between the -10 and +10 regions of the target gene nucleotide sequence of interest.
  • the antisense nucleic acid molecule is antisense to a “noncoding region” of the coding strand of a nucleotide sequence.
  • An antisense oligonucleotide can be, for example, about 7, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more nucleotides in length.
  • An antisense nucleic acid can be constructed using chemical synthesis or enzyme ligation reactions using standard procedures.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used).
  • Antisense nucleic acid also can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation.
  • An antisense nucleic acid is a ribozyme in some embodiments.
  • a ribozyme having specificity for a target nucleotide sequence can include one or more sequences complementary to such a nucleotide sequence, and a sequence having a known catalytic region responsible for mRNA cleavage (e.g., U.S. Pat. No. 5,093,246 or Haselhoff and Gerlach, Nature 334: 585-591 (1988)).
  • a derivative of a Tetrahymena L-19 IVS RNA is sometimes utilized in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an mRNA (e.g., Cech et al. U.S. Pat. No. 4,987,071; and Cech et al. U.S. Pat. No. 5,116,742).
  • Target mRNA sequences may be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules (e.g., Bartel & Szostak, Science 261: 1411-1418 (1993)).
  • the antagonist is a gene-editing system, such as a CRISPR/Cas gene editing system, Transcription activator-like effector nuclease or TAUEN gene editing system, Zine-finger gene editing system, etc.
  • the antagonist is a geneediting system that knocks-down a target protein, e.g., in a tissue-specific manner.
  • the antagonist is a gene-editing system that silences expression of the target protein.
  • the gene-editing system comprises a guided nuclease such as an engineered (e.g., programmable or targetable) nuclease to induce gene editing of a target sequence (e.g., DNA sequence or RNA sequence) encoding the target protein.
  • a guided nucleases such as an engineered (e.g., programmable or targetable) nuclease to induce gene editing of a target sequence (e.g., DNA sequence or RNA sequence) encoding the target protein.
  • Any suitable guided nucleases can be used including, but not limited to, CRISPR-associated protein (Cas) nucleases, zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TAEENs), meganucleases, other endo- or exo-nucleases, variants thereof, fragments thereof, and combinations thereof.
  • Cas CRISPR-associated protein
  • ZFNs zinc finger nucleases
  • TAEENs transcription activator-
  • the gene-editing system comprises a guided nuclease fused to a transcription suppressor. In some embodiments, the gene-editing system further comprises an engineered nucleic acid that hybridizes to a target sequence encoding the target protein. In some embodiments, the gene-editing system is a CRISPR-Cas system comprising a Cas nuclease (e.g., Cas9) and a guide RNA (i.e., gRNA).
  • Cas nuclease e.g., Cas9
  • gRNA guide RNA
  • nucleotide sequences encoding heterologous proteins (e.g., sialidase and multispecific immune cell engagers) or nucleic acids described herein can be operably linked to a promoter.
  • at least a first nucleotide sequence encoding the sialidase and a second nucleotide sequence encoding a multispecific immune cell engager are operably linked to the same promoter.
  • at least a first nucleotide sequence encoding the sialidase and an additional nucleotide sequence encoding an additional heterologous protein or nucleic acid are operably linked to the same promoter.
  • all of the nucleic acids encoding the heterologous proteins or nucleic acids are operably linked to the same promoter. In some embodiments, all of the nucleic acids encoding the heterologous proteins or nucleic acids are operably linked to different promoters.
  • the promoter is a viral promoter.
  • Viral promoters can include, but are not limited to, VV promoter, poxvirus promoter, adenovirus late promoter, Cowpox ATI promoter, or T7 promoter.
  • the promoter may be a vaccinia virus promoter, a synthetic promoter, a promoter that directs transcription during at least the early phase of infection, a promoter that directs transcription during at least the intermediate phase of infection, a promoter that directs transcription during early/late phase of infection, or a promoter that directs transcription during at least the late phase of infection.
  • the promoter comprises the nucleic acid sequence of SEQ ID NO: 107.
  • the promoter described herein is a constitutive promoter. In some embodiments, the promoter described herein is an inducible promoter.
  • Promoters suitable for constitutive expression in mammalian cells include but are not limited to the cytomegalovirus (CMV) immediate early promoter (US 5,168,062), the RSV promoter, the adenovirus major late promoter, the phosphoglycerate kinase (PGK) promoter (Adra et al., 1987, Gene 60: 65-74), the thymidine kinase (TK) promoter of herpes simplex virus (HSV)-l and the T7 polymerase promoter (W098/10088).
  • Vaccinia virus promoters are particularly adapted for expression in oncolytic poxviruses.
  • Representative examples include without limitation the vaccinia 7.5K, H5R, 11K7.5 (Erbs etal., 2008, Cancer Gene Ther. 15(1): 18-28), TK, p28, pll, pB2R, pA35R and K1L promoters, as well as synthetic promoters such as those described in Chakrabarti e/ al. (1997, Biotechniques 23: 1094-7; Hammond etal, 1997, J. Virol Methods 66: 135-8; and Kumar and Boyle, 1990, Virology 179: 151-8) as well as early/late chimeric promoters.
  • Promoters suitable for oncolytic measles viruses include without limitation any promoter directing expression of measles transcription units (Brandler and Tangy, 2008, CIMID 31: 271).
  • Inducible promoters belong to the category of regulated promoters.
  • the inducible promoter can be induced by one or more conditions, such as a physical condition, microenvironment of the host cell, or the physiological state of the host cell, an inducer (i.e., an inducing agent), or a combination thereof.
  • Appropriate promoters for expression can be tested in vitro (e.g. in a suitable cultured cell line) or in vivo (e.g. in a suitable animal model or in the subject).
  • the encoded immune checkpoint modulator(s) comprise(s) an antibody and especially a mAb
  • suitable promoters for expressing the heavy component of said immune checkpoint modulator comprise CMV, SV and vaccinia virus pH5R, F17R and pllK7.5 promoters
  • suitable promoters for expressing the light component of said immune checkpoint modulator comprise PGK, beta-actin and vaccinia virus p7.5K, F17R and pA35R promoters.
  • Promoters can be replaced by stronger or weaker promoters, where replacement results in a change in the attenuation of the virus.
  • replacement of a promoter with a stronger promoter refers to removing a promoter from a genome and replacing it with a promoter that effects an increased the level of transcription initiation relative to the promoter that is replaced.
  • a stronger promoter has an improved ability to bind polymerase complexes relative to the promoter that is replaced.
  • an open reading frame that is operably linked to the stronger promoter has a higher level of gene expression.
  • replacement of a promoter with a weaker promoter refers to removing a promoter from a genome and replacing it with a promoter that decreases the level of transcription initiation relative to the promoter that is replaced.
  • a weaker promoter has a lessened ability to bind polymerase complexes relative to the promoter that is replaced.
  • an open reading frame that is operably linked to the weaker promoter has a lower level of gene expression.
  • the viruses can exhibit differences in characteristics, such as attenuation, as a result of using a stronger promoter versus a weaker promoter.
  • vaccinia virus synthetic early/late and late promoters are relatively strong promoters
  • vaccinia synthetic early, P7.5k early/late, P7.5k early, and P28 late promoters are relatively weaker promoters (see e.g., Chakrabarti et al. (1997) BioTechniques 23 (6) 1094-1097).
  • the promoter described herein is a weak promoter.
  • the promoter described herein is a strong promoter.
  • the promoter is a viral promoter of the oncolytic virus.
  • the promoter is an early viral promoter, a late viral promoter, an intermediate viral promoter, or an early/late viral promoter.
  • the promoter is a synthetic viral promoter, such as a synthetic early, early/late, or late viral promoter.
  • the promoter is a vaccinia virus promoter.
  • Exemplary vaccinia viral promoters for use in the present invention can include, but are not limited to, P?.5k, Piik, PSE, PSEL, PSL, H5R, IK, P28, C11R, G8R, F17R, I3L, I8R, AIL, A2L, A3L, H1L, H3L, H5L, H6R, H8R, DIR, D4R, D5R, D9R, DHL, D12L, D13L, MIL, N2L, P4b or KI promoters.
  • Exemplary vaccinia early, intermediate and late stage promoters include, for example, vaccinia P?.5k early/late promoter, vaccinia PEL early/late promoter, vaccinia P13 early promoter, vaccinia Pi ik late promoter and vaccinia promoters listed elsewhere herein.
  • Exemplary synthetic promoters include, for example, PSE synthetic early promoter, PSEL synthetic early/late promoter, PSL synthetic late promoter, vaccinia synthetic promoters listed elsewhere herein (Patel et al., Proc. Natl. Acad. Set.
  • the promoter directs transcription during at least the late phase of infection (such as F17R promoter, shown in SEQ ID NO: 61) is employed.
  • the late promoter is selected from the group consisting of F17R, I2L late promoter, L4R late promoter, P?.5k early/late promoter, PEL early/late promoter, Pi ik late promoter, PSEL synthetic early/late promoter, and PSL synthetic late promoter.
  • the late vaccinia viral promoter F17R is only activated after VV infection in tumor cells, thus tumor selective expression of the heterologous protein or nucleic acid from VV will be further enhanced by the use of F17R promoter.
  • the late expression of a heterologous protein or nucleic acid of the present invention allows for sufficient viral replication before T-cell activation and mediated tumor lysis.
  • the promoter is a hybrid promoter.
  • the hybrid promoter is a synthetic early/late viral promoter.
  • the promoter comprises a partial or complete nucleotide sequence of a human promoter.
  • the human promoter is a tissue or tumor-specific promoter.
  • the tumor-specific promoter can be a promoter that drives enhanced expression in tumor cells, or that drives expression specifically in tumor cells (e.g. , a promoter that drives expression of a tumor tumor-associated antigen (TAA) or a tumor-specific antigen (TSA)).
  • TAA tumor tumor-associated antigen
  • TSA tumor-specific antigen
  • the hybrid promoter comprises a partial or complete nucleotide sequence of a tissue or tumor-specific promoter and a nucleotide sequence (e.g., a CMV promoter sequence) that increase the strength of the hybrid promoter relative to the tissue- or tumorspecific promoter.
  • a nucleotide sequence e.g., a CMV promoter sequence
  • hybrid promoters comprising tissue- or tumor-specific promoters include hTERT and CMV hybrid promoters or AFP and CMV hybrid promoters.
  • the one or more promotors comprise a first promoter that is operably linked to the first nucleotide sequence and a second promoter that is operably linked to the second nucleotide sequence.
  • the first promoter is an F17R promoter and the second promoter is a pE/L promoter.
  • the F17R promoter comprised the nucleic acid sequence of SEQ ID NO: 61.
  • the pE/L promoter comprises the nucleic acid sequence of SEQ ID NO: 107.
  • the engineered immune cells can be T-cells (e.g., apT cells or yST cells), natural killer (NK) cells, natural killer T (NKT) cells, dendritic cells (DC), cytokine -induced killer (CIK) cells, cytokine-induced natural killer (CINK) cells, lymphokine-activated killer (LAK) cells, tumor-infiltrating lymphocytes (TILs), macrophages, peripheral blood mononuclear cells (PBMCs) or combinations thereof.
  • T-cells e.g., apT cells or yST cells
  • NK natural killer
  • NKT natural killer T
  • DC dendritic cells
  • CIK cytokine -induced killer
  • CINK cytokine-induced natural killer
  • LAK lymphokine-activated killer
  • TILs tumor-infiltrating lymphocytes
  • macrophages es
  • PBMCs peripheral blood mononuclear cells
  • the cell therapy comprises PBMC cells that have been stimulated with various cytokine and antibody combinations to activate effector T cells (CD3, CD38 and IL-2) or, in some cases, T cells and NK cells (CD3, CD28, IL-15 and IL-21).
  • the engineered immune cells are CAR-T, CAR-NK, CAR-macrophage or CAR-NKT cells.
  • the immune cell is selected from the group consisting of a cytotoxic T cell, a helper T cell, a suppressor T cell, an NK cell, an NK-T cell, and combinations thereof.
  • the engineered immune cell is an NK cell.
  • the engineered immune cell is a T cell.
  • the engineered immune cell is an NKT cell.
  • engineered immune cells described herein comprise one or more engineered chimeric receptors, which are capable of activating an immune cell (e.g., T cell or NK cell) directly or indirectly against a tumor cell expressing a target antigen.
  • engineered receptors include, but are not limited to, chimeric antigen receptor (CAR), engineered T cell receptor, and TCR fusion protein.
  • the engineered immune cells are autologous cells (cells obtained from the subject to be treated). In some embodiments, the engineered immune cells are allogeneic cells, which can include a variety of readily isolable and/or commercially available cells/cell lines. Chimeric antisen receptor (CAR)
  • Chimeric antigen receptor refers to an engineered receptor that can be used to graft one or more target-binding specificities onto an immune cell, such as T cells, macrophages, and/or NK cells.
  • the chimeric antigen receptor comprises an extracellular target binding domain, a transmembrane domain, and an intracellular signaling domain of a T cell receptor and/or other receptors.
  • the engineered immune cells described herein comprise a chimeric antigen receptor (CAR).
  • the CAR comprises an antigenbinding moiety and an effector protein or fragment thereof comprising a primary immune cell signaling molecule or a primary immune cell signaling domain that activates the immune cell expressing the CAR directly or indirectly.
  • the CAR comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.
  • an engineered immune cells e.g., T cell or NK cell comprising the CAR.
  • the antigen-binding moiety and the effector protein or fragment thereof may be present in one or more polypeptide chains.
  • CAR constructs have been described, for example, in US9765342B2, W02002/077029, and WO2015/142675, which are hereby incorporated by reference. Any one of the known CAR constructs may be used in the present application.
  • the primary immune cell signaling molecule or primary immune cell signaling domain comprises an intracellular domain of a molecule selected from the group consisting of CD3 ⁇ , FcRy, FcR , CD3y, CD35, CD3s. CD5, CD22, CD79a, CD79b, and CD66d.
  • the intracellular signaling domain consists of or consists essentially of a primary immune cell signaling domain.
  • the intracellular signaling domain comprises an intracellular signaling domain of CD3 ⁇ .
  • the CAR further comprises a costimulatory molecule or fragment thereof.
  • the costimulatory molecule or fragment thereof is derived from a molecule selected from the group consisting of CD27, CD28, 4-1BB, 0X40, CD30, CD40, PD-1, ICOS, LFA-1, CD2, CD7, LIGHT, NKG2C, B7-H3, and a ligand that specifically binds CD83.
  • the intracellular signaling domain further comprises a co-stimulatory domain comprising a CD28 intracellular signaling sequence.
  • the intracellular signaling domain comprises a CD28 intracellular signaling sequence and an intracellular signaling sequence of CD3 ⁇ .
  • the transmembrane domain may be derived either from a natural or from a synthetic source. Where the source is natural, the domain may be derived from any membrane -bound or transmembrane protein. Transmembrane regions of particular use in this invention may be derived from (i.e. comprise at least the transmembrane region(s) of) the CD28, CD3a, CD3 ⁇ , CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154.
  • the CAR is a CD-19 CAR comprising including CD19 scFv from clone FMC63 (Nicholson IC, et al.
  • the transmembrane domain may be synthetic, in which case it may comprise predominantly hydrophobic residues such as leucine and valine.
  • a triplet of phenylalanine, tryptophan and valine may be found at each end of a synthetic transmembrane domain.
  • a short oligo- or polypeptide linker having a length of, for example, between about 2 and about 10 (such as about any of 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acids in length may form the linkage between the transmembrane domain and the intracellular signaling domain.
  • the linker is a glycine-serine doublet.
  • the transmembrane domain that is naturally associated with one of the sequences in the intracellular domain is used (e.g., if an intracellular domain comprises a CD28 co-stimulatory sequence, the transmembrane domain is derived from the CD28 transmembrane domain).
  • the transmembrane domain can be selected or modified by amino acid substitution to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins to minimize interactions with other members of the receptor complex.
  • the intracellular signaling domain of the CAR is responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR has been placed in.
  • Effector function of a T cell for example, may be cytolytic activity or helper activity including the secretion of cytokines.
  • intracellular signaling domain refers to the portion of a protein, which transduces the effector function signal and directs the cell to perform a specialized function. While usually the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain.
  • intracellular signaling sequence is thus meant to include any truncated portion of the intracellular signaling domain sufficient to transduce the effector function signal.
  • intracellular signaling domains for use in the CAR of the present application include the cytoplasmic sequences of the TCR and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any synthetic sequence that has the same functional capability.
  • T cell activation can be said to be mediated by two distinct classes of intracellular signaling sequence: those that initiate antigen-dependent primary activation through the TCR (primary signaling sequences) and those that act in an antigen-independent manner to provide a secondary or co-stimulatory signal (co-stimulatory signaling sequences).
  • Primary signaling sequences regulate primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way.
  • Primary signaling sequences that act in a stimulatory manner may contain signaling motifs, which are known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • ITAMs immunoreceptor tyrosine-based activation motifs or ITAMs.
  • the CAR constructs in some embodiments comprise one or more ITAMs. Examples of ITAM containing primary signaling sequences that are of particular use in the invention include those derived from CD3 ⁇ , FcRy, FcR[3, CD3y, CD35, CD3s. CD5, CD22, CD79a, CD79b, and CD66d.
  • the CAR comprises a primary signaling sequence derived from CD3 ⁇ .
  • the intracellular signaling domain of the CAR can comprise the CD3 ⁇ intracellular signaling sequence by itself or combined with any other desired intracellular signaling sequence(s) useful in the context of the CAR described herein.
  • the intracellular domain of the CAR can comprise a CD3 ⁇ intracellular signaling sequence and a costimulatory signaling sequence.
  • the costimulatory signaling sequence can be a portion of the intracellular domain of a costimulatory molecule including, for example, CD27, CD28, 4- 1BB (CD137), 0X40, CD30, CD40, ICOS, lymphocyte function-associated antigen-1 (LFA- 1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, and the like.
  • a costimulatory molecule including, for example, CD27, CD28, 4- 1BB (CD137), 0X40, CD30, CD40, ICOS, lymphocyte function-associated antigen-1 (LFA- 1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, and the like.
  • the intracellular signaling domain of the CAR comprises the intracellular signaling sequence of CD3 ⁇ and the intracellular signaling sequence of CD28. In some embodiments, the intracellular signaling domain of the CAR comprises the intracellular signaling sequence of CD3 ⁇ and the intracellular signaling sequence of 4- IBB. In some embodiments, the intracellular signaling domain of the CAR comprises the intracellular signaling sequence of CD3 ⁇ and the intracellular signaling sequences of CD28 and 4-1BB. [0286] In some embodiments, the antigen binding moiety comprises an scFv or a Fab.
  • the antigen binding moiety is targeted to a tumor-associated or tumor-specific antigen, such as, without limitation: carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR1, WT1, NY-ESO-1, CDH17, and other tumor antigens with clinical significance.
  • a tumor-associated or tumor-specific antigen such as, without limitation: carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2,
  • the antigen binding moiety is directed to a foreign antigen that is delivered to tumor cells (e.g., by a recombinant oncolytic virus).
  • the foreign antigen is DAS 181 or its derivatives (e.g. a transmembrane form of the sialidase domain of DAS181 without anchoring domain, as described in Examples 11 and 15).
  • the sialidase domain (e.g., a non-human sialidase or a derivative thereof, such as a sialidase domain of DAS 181) delivered to tumor cells using an oncolytic virus functions both to remove sialic acid from the surface of tumor cells and as a foreign antigen that enhances immune cell -mediated killing of tumor cells.
  • the sialidase-armed oncolytic virus is combined with an engineered immune cell that specifically targets the sialidase domain (e.g., DAS 181), thereby enhancing killing of tumor cells infected by the oncolytic virus.
  • engineered immune cells such as lymphocytes, e.g., T cells, NK cells
  • a method of producing an engineered immune cell expressing any one of the CARs described herein comprising introducing a vector comprising a nucleic acid encoding the CAR into the immune cell.
  • introducing the vector into the immune cell comprises transducing the immune cell with the vector.
  • introducing the vector into the immune cell comprises transfecting the immune cell with the vector. Transduction or transfection of the vector into the immune cell can be carried about using any method known in the art.
  • the chimeric receptor is a T cell receptor.
  • the T cell receptor is an endogenous T cell receptor.
  • the engineered immune cell with the TCR is pre-selected.
  • the T cell receptor is a recombinant TCR.
  • the TCR is specific for a tumor antigen.
  • the tumor antigen is selected from the group consisting of carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican- 3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR1, WT1, NY-ESO- 1, Fibulin-3, CDH17, and other tumor antigens with clinical significance.
  • TCRs specific for tumor antigens include tumor-associated antigens
  • TCRs for tumor antigens in melanoma e.g., MARTI , gp 100
  • leukemia e.g., WT1, minor histocompatibility antigens
  • breast cancer HER2, NY-BR1, for example.
  • the TCR has an enhanced affinity to the tumor antigen.
  • Exemplary TCRs and methods for introducing the TCRs to immune cells have been described, for example, in US5830755, and Kessels et al. Immunotherapy through TCR gene transfer. Nat. Immunol. 2, 957-961 (2001).
  • the engineered immune cell is a TCR-T cell.
  • TCR fusion protein TCP
  • the engineered immune cell comprises a TCR fusion protein (TFP).
  • TCR fusion protein or “TFP” as used herein refers to an engineered receptor comprising an extracellular target-binding domain fused to a subunit of the TCR-CD3 complex or a portion thereof, including TCRa chain, TCRp chain, TCRy chain, TCRS chain, CD3s, CD38, or CD3y.
  • the subunit of the TCR-CD3 complex or portion thereof comprise a transmembrane domain and at least a portion of the intracellular domain of the naturally occurring TCR-CD3 subunit.
  • the TFP comprises the extracellular domain of the TCR-CD3 subunit or a portion thereof.
  • TFP constructs comprising an antibody fragment as the target-binding moiety have been described, for example, in WO2016187349 and WO2018098365, which are hereby incorporated by reference.
  • Engineered immune cells can be targeted to any of a variety of tumor-associated antigens (TAAs) or immune cell receptors, which may include without limitation: EGFRvIII, PD-L1, EpCAM, carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD- 19, etc.
  • TAAs tumor-associated antigens
  • engineered immune cells can be used to deliver recombinant oncolytic viruses provided herein to cancer cells expressing these or any number of known cancer antigens.
  • engineered immune cells can be targeted to a foreign antigen (e.g., a bacterial peptide or a bacterial sialidase) that is delivered to tumor cells using a recombinant oncolytic virus.
  • Engineered immune cells can also be targeted to a variety of immune cells expressing various immune cell antigens, such as, without limitation: carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR1, WT1, NY-ESO- 1, Fibulin-3, CDH17, and other tumor antigens with clinical significance
  • a foreign antigen e.g., a bacterial peptide or a bacterial sialidase
  • Engineered immune cells
  • Engineered immune cells can be delivered to the patient in any way known in the art for delivering engineered immune cells (e.g. , CART-T, CAR-NK, or CAR-NKT cells).
  • sialidase expressed on the surface of or secreted by sialidase expressing engineered immune cells may remove sialic acids from sialoglycans expressed on immune cells and/or tumor cells. The removal of the sialic acid on tumor cell can further activate the Dendritic cells, macrophages, T and NK cell that are no longer engaged with the inihibitory signals of the tumor cells via Siglecs/sialic acid axis and other Selectins interactions.
  • TEE tumor microenvironment
  • the engineered immune cells set forth herein can be engineered to express sialidase, such as, without limitation, sialidase domain of DAS 181 fused to a transmembrane domain, on the immune cell surface membrane, such that the sialidase is membrane bound.
  • sialidase can be fused to a e.g. transmembrane domain.
  • membrane bound sialidases will not be freely circulating and will only come into contact with the target cells of the CAR-T, namely tumor cells expressing the antigens that the CAR-T receptor targets.
  • the CAR- T is a CD- 19 receptor or mAb to CD 19 expressing CAR-T
  • the membrane bound sialidases will primarily only come into contact with tumor cells that express CD- 19.
  • the sialidases will not desialylate non-targeted cells, such as erythrocytes, but will instead eliminate sialic acid primarily only from tumor cells.
  • the CAR-Ts set forth herein can also be engineered so that they express secreted sialidase, such as, without limitation, secreted form of DAS 181.
  • the present application provides a carrier cell comprising any one of the recombinant oncolytic viruses described herein.
  • the carrier cell is an immune cell or a stem cell (e.g., a mesenchymal stem cell).
  • the immune cell is an engineered immune cell, such as any of the engineered immune cells described in subsection C above.
  • the population of carrier cells e.g., immune cells or stem cells
  • the sialidase containing virus may be administered in any appropriate physiologically acceptable cell carrier.
  • the multiplicity of infection will generally be in the range of about 0.001 to 1000, e.g., in the range of 0.001 to 100.
  • the virus-containing cells may be administered one or more times.
  • viral DNA may be used to transfect the effector cells, employing liposomes, general transfection methods that are well known in the art (such as calcium phosphate precipitation and electroporation), etc. Due to the high efficiency of transfection of viruses, one can achieve a high level of modified cells.
  • the engineered immune cell comprising the recombinant oncolytic virus can be prepared by incubating the immune cell with the virus for a period of time.
  • the immune cell can be incubated with the virus for a time sufficient for infection of the cell with virus, and expression of the one or more virally encoded heterologous protein(s) (e.g., sialidase and/or any of the immunomodulatory proteins described herein).
  • the one or more virally encoded heterologous protein(s) e.g., sialidase and/or any of the immunomodulatory proteins described herein.
  • the population of carrier cells e.g., immune cells or stem cells
  • the recombinant oncolytic virus may be injected into the recipient. Determination of suitability of administering cells of the invention will depend, inter alia, on assessable clinical parameters such as serological indications and histological examination of tissue biopsies.
  • a pharmaceutical composition is administered. Routes of administration include systemic injection, e.g. intravascular, subcutaneous, or intraperitoneal injection, intratumor injection, etc.
  • the present application provides methods of treating a cancer (e.g., solid tumor) in an individual in need thereof, comprising administering to the individual an effective amount of any one of the recombinant oncolytic viruses, pharmaceutical compositions, or engineered immune cells described herein.
  • a cancer e.g., solid tumor
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a sialidase, wherein the nucleotide sequence encoding the heterologous protein is operably linked to a promoter.
  • the oncolytic virus is a vaccinia virus, reovirus, Seneca Valley virus (SVV), vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), herpes simplex virus (HSV), morbillivirus virus, retrovirus, influenza virus, Sinbis virus, poxvirus, measles virus, cytomegalovirus (CMV), lentivirus, adenovirus, or coxsackievirus, or a derivative thereof.
  • the oncolytic vims is Talimogene Laherparepvec.
  • the oncolytic vims is a reovims.
  • the oncolytic vims is an adenovims (e.g, an adenovims having an E1ACR2 deletion).
  • the oncolytic vims is a poxvims.
  • the poxvims is a vaccinia vims.
  • the vaccinia vims is of a strain such as Dryvax, Lister, M63, LIVP, Tian Tan, Modified Vaccinia Ankara, New York City Board of Health (NY CBOH), Dairen, Ikeda, LC16M8, Tashkent, IHD-J, Brighton, Dairen I, Connaught, Elstree, Wyeth, Copenhagen, Western Reserve, Elstree, CL, Lederle-Chorioallantoic, or AS, or a derivative thereof.
  • the vims is vaccinia vims Western Reserve.
  • the recombinant oncolytic vims is administered via a carrier cell (e.g., an immune cell or stem cell, such as a mesenchymal stem cell).
  • a carrier cell e.g., an immune cell or stem cell, such as a mesenchymal stem cell.
  • the recombinant oncolytic vims is administered as a naked vims.
  • the recombinant oncolytic vims is administered via intratumoral injection.
  • the method comprises administering a recombinant oncolytic vims comprising a nucleotide sequence encoding a sialidase, wherein the nucleotide sequence encoding the heterologous protein is operably linked to a promoter, and wherein the recombinant oncolytic vims comprises one or more mutations that reduce immunogenicity of the vims compared to a corresponding wild-type strain.
  • the vims is a vaccinia vims (e.g., a vaccinia vims Western Reserve), and the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R or other immunogenic proteins (e.g., A14, A17, A13, LI, H3, D8, A33, B5, A56, L13, A28, and A27). In some embodiments, the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R.
  • the one or more mutations are in one or more proteins selected from the group consisting of A27L, H3L, D8L and L1R.
  • the vims comprises one or more proteins selected from the group consisting of: (a) a variant vaccinia vims (VV) H3L protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to any one of SEQ ID NOS: 66-69; (b) a variant vaccinia vims (VV) D8L protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to any one of SEQ ID NOS: 70-72 or 85; (c) a variant vaccinia vims (VV) A27L protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to SEQ ID NO: 73; and (d) a variant vaccinia vims (VV) L1R protein that comprises an amino acid sequence having at least 90% amino acid sequence identity to SEQ ID NO: 74.
  • the method comprises administering a recombinant oncolytic vims comprising a nucleotide sequence encoding a sialidase, wherein the sialidase is operably linked to a promoter.
  • the sialidase is aNeu5Ac alpha(2,6)-Gal sialidase or a Neu5Ac alpha(2,3)-Gal sialidase.
  • the sialidase is a bacterial sialidase (e.g., a Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase) or a derivative thereof.
  • bacterial sialidase e.g., a Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase
  • the sialidase comprises all or a portion of the amino acid sequence of a large bacterial sialidase or can comprise amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to all or a portion of the amino acid sequence of a large bacterial sialidase.
  • the sialidase domain comprises SEQ ID NO: 2 or 27, or a sialidase sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 12.
  • a sialidase domain comprises the catalytic domain of the Actinomyces viscosus sialidase extending from amino acids 274-666 of SEQ ID NO: 26, having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% sequence identity to amino acids 274-666 of SEQ ID NO: 26.
  • the sialidase is a human sialidase (e.g., NEU1, NEU2, NEU3, or NEU4), or a derivative thereof.
  • a human sialidase e.g., NEU1, NEU2, NEU3, or NEU4
  • the sialidase is a naturally occurring sialidase. In some embodiments, the sialidase is a fusion protein comprising a sialidase catalytic domain.
  • the sialidase comprises an anchoring moiety.
  • the sialidase is a fusion protein comprising a sialidase catalytic domain fused to an anchoring domain.
  • the anchoring domain is positively charged at physiologic pH.
  • the anchoring domain is a glycosaminoglycan (GAG)- binding domain.
  • the sialidase comprises an amino acid sequence having at least about 80% (e.g. , at least about 85%, 90%, or 95%) sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-33, 53-54, and 105. In some embodiments, the sialidase comprises an amino acid sequence having at least about 80% (e.g., at least about 85%, 90%, or 95%) sequence identity to the amino acid sequence of SEQ ID NO: 1. In some embodiments, the sialidase is DAS 181 or a derivative thereof.
  • the nucleotide sequence encoding the sialidase including a secretory peptide e.g., a signal sequence or signal peptide operably linked to the sialidase.
  • the secretion sequence comprises the amino acid sequence of any one of SEQ ID NOS: 40, 102, and 103.
  • the sialidase comprises a transmembrane domain.
  • the anchoring domain or the transmembrane domain is located at the carboxy terminus of the sialidase.
  • a method of treating cancer in an individual in need thereof comprising administering an effective amount of a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having at least any one of 85%, 90%, 95%, or 100% identity to the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager having at least any one of 85%, 90%, 95%, or 100% identity the amino acid sequence of SEQ ID NO: 100.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • the cancer is an FAP positive cancer, and/or is associated with FAP positive stromal cells.
  • FAP is expressed by primary cancer cells or is expressed by tumor-associated fibroblasts (also referred to as cancer- associated fibroblasts).
  • the cancer is selected from lung cancer, colon cancer, and breast cancer.
  • administering the recombinant oncolytic virus activates and/or expands CD4 + and/or CD8 + T-cells in the individual.
  • administering the recombinant oncolytic virus increases tumor-infiltrating lymphocytes in the individual.
  • a method of treating cancer in an individual in need thereof comprising administering an effective amount of a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having at least any one of 85%, 90%, 95%, or 100% identity to the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager comprising a first scFv that recognizes FAP and a second scFv that recognizes CD3.
  • the first scFv comprises: (i) a first light chain complementarity-determining region (CDR-L1) having the amino acid sequence of SEQ ID NO: 86, (i) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 87, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 88, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 89, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 90, and (vi) a third heavy chain complementarity-determining region (CDR-H3) having the amino acid sequence of SEQ ID NO: 91.
  • CDR-L1 first light chain complementarity-determining region having the amino acid sequence of SEQ ID NO: 86
  • CDR-L2 having the
  • the first scFv comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 98. In some embodiments, the first scFv comprises the amino acid sequence of SEQ ID NO: 98.
  • the second scFv comprises: (i) a first light chain complementarity-determining region (CDR-L1) having the amino acid sequence of SEQ ID NO: 92, (ii) a second light chain complementarity-determining region (CDR-L2) having the amino acid sequence of SEQ ID NO: 93, (iii), a third light chain complementarity-determining region (CDR-L3) having the amino acid sequence of SEQ ID NO: 94, (iv) a first heavy chain complementarity-determining region (CDR-H1) having the amino acid sequence of SEQ ID NO: 95, (v) a second heavy chain complementarity-determining region (CDR-H2) having the amino acid sequence of SEQ ID NO: 96, and (vi) a third heavy chain complementarity-determining region (CDR-H3) having the amino acid sequence of SEQ ID NO: 97.
  • CDR-L1 having the amino acid sequence of SEQ ID NO: 92
  • CDR-L2 having the amino acid sequence of SEQ ID NO
  • the second scFv comprises an amino acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99% identity to the amino acid sequence of SEQ ID NO: 99. In some embodiments, the second scFv comprises the amino acid sequence of SEQ ID NO: 99. In some embodiments, the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters. In some embodiments, the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • TK thymidine kinase
  • VVF vaccinia growth factor
  • the cancer is an FAP positive cancer.
  • FAP is expressed by primary cancer cells or is expressed by tumor-associated fibroblasts (also referred to as cancer-associated fibroblasts).
  • the cancer is selected from lung cancer, colon cancer, and breast cancer.
  • administering the recombinant oncolytic virus activates and/or expands CD4 + and/or CD8 + T- cells in the individual.
  • administering the recombinant oncolytic virus increases tumor-infiltrating lymphocytes in the individual.
  • a method of treating cancer in an individual in need thereof comprising administering an effective amount of a recombinant vaccinia virus of a Western Reserve strain comprising a first nucleotide sequence encoding a sialidase having at least any one of 85%, 90%, 95%, or 100% identity to the amino acid sequence of SEQ ID NO: 105, and a second nucleotide sequence encoding a bispecific immune cell engager comprising a first scFv that recognizes FAP and a second scFv that recognizes CD3.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • the cancer is an FAP positive cancer.
  • FAP is expressed by primary cancer cells or is expressed by tumor-associated fibroblasts (also referred to as cancer-associated fibroblasts).
  • the cancer is selected from lung cancer, colon cancer, and breast cancer.
  • administering the recombinant oncolytic virus activates and/or expands CD4 + and/or CD8 + T- cells in the individual.
  • administering the recombinant oncolytic virus increases tumor-infiltrating lymphocytes in the individual.
  • a method of treating cancer in an individual in need thereof comprising administering an effective amount of a recombinant vaccinia virus of a Western Reserve strain comprising a nucleic acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 99%, or 100% identity to the nucleic acid sequence of SEQ ID NO: 108.
  • the first nucleotide sequence and the second nucleotide sequence are operably linked to one or more promoters.
  • the recombinant vaccinia virus comprises a disruption of a thymidine kinase (TK) gene and a disruption of a vaccinia growth factor (VGF) gene.
  • TK thymidine kinase
  • VVF vaccinia growth factor
  • the nucleic acid sequence having at least any one of 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 99%, or 100% identity to the nucleic acid sequence of SEQ ID NO: 108 is integrated into the TK gene of the vaccinia virus.
  • the cancer is an FAP positive cancer.
  • FAP is expressed by primary cancer cells or is expressed by tumor- associated fibroblasts (also referred to as cancer-associated fibroblasts).
  • the cancer is selected from lung cancer, colon cancer, and breast cancer.
  • administering the recombinant oncolytic virus activates and/or expands CD4 + and/or CD8 + T- cells in the individual.
  • administering the recombinant oncolytic virus increases tumor-infiltrating lymphocytes in the individual.
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual an effective amount of a carrier cell (e.g., an immune cell or a stem cell such as a mesenchymal stem cell) comprising a recombinant oncolytic virus, wherein the recombinant oncolytic virus comprises a nucleotide sequence encoding a sialidase.
  • a carrier cell e.g., an immune cell or a stem cell such as a mesenchymal stem cell
  • the recombinant oncolytic virus comprises a nucleotide sequence encoding a sialidase.
  • the sialidase is a bacterial sialidase (e.g., a Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase) or a derivative thereof.
  • the sialidase is derived from a Actinomyces viscosus sialidase.
  • the sialidase is DAS181 or a derivative thereof.
  • the nucleotide sequence encoding the sialidase further encodes a secretion sequence (e.g, a secretory sequence or secretory peptide) operably linked to the sialidase.
  • the molecule comprises a sialidase linked to a transmembrane domain.
  • the carrier cell is an engineered immune cell.
  • the engineered immune cell expresses a chimeric receptor, such as a CAR.
  • the chimeric receptor specifically recognizes a tumor associated antigen and encoded other molecule that can stimulate antitumor immune response and tumor killing functions.
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual: (a) an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a sialidase, or an effective amount of carrier cells comprising the recombinant oncolytic virus; and (b) an effective amount of engineered immune cells expressing a chimeric receptor.
  • the sialidase is a bacterial sialidase (e.g., Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase).
  • the sialidase comprises an anchoring domain.
  • the anchoring domain is a GAG- binding protein domain, e.g., the epithelial anchoring domain of human amphiregulin.
  • the anchoring domain is positively charged at physiologic pH.
  • the anchoring domain is a GPI linker.
  • the sialidase is DAS 181. In some embodiments, the sialidase comprises a transmembrane domain. In some embodiments, the chimeric receptor recognizes a tumor-associated antigen or tumor-specific antigen. In some embodiments, the engineered immune cells are T cells or NK cells. In some embodiments, the chimeric receptor is a CAR.
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual: (a) an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a sialidase, or an effective amount of carrier cells comprising the recombinant oncolytic virus; and (b) an effective amount of engineered immune cells expressing a chimeric receptor specifically recognizing the sialidase.
  • the sialidase is a bacterial sialidase (e.g., Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter ureafaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase).
  • the sialidase comprises an anchoring domain.
  • the anchoring domain is a GAG-binding protein domain, e.g., the epithelial anchoring domain of human amphiregulin.
  • the anchoring domain is positively charged at physiologic pH.
  • the anchoring domain is a GPI linker.
  • the sialidase is DAS 181. In some embodiments, the sialidase comprises a transmembrane domain. In some embodiments, the chimeric receptor specifically recognizes the sialidase (e.g., DAS 181) and is not cross-reactive with human native amphiregulin or any other human antigen. In some embodiments, the engineered immune cells are T cells or NK cells. In some embodiments, the chimeric receptor is a CAR.
  • a method of delivering a foreign antigen to cancer cells in an individual comprising administering to the individual an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a foreign antigen.
  • the foreign antigen is a bacterial protein.
  • the foreign antigen is a sialidase.
  • the foreign antigen is a bacterial sialidase (e.g., Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase).
  • the sialidase is a sialidase catalytic domain of DAS 181.
  • the method further comprises administering engineered immune cells.
  • the engineered immune cells express a chimeric receptor specifically recognizing the foreign antigen or any relevant tumor associated antigen or tumor specific antigen of the tumor being treated.
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual: (a) an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a foreign antigen; and (b) an effective amount of engineered immune cells expressing a chimeric receptor specifically recognizing said foreign antigen.
  • a method of treating cancer comprising administering to the individual: (a) an effective amount of a recombinant oncolytic virus comprising a first nucleotide sequence encoding a sialidase and a second nucleotide sequence encoding a multispecific immune cell engager; and (b) an effective amount of an immunotherapeutic agent.
  • the immunotherapeutic agent is a cell therapy.
  • the immunotherapeutic agent is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is an inhibitor of CTLA-4, PD-1, PD-L1, B7-H4, TIGIT, LAG3, TIM-3, VISTA, or HLA-G.
  • a method of sensitizing a tumor in an individual to an immunotherapy comprising administering to the individual an effective amount of any one of the recombinant oncolytic viruses comprising a nucleotide sequence encoding a sialidase and a multispecific immune cell engager described above.
  • the sialidase is a bacterial sialidase (e.g., a Clostridium perfringens sialidase, Actinomyces viscosus sialidase, and Arthrobacter urecifaciens sialidase, Salmonella typhimurium sialidase or Vibrio cholera sialidase) or a derivative thereof.
  • the sialidase is derived from a Actinomyces viscosus sialidase. In some embodiments, the sialidase is DAS 181. In some embodiments, the nucleotide sequence encoding the sialidase further encodes a secretion sequence (e.g., a secretory signal peptide) operably linked to the sialidase. In some embodiments, the sialidase further comprises a transmembrane domain. In some embodiments, the method further comprises administering an effective amount of the immunotherapy to the individual.
  • the immunotherapy is a multi-specific immune cell engager (e.g., a bispecific molecule), a cell therapy, a cancer vaccine (e.g., a dendritic cell (DC) cancer vaccine), a cytokine (e.g., IL-15, IL- 12, modified IL-2 having no or reduced binding to the alpha receptor, modified IL- 18 with no or reduced binding to IL- 18 BP, CXCL10, or CCL4), an immune checkpoint inhibitor (e.g., an inhibitor of CTLA-4, PD-1, PD-L1, B7-H4, or HLA), a master switch anti-LILRB, and bispecific anti-LILRB-4-lBB, Anti-FAP-CD3, a PI3Kgamma inhibitor, a TLR9 ligand, an HDAC inhibitor, a LILRB2 inhibitor, a MARCO inhibitor, etc.
  • a cancer vaccine e.g., a dendritic cell (DC) cancer vaccine
  • the immunotherapeutic agent is a cell therapy.
  • a cell therapy comprises administering an effective amount of live cells (e.g. , immune cells) to the individual.
  • the immune cells can be T-cells, natural killer (NK) cells, natural killer T (NKT) cells, dendritic cells (DC), cytokine-induced killer (CIK) cells, cytokine- induced natural killer (CINK) cells, lymphokine-activated killer (LAK) cells, tumor-infiltrating lymphocytes (TILs), macrophages, or combinations thereof.
  • the cell therapy can comprise administering a developmental intermediate (e.g., a progenitor) of any one of the immune cell types described herein.
  • the cell therapy agents can comprise indiscrete heterogeneous cell populations, such as expanded PBMCs that have proliferated and acquired killing activity on ex vivo culture. Suitable cell therapies have been described, for example, in Hayes, C. “Cellular immunotherapies for cancer.” Ir J Med Sci (2020).
  • the cell therapy comprises PBMC cells that have been stimulated with various cytokine and antibody combinations to activate effector T cells (CD 3, CD38 and IL-2) or, in some cases, T cells and NK cells (CD3, CD28, IL-15 and IL-21).
  • the cell therapy comprises administering to the individual an effective amount of immune cells, wherein the immune cells have been primed to respond to a tumor antigen, e.g, by exposure to the antigen either in vivo or ex vivo.
  • the cell therapy comprises administering to the individual an effective amount of engineered immune cells expressing a chimeric receptor, such as any one of the chimeric receptors described in the “Engineered immune cells” section above.
  • the cell therapy comprises administering an effective amount of CAR-T, CAR- NK, or CAR-NKT cells.
  • the chimeric receptor recognizes an antigen expressed by tumor cells, such as an endogenous tumor-associated or tumor-specific antigen.
  • the chimeric receptor can recognize tumor antigens such as carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR1, WT1, NY-ESO-1, Fibulin-3, CDH17, and other tumor antigens with clinical significance.
  • tumor antigens such as carcinoembryonic antigen, alphafetoprotein, MUC16, survivin, glypican-3, B7 family members, LILRB, CD19, BCMA, NY-ESO-1, CD20, CD22, CD33, CD38, CEA, EGFR (such as EGFRvIII), GD2, HER2, IGF1R, mesothelin, PSMA, ROR
  • the chimeric receptor recognizes a foreign antigen expressed by tumor cells, such as a heterologous protein delivered to the tumor cells via any one of the recombinant oncolytic viruses provided herein.
  • the foreign antigen delivered by the recombinant oncolytic virus is a bacterial peptide or a bacterial sialidase, e.g., DAS181 (SEQ ID NO: 2).
  • the foreign antigen is a sialidase comprising a transmembrane domain.
  • the foreign antigen is DAS 181 without an AR tag and fused to a C-terminal transmembrane domain (e.g., SEQ ID NO: 31).
  • a method of increasing efficacy of an immunotherapy in an individual in need of the immunotherapy comprising administering an effective amount of a recombinant oncolytic virus encoding a sialidase and an effective amount of an immunotherapy.
  • the immunotherapy is a multi-specific immune cell engager (e.g., a BiTE), a cell therapy, a cancer vaccine (e.g., a dendritic cell (DC) cancer vaccine), a cytokine (e.g., IL-15, IL-12, modified IL-2, modified IL-18, CXCL10, or CCL4), and an immune checkpoint inhibitor (e.g., an inhibitor of CTLA-4, PD-1, PD-L1, B7-H4, TIGIT, LAG3, TIM3 or HLA-G).
  • a cancer vaccine e.g., a dendritic cell (DC) cancer vaccine
  • a cytokine e.g., IL-15, IL-12, modified IL-2, modified IL-18, CXCL10, or CCL4
  • an immune checkpoint inhibitor e.g., an inhibitor of CTLA-4, PD-1, PD-L1, B7-H4, TIGIT, LAG3, TIM3 or HLA-
  • the immunotherapy is cell therapy, e.g., a cell therapy comprising T-cells, natural killer (NK) cells, natural killer T (NKT) cells, dendritic cells (DC), cytokine -induced killer (CIK) cells, cytokine-induced natural killer (CINK) cells, lymphokine-activated killer (LAK) cells, tumor-infiltrating lymphocytes (TILs), macrophages, or combinations thereof.
  • the recombinant oncolytic virus is administered before, after, or simultaneously with the immunotherapy.
  • administering the recombinant oncolytic vims increases tumor cell killing by at least 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, or 100% compared to the immunotherapy alone.
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual an effective amount of engineered immune cells, wherein the immune cells express a recombinant oncolytic vims encoding a heterologous protein.
  • the immune cells express a chimeric receptor that specifically recognizes a target molecule associated with the cancer.
  • the immune cells express a chimeric receptor that specifically recognizes the sialidase encoded by the vims.
  • a method of treating a cancer in an individual in need thereof comprising administering to the individual an effective amount of engineered immune cells, wherein the immune cells express a recombinant oncolytic vims encoding a heterologous protein, wherein the heterologous protein is a sialidase.
  • the immune cells express a chimeric receptor that specifically recognizes a target molecule associated with the cancer.
  • the immune cells express a chimeric receptor that specifically recognizes the sialidase encoded by the vims.
  • One aspect of the present application provides methods of reducing sialylation of cancer cells in an individual, comprising administering to the individual an effective amount of any one of the recombinant oncolytic vimses, pharmaceutical compositions, or engineered immune cells described above.
  • the sialidase reduces surface sialic acid on tumor cells.
  • the sialidase reduces surface sialic acid on tumor cells by at least 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, or 90%.
  • the sialidase cleaves both a2,3 and a2,6 sialic acids from the cell surface of tumor cells.
  • the sialidase increases cleavage of both a2,3 and a2,6 sialic acids by at least 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, or 90%.
  • a method of promoting an immune response in an individual comprising administering an effective amount of a recombinant oncolytic virus encoding a sialidase.
  • the method promotes a local immune response in a tumor microenvironment of the individual.
  • a method of promoting dendritic cell (DC) maturation in an individual comprising administering an effective amount of a recombinant oncolytic virus encoding a sialidase (e.g., DAS 181).
  • DC maturation can be determined based on the expression of dendritic cell markers, such as CD 80 and DC MHC I and MHC-II proteins.
  • the recombinant oncolytic virus increases DC maturation by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50%.
  • Example 9 provides results demonstrating increased DC maturation following administration of a recombinant oncolytic virus encoding a sialidase.
  • a method of increasing immune cell killing of tumor cells in an individual comprising administering an effective amount of a recombinant oncolytic virus encoding a sialidase.
  • the method increases killing by NK cells.
  • the recombinant oncolytic virus encoding a sialidase increases killing by NK cells by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50%.
  • the recombinant oncolytic virus encoding a sialidase increases killing by NK cells by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50% compared to recombinant oncolytic virus lacking sialidase.
  • Example 3 demonstrates enhanced NK cell-mediated killing of tumor cells with administration of a recombinant oncolytic virus encoding sialidase.
  • the method increases killing by T cells.
  • the recombinant oncolytic virus encoding a sialidase increases killing by T cells by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50% .
  • the recombinant oncolytic virus encoding a sialidase increases killing by T cells by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50% compared to recombinant oncolytic virus lacking sialidase.
  • Example 10 demonstrates enhanced NK cell-mediated killing of tumor cells with administration of a recombinant oncolytic virus encoding sialidase.
  • the method increases killing by PBMCs.
  • the recombinant oncolytic virus encoding a sialidase increases killing by PBMCs by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50%.
  • the recombinant oncolytic virus encoding a sialidase increases killing by PBMCs by at least at least 5%, 10%, 15%, 20%, 30%, 40%, or 50% compared to recombinant oncolytic virus lacking sialidase.
  • Example 6 demonstrates enhanced PBMC-mediated killing of tumor cells with administration of a recombinant oncolytic virus encoding sialidase.
  • a method of increasing oncolytic killing of an oncolytic virus in an individual comprising administering an effective amount of a sialidase.
  • the sialidase is encoded by the oncolytic virus.
  • oncolytic killing by a recombinant oncolytic virus encoding a sialidase is increased by at least at least 5%, 10%, 20%, 30%, 40%, or 50% compared to recombinant oncolytic virus lacking sialidase.
  • Example 5 provides results demonstrating enhanced oncolytic killing by a recombinant oncolytic virus encoding a sialidase.
  • a method of enhancing cytokine production and oncolytic activity in an individual comprising administering an effective amount of a recombinant oncolytic vims encoding a sialidase.
  • the method enhances cytokine production by T-lymphocytes.
  • method enhances T- lymphocyte mediated cytokine production locally in a tumor microenvironment of the individual.
  • the cytokines include IL2 and IFN-gamma.
  • administering recombinant oncolytic vims encoding a sialidase increases cytokine production by at least at least 5%, 10%, 20%, 30%, 40%, or 50% compared to administering an oncolytic vims lacking sialidase. In some embodiments, administering recombinant oncolytic vims encoding a sialidase increases IL2 production by at least 2.5-fold, at least 3 -fold, or at least 4-fold compared to administering an oncolytic vims lacking sialidase.
  • administering recombinant oncolytic vims encoding a sialidase increases IFN-gamma production by at least 5%, 10%, 20%, 30%, 40%, or 50% compared to administering an oncolytic vims lacking sialidase.
  • Example 10 demonstrates enhanced cytokine production and killing by T-lymphocytes following administration of a recombinant oncolytic vims encoding a sialidase.
  • cancer is a term for diseases caused by or characterized by any type of malignant tumor or hematological malignancy, including metastatic cancers, solid tumors , lymphatic tumors, and blood cancers.
  • Cancers include leukemias, lymphomas (Hodgkins and non-Hodgkins), sarcomas, melanomas, adenomas, carcinomas of solid tissue including breast cancer and pancreatic cancer, hypoxic tumors, squamous cell carcinomas of the mouth, throat, larynx, and lung, genitourinary cancers such as cervical and bladder cancer, hematopoietic cancers, head and neck cancers, and nervous system cancers, such as gliomas, astrocytomas, meningiomas, etc., benign lesions such as papillomas, and the like.
  • lymphomas Hodgkins and non-Hodgkins
  • sarcomas melanomas
  • adenomas carcinomas of solid tissue including breast cancer and pancreatic cancer
  • hypoxic tumors squamous cell carcinomas of the mouth, throat, larynx, and lung
  • genitourinary cancers such as cervical and bladder cancer
  • delivery of the sialidase can reduce sialic acid present on tumor cells and render the tumor cells more vulnerable to killing by immune cells, immune cell-based therapies and other therapeutic agents whose effectiveness is diminished by hyper sialylation of cancer cells.
  • the method further comprises administering to the individual an effective amount of an immunotherapeutic agent.
  • the immunotherapeutic agent can be a multi-specific immune cell engager, a cell therapy, a cancer vaccine, a cytokine, a PI3Kgamma inhibitor, a TLR9 ligand, an HD AC inhibitor, a LILRB2 inhibitor, a MARCO inhibitor, or an immune checkpoint inhibitor. Suitable immune cell engagers and immune checkpoint inhibitors are described in the “Other heterologous proteins or nucleic acids” subsection above.
  • the cancer comprises a solid tumor.
  • the cancer is an adenocarcinoma, a metastatic cancer and/or is a refractory cancer.
  • the cancer is a breast, colon or colorectal, lung, ovarian, pancreatic, prostate, cervical, endometrial, head and neck, liver, renal, skin, stomach, testicular, thyroid or urothelial cancer.
  • the cancer is an epithelial cancer, e.g., an endometrial cancer, ovarian cancer, cervical cancer, vulvar cancer, uterine cancer, fallopian tube cancer, breast cancer, prostate cancer, lung cancer, pancreatic cancer, urinary cancer, bladder cancer, head and neck cancer, oral cancer or liver cancer.
  • the cancer is selected from human alveolar basal epithelial adenocarcinoma, human mamillary epithelial adenocarcinoma, and glioblastoma.
  • the method comprises administering to the individual an effective amount of any one of the recombinant oncolytic viruses, pharmaceutical compositions, or engineered immune cells described above and an effective amount of engineered immune cells expressing a chimeric receptor.
  • the chimeric receptor targets a heterologous protein expressed by the recombinant oncolytic virus.
  • the heterologous protein is a sialidase (e.g., DAS181 or a derivative thereof, such as a membrane-bound form of DAS 181), and the chimeric receptor specifically recognizes the sialidase.
  • the sialidase is DAS 181 or a derivative thereof, and wherein the chimeric receptor comprises an anti-DAS 181 antibody that is not cross-reactive with human native amphiregulin or any other human antigen.
  • the present application provides a method of treating a tumor in an individual in need thereof comprising administering to the individual: (a) an effective amount of a recombinant oncolytic virus comprising a nucleotide sequence encoding a foreign antigen; and (b) an effective amount of an engineered immune cell expressing a chimeric receptor specifically recognizing said foreign antigen.
  • the foreign antigen is a non-human protein (e.g., a bacterial protein).
  • the engineered immune cells and the recombinant oncolytic virus are administered separately (e.g., as monotherapy) or together simultaneously (e.g. , in the same or separate formulations) as combination therapy.
  • the recombinant oncolytic virus is administered prior to administration of the engineered immune cells.
  • the recombinant oncolytic virus can be administered 1 or more, 2 or more, 4 or more, 6 or more, 8 or more, 10 or more, 12 or more, 24 or more, or 48 or more hours prior to the engineered immune cells comprising the chimeric receptor.
  • a population of engineered immune cells expressing the recombinant oncolytic virus is administered prior to a population of engineered immune cells expressing a chimeric antigen receptor targeting a heterologous protein expressed by the recombinant oncolytic virus.
  • the engineered immune cells comprising the recombinant oncolytic virus can be administered 1 or more, 2 or more, 4 or more, 6 or more, 8 or more, 10 or more, 12 or more, 24 or more, or 48 or more hours prior to the engineered immune cells comprising the chimeric receptor targeting a heterologous protein expressed by the recombinant oncolytic virus.
  • the time period between administration of the recombinant oncolytic virus (e.g., in a pharmaceutical composition or a carrier cell comprising the recombinant oncolytic virus) and administration of the engineered immune cells expressing the chimeric receptor is sufficient to allow the virus to express the heterologous protein or nucleic acid in the tumor cells.
  • the recombinant oncolytic virus, and in some embodiments, the engineered immune cells and/or additional immunotherapeutic agent(s) may be administered using any suitable routes of administration and suitable dosages.
  • the determination of the appropriate dosage or route of administration is well within the skill of an ordinary artisan. Animal experiments provide reliable guidance for the determination of effective doses for human therapy. Interspecies scaling of effective doses can be performed following the principles laid down by Mordenti, J. and Chappell, W. “The Use of Interspecies Scaling in Toxicokinetics,” In Toxicokinetics and New Drug Development, Yacobi et al., Eds, Pergamon Press, New York 1989, pp. 42-46.
  • the recombinant oncolytic virus, the engineered immune cells and/or additional immunotherapeutic agent(s) are administered sequentially (e.g., the recombinant oncolytic virus can be administered prior to the engineered immune cells, and/or prior to other therapeutic agents such as bi-specific antibody of FAP/CD3, bi-specific or trispecific antibody of LILRB-4-1BB, PD-1 antibody, etc as described above).
  • the recombinant oncolytic virus, the engineered immune cells and/or additional immunotherapeutic agent(s) are administered simultaneously or concurrently.
  • the recombinant oncolytic virus, the engineered immune cells and/or additional immunotherapeutic agent(s) are administered in a single formulation.
  • the recombinant oncolytic virus, the engineered immune cells and/or additional immunotherapeutic agent(s) are administered as separate formulations.
  • compositions, kits and articles of manufacture may be combined with conventional chemotherapeutic, radiologic and/or surgical methods of cancer treatment.
  • IV. Pharmaceutical compositions, kits and articles of manufacture
  • compositions comprising any one of the recombinant oncolytic viruses, carrier cells comprising a recombinant oncolytic virus, and/or engineered immune cells (s) described herein, and a pharmaceutically acceptable carrier.
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising an oncolytic virus (such as VV) comprising a first nucleotide sequence encoding a sialidase and/or any one of the other heterologous proteins or nucleic acids described herein, and an engineered immune cell expressing a chimeric receptor (e.g., a CAR-T, CAR-NK, or CAR-NKT cell) or any of the heterologous proteins or nucleic acids described herein that can modulate and enhance immune cell function such as anti LILRB, Anti-folate receptor beta, bispecific antibody such as anti-LILRB/4-lBB, etc.
  • an oncolytic virus such as VV
  • an engineered immune cell expressing a chimeric receptor (e.g., a CAR-T, CAR-NK, or CAR-NKT cell) or any of the heterologous proteins or nucleic acids described herein that can modulate and enhance immune cell function such as anti LILRB, Anti-folate
  • the present application provides a first pharmaceutical composition comprising a recombinant oncolytic virus (such as VV) comprising a first nucleotide sequence encoding a sialidase and/or any one of the other heterologous proteins or nucleic acids described herein, and optionally a pharmaceutically acceptable carrier; and a second pharmaceutical composition comprising an engineered immune cell expressing a chimeric receptor (e.g., a CAR-T, CAR-NK, or CAR-NKT cell), and optionally a pharmaceutically acceptable carrier.
  • a recombinant oncolytic virus such as VV
  • a second pharmaceutical composition comprising an engineered immune cell expressing a chimeric receptor (e.g., a CAR-T, CAR-NK, or CAR-NKT cell), and optionally a pharmaceutically acceptable carrier.
  • compositions can be prepared by mixing the recombinant oncolytic viruses and/or engineered immune cells described herein having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers, antioxidants including ascorbic acid, methionine, Vitamin E, sodium metabisulfite; preservatives, isotonicifiers (e.g. sodium chloride), stabilizers, metal complexes (e.g. Zn-protein complexes); chelating agents such as EDTA and/or non-ionic surfactants.
  • the formulation can include a carrier.
  • the carrier is a macromolecule which is soluble in the circulatory system and which is physiologically acceptable where physiological acceptance means that those of skill in the art would accept injection of said carrier into a patient as part of a therapeutic regime.
  • the carrier preferably is relatively stable in the circulatory system with an acceptable plasma half-life for clearance.
  • macromolecules include but are not limited to soy lecithin, oleic acid and sorbitan trioleate.
  • the formulations can also include other agents useful for pH maintenance, solution stabilization, or for the regulation of osmotic pressure. Examples of the agents include but are not limited to salts, such as sodium chloride, or potassium chloride, and carbohydrates, such as glucose, galactose or mannose, and the like.
  • the pharmaceutical composition is contained in a single-use vial, such as a single-use sealed vial. In some embodiments, the pharmaceutical composition is contained in a multi-use vial. In some embodiments, the pharmaceutical composition is contained in bulk in a container. In some embodiments, the pharmaceutical composition is cryopreserved.
  • the systems provided herein can be stably and indefinitely stored under cryopreservation conditions, such as, for example, at -80 °C, and can be thawed as needed or desired prior to administration.
  • cryopreservation conditions such as, for example, at -80 °C
  • the systems provided herein can be stored at a preserving temperature, such as - 20 °C or -80 °C, for at least or between about a few hours,.
  • 1, 2, 3, 4 or 5 hours, or days including at least or between about a few years, such as, but not limited to, 1 , 2, 3 or more years, for example for at least or about 1, 2, 3, 4 or 5 hours to at least or about 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 or 72 hours or 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or 30 days or 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5 or 12 months or 1, 2, 3, 4 or 5 or more years prior to thawing for
  • the systems provided herein also stably can be stored under refrigeration conditions such as, at 4 °C and/or transported on ice to the site of administration for treatment.
  • the systems provided herein can be stored at 4 °C or on ice for at least or between about a few hours, such as, but not limited to, 1 , 2, 3, 4 or 5 hours, to at least or about 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47 or 48 or more hours prior to administration for treatment.
  • kits and articles of manufacture for use in any embodiment of the treatment methods described herein.
  • the kits and articles of manufacture may comprise any one of the formulations and pharmaceutical compositions described herein.
  • kits comprising one or more nucleic acid constructs for expression any one of the recombinant oncolytic viruses described herein, and instructions for producing the recombinant oncolytic virus.
  • the kit further comprises instructions for treating a cancer.
  • kits comprising any one of the recombinant oncolytic viruses described herein, and instructions for treating a cancer.
  • the kit further comprises an immunotherapeutic agent (e.g., a cell therapy or any one of the immunotherapies described herein).
  • the kit further comprises one or more additional therapeutic agents for treating the cancer.
  • the antagonist, the recombinant oncolytic virus and/or the one or more immunotherapeutic agents are in a single composition (e.g., a composition comprising a cell therapy and a recombinant oncolytic virus).
  • the recombinant oncolytic virus and optionally the one or more additional immunotherapeutic agents and/or additional therapeutic agents for treating the cancer are in separate compositions.
  • kits of the invention are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Kits may optionally provide additional components such as buffers and interpretative information.
  • the present application thus also provides articles of manufacture, which include vials (such as sealed vials), bottles, jars, flexible packaging, and the like.
  • sialic acid is most often attached to the penultimate sugar by an a-2,3 linkage or an a-2,6 linkage, which can that can be detected by Maackia Amurensis Lectin II (MAL II) and Sambucus Nigra Lectin (SNA), respectively.
  • MAL II Maackia Amurensis Lectin II
  • SNA Sambucus Nigra Lectin
  • surface galactose e.g., galactose exposed after sialic acid removal
  • PNA Peanut Agglutinin
  • FIG 1 depicts the detection of a-2,6 sialic acid by FITC-SNA on A549 and MCF cells by fluorescence imaging.
  • A549 cells were treated with various concentrations of DAS 181 and them stained to image 2,6 linked sialic acid (FITC-SNA), a-2,3 linked sialic acid (FITC-MALII) or galactose (FITC-PNA).
  • FITC-SNA 2,6 linked sialic acid
  • FITC-MALII a-2,3 linked sialic acid
  • FITC-PNA galactose
  • DAS 185 a variant of DAS181 lacking sialidase activity due to Y348F mutation, was not able to remove a-2,6 linked sialic acid or a-2,3 linked sialic acid.
  • incubation of A549 cells with DAS 185 had essentially no impact on surface a-2,3 linked sialic acid, while DAS 181 reduced surface a-2,3 linked sialic acid in a concentration dependent manner (cells stained with FITC-MALII; results shown in FIG. 3).
  • Example 2 DAS181 Treatment Increases PBMC-Mediated Tumor Cell Killing
  • Example 1 demonstrated that DAS 181 effectively reduces the sialic acid burden of tumor cells with broad specificity (e.g., cleaving both a-2,3 vs. a-2,6 linkages).
  • Example 2 demonstrates that treatment of tumor cells with DAS 181 significantly enhances PBMC- mediated killing of the treated tumor cells compared to untreated tumor cells.
  • A549 cells were genetically labelled with a red fluorescent protein (A549-red).
  • Fresh human PBMCs were harvested and stimulated with various cytokine and antibody combinations to activate effector T cells (CD3, CD38 and IL-2) or, in some cases, T cells and NK cells (CD3, CD28, IL-15 and IL-21).
  • Activated PBMCs were then co-cultured with A549- red cells that had been exposed to DAS181 (100 nM). Tumor cell killing by PBMCs was monitored by live cell imaging and quantification with IncuCyte. The cell culture medium was collected and analyzed by ELISA to assess cytokine production by PBMCs.
  • FIG. 6 shows that neither the treatments used to stimulate PBMC nor DAS 181 in combination with treatment used to stimulate PBMC impact A549-red cell proliferation.
  • FIG. 7 shows that DAS 181 significantly increases tumor cell toxicity mediated by PBMC (Donor 1), both T cell mediated and NK cell mediated, compared to a vehicle only control. Similar results were observed using PBMC from a different donor (Donor 2; FIG. 8).
  • FIGS. 9A-C presents a quantification of the data presented in FIG. 7.
  • FIG. 9A shows quantification of A549-red cells following treatment with PBMCs with or without DAS 181 at the indicated effector cell : tumor cell ratios.
  • FIG. 9B shows quantification of A549-red cells following treatment with PBMCs stimulated with CD3, CD38 and IL-2 to activate effector T cells with or without DAS 181 at the indicated effector cell : tumor cell ratios.
  • FIG. 9C shows quantification of A549-red cells following treatment with PBMCs stimulated with CD3, CD28, IL- 15 and IL-21 to activate effector T and NK cells with or without DAS181 at the indicated effector cell : tumor cell ratios.
  • FIGS. 10A-10C show the same quantifications, respectively, using PBMCs from a different donor (Donor 2).
  • Example 3 NK Cell Mediated Killing of Tumor Cells by Oncolytic Vaccinia Virus and DAS181
  • DAS181 a variant protein lacking sialidase activity was used as a control.
  • monocyte-derived dendritic cells were prepared by resuspending 5xl0 6 adherent PBMC in 3 ml of medium supplemented with 100 ng/ml of GM-CSF and 50 ng/ml of IL-4. After 48 hrs, 2 ml of fresh medium supplemented with 100 ng/ml of GM-CSF and 50 ng/ml of IL-4 was added to each well. After another 72 hrs, tumor cells (U87-GFP) were plated in 24-well plates in DMEM. The tumor cells were infected with VV at various MOI in FBS free medium for 2 hours. DC cultured in the presence of InM DAS 181 or DAS 185 were mixed with tumor cells at 1: 1 tumor cell:DC ratio. Dendritic cell maturation (expression of CD86, CD80, MHC-II, MHC-I).
  • THP-1 cells were cultured in RPMI 1640 medium (Invitrogen) containing 10% heat-inactivated FBS.
  • THP-1 cells in a 6-well plate (3xl0e6 cells/well) were stimulated with PMA (20 ng/ml) in the absence and in the presence of InM of Sialidase DAS181 or DAS 185.
  • Cell culture medium volume was 2ml.
  • tumor cells U87- GFP, DMEM cell culture medium
  • Tumor were infected with VV at various MOI (i.e. 0.5, 1, 2) in FBS free medium for 2 hours.
  • MOI i.e. 0.5, 1, 2
  • the differentiated THP-1 cells were further stimulated for 12 h by ionomycin (lug/ml) and PMA (20 ng/ml) also in the absence and in the presence of InM of Sialidase DAS 181 or DAS 185 and tumor cells-VV at tumormacrophage ratio of 1 : 1.
  • the THP- 1 cells were cultured in medium supplemented with 2% FBS in order to decrease neuraminidase background. On day 6, the concentration of cytokine in the culture medium was measured by ELISA array.
  • DAS 181 significant enhanced expression of dendritic cell maturation markers whether the cells were cultured alone or with vaccinia virus infected tumor cells.
  • This Example provides unexpected results demonstrating that treatment with DAS181 increases oncolytic virus tumor cell killing, even in the absence of immune cells.
  • A549 cells were genetically labelled with red fluorescent protein (A549-red). Tumor cell proliferation and killing by oncolytic adenovirus (Ad5) in the presence or absence of DAS181 was monitored by live cell imaging and quantification with IncuCyte. The cell culture medium was collected for ELISA measurement of cytokine production by PBMCs. As shown in FIG 15, DAS181 increased oncolytic adenovirus-mediated tumor cell killing and growth inhibition.
  • Example 6 DAS181 Increases Oncolytic Adenovirus Tumor Cell Killing in the Presence of PBMC
  • Example 5 As shown in Example 5, treatment with DAS 181 increases killing of tumor cells by an oncolytic virus in the absence of immune cells.
  • Example 6 provides results demonstrating that treatment with DAS181 also increases tumor cell killing when present together with oncolytic virus in the presence of PBMC
  • A549 cells were genetically labelled by a red fluorescent protein (A549-red).
  • Fresh human PBMCs were harvested and stimulated with proper cytokine and antibody combinations to activate effector T cells.
  • Activated PBMCs were then co-cultured with A549-red cells that have been treated with DAS 181 with or without the oncolytic adenovirus (Ad5).
  • Tumor cell killing by PBMCs was monitored by live cell imaging and quantification with IncuCyte. The cell culture medium was collected for ELISA measurement of cytokine production by PBMCs.
  • DAS 181 significantly increased tumor cell killing when present together with oncolytic adenovirus in the presence of PBMC.
  • a construct designed for expression of DAS 181 is depicted schematically in FIG 17.
  • a pSEM-1 vector was modified to include a sequence encoding DAS 181 as well as two loxP sites (loxP site sequences are shown in SEQ ID NO: 62) with the same orientation flanking the sequence encoding the GFP protein (the GFP coding sequence is shown in SEQ ID NO: 63).
  • pSEM-l-TK-DAS181-GFP DAS181 expression is under the transcriptional control of the F17R late promoter in order to limit the expression within tumor tissue.
  • the sequence of a portion of an exemplary construct is shown in SEQ ID NO: 65.
  • Recombinant virus can be generated as follows.
  • CV-1 cells in 6-well plates at 5xl0 5 cells/2ml DMEM-10% FBS/well and grow overnight.
  • CV-1 cells should be 60-80% confluent when receiving cell lysate.
  • CV-1 cells 5xl0 5 cells/2ml DMEM-10% FBS/well and grow overnight in 6- well plate. CV-1 should be confluent when starting the experiment. Infect 1 well with 250 ul of plaque lysate/lml DMEM-2% FBS, and incubate at 37°C for 2 h. Remove the plaque lysate and add 2 ml fresh DMEM-2% FBS, and incubate for 48-72 hr until cells round up. Collect the cells by repeatedly pipetting, freeze-thaw 3 times and sonicate.
  • CV-1 cells were infected with VV-DAS181 at MOI 0.2. 48 hours later, CV-1 cells were collected. DNA was extracted using Wizard SV Genomic DAN Purification System and used as template for DAS181 PCR amplification. PCR was conducted using standard PCR protocol and primer sequences (SialF:
  • Example 8 DAS181 Expressed by Vaccinia Virus is Active In Vitro
  • Example 8 provides results demonstrating that delivery of DAS 181 to cells using an oncolytic virus results in sialidase activity equivalent to treatment with approximately 0.78nM- 1.21 nM of purified DAS181 in 1 ml medium.
  • CV-1 cells were plated in six well plate. The cells were transduced with Sialidase- VV or control VV at MOI 0.1 or MOI 1. After 24 hrs, transfected cells were collected, and single cell suspension were made in PBS at 3xl0 6 /500 pl. Cell lysate was prepared using Sigma’s Mammalian cell lysis kit for protein extraction (Sigma, MCL1-1KT), and supernatant was collected. The sialidase (DAS 181) activity was measured using Neuraminidase Assay Kit (Abeam, abl38888) according to manufacturer’s instruction. 1 nM, 2 nM, and 10 nM DAS181 was added to the VV-cell lysate as control and generated the standard curve.
  • DAS 181 Neuraminidase Assay Kit
  • IxlO 6 cells infected with Sialidase-VV express DAS 181 equivalent to 0.78nM-1.21 nM of DAS 181 in 1 ml medium. As shown in FIG 18, the DAS 181 has sialidase activity in vitro.
  • Example 9 Vaccinia Virus-Sialidase Promotes Dendritic Cell Maturation
  • Example 9 provides results demonstrating that an oncolytic virus encoding a sialidase promotes dendritic cell maturation compared to an oncolytic virus without a sialidase.
  • adherent human PBMC were re-suspend at 5xl0 6 cells in 3 ml medium supplemented with 100 ng/ml of GM-CSF and 50 ng/ml of IL-4 then cultured in 6-well plates with 2ml per well of fresh medium supplemented with same concentrations of GM-CSF and IL-4. 6 days post cell culture, the cells were cultured in the presence of Sialidase-VV infected tumor cell lysate, VV- infected tumor cell lysate, VV-infected tumor cell lysate plus synthetic DAS181 protein, or LPS (positive control).
  • Example 10 Sialidase-VV enhances T lymphocyte-mediated cytokine production and oncolytic activity
  • Sialidase-VV induces a significantly greater IL-2 and IFN-gamma expression by CD3 activated T cells than does VV.
  • Sialidase-VV elicits stronger anti-tumor response than VV at an E:T of 5: 1.
  • a DNA sequence encoding the signal peptide of the mouse Immunoglobulin kappa chain was added to the N-terminus of DAS 181 sequence by gene synthesis and then together cloned into a mammalian expression vector pcDNA3.4.
  • a DNA sequence encoding the DAS 181 catalytic domain was synthesized and cloned in-frame with the human PDGFR beta transmembrane domain in a mammalian expression vector pDisplay .
  • DNA sequences encoding secreted and transmembrane versions of DAS 185, a mutant protein lacking sialidase activity, were similarly synthesized and cloned into pcDNA3.4 and pDisplay vectors, respectively.
  • constructs expressing secreted and transmembrane versions of human Neu2 sialidases were generated in the same manner.
  • construct 1 secreted DAS181; SEQ ID NO: 34
  • construct 4 transmembrane DAS181; SEQ ID NO: 37
  • construct 2 secreted DAS185; SEQ ID NO: 35
  • construct 5 transmembrane DAS 185; SEQ ID NO: 38
  • construct 3 secreted human Neu2; SEQ ID NO: 36
  • construct 6 transmembrane human Neu2; SEQ ID NO: 39
  • Example 12 Enzymatic activity of secreted and transmembrane sialidases
  • mammalian expression vectors (detailed in Example 11) were transfected into HEK293 cells using jetPRIME transfection reagent (Polyplus Transfection #114-15) following the manufacturer’s protocol. Briefly, Human embryonic kidney cells (HEK293) were plated at ⁇ 2 x 10 5 live cells per well in 6-well tissue culture plates and grown to confluency by incubation at 37°C, 5% CO2, and 95% relative humidity (typically overnight). Two microliters equivalent to 2 micrograms ofDNA was diluted into 200 microliters jetPRIME Buffer followed by 4 microliters of jetPRIME reagent.
  • jetPRIME transfection reagent Polyplus Transfection #114-15
  • Tubes were vortexed, briefly centrifuged at 1,000 x g ( ⁇ 10 seconds) and incubated for 10 minutes at room temperature. During the incubation, the media on all wells was replenished with fresh culture media (MEM + 10% FBS). Transfections were added to individual wells and the plate returned to the incubator for 24 hours. Following incubation, supernatants were reserved. Single cell suspensions were created using non-enzymatic cell dissociation reagent Versene (Gibco #15040-066). Monolayers were washed 1 time with DPBS and 500 microliters Versene was added the plate incubated until cells dissociated from vessel surface; 500 microliters complete media was added and the cells were centrifuged for 5 minutes at 300xg.
  • the supernatant was aspirated and cells were suspended in 300 microliters compete media for enzymatic assay.
  • supernatant and resuspended cells were evaluated for activity utilizing the ability of the sialidase to enzymatically cleave the Anorogenic substrate, 2'-(4-Methylumbelliferyl)-a-D-N-acetylneuraminic acid sodium salt hydrate (MuNaNa) to release the Huorescent molecule 4-methylumbelliferone (4-Mu).
  • the resulting free 4-Mu is excited at 365 nM and the emission is read at 445 nm using a Huorescent plate reader.
  • BrieHy 100 pl of each sample was plated into a black, non-treated 96-well plate. The plate was incubated in a water bath at 37°C for approximately 30 minutes and subsequently mixed with pre-incubated (37°C, 30 minutes) 100 pM MuNaNa. The Huorescence was kinetically measured at 30 second intervals for 60 minutes using a Molecular Devices SpectraMax M5e multi-mode plate reader. The amount of 4-Mu generated by cleavage was quantified by comparison to a standard curve of pure 4-Mu, ranging from 100-5 pM. Reaction rates were determined for each sample by dividing the amount of 4-Mu produced ( ⁇ 20 pM) by the time (seconds) required to do so.
  • the observed reaction rates were compared to determine the approximate relative activity of each sample solution (Table 6). It was shown that the supernatant from the secreted DAS181 transfection and the resuspended cells from the transmembrane DAS181 transfection were the most active and approximately equal. All DAS 185 and Neu2 sample solutions showed negligible activity compared to the DAS181 sample solutions. The Neu2 sample solutions were equivalent to the background. Furthermore, the observed reaction rates were compared to a standard curve of known concentrations of DAS 181, ranging from 1000-60 pM.
  • the supernatant from the secreted DAS181 transfection and the resuspended cells from the transmembrane DAS 181 transfection were extrapolated to be approximately equivalent to 4000 pM DAS 181. All other samples were observed to be approximately equivalent to or less than 90 pM DAS 181.
  • **Cells were harvested, spun down and resuspended in 300 L media.
  • Example 13 Secreted DAS181 and transmembrane DAS181 reduce surface sialic acids on tumor cells
  • A549-Red cell were plated at 2 x 10 5 cells per well in 2 ml of A549-Red complete growth medium in 6-well plates.
  • plasmid DNA and 9 pl of Fugene HD were diluted into 150 pl of Opti-MEM® I Reduced Serum Medium, mixed gently and incubated for 5 minutes at room temperature to form DNA-Fugene HD complexes.
  • the above DNA-Fugene HD complexes directly to each well containing cells and the cells were incubated at 37°C in a CO2 incubator overnight before further experiments.
  • transfected cells were re-seeded as 8,000 cells per well in 96-well plates. Then cells were fixed and stained for a2,3-sialic acid; a2,6-sialic acid; and galactose following cell culture for 24 hr, 48 hr or 72 hr. Cells were incubated with SNA-FITC at 40pg/ml, PNA-FITC at 20pg/ml for Ih at room temperature to stain a2,6-sialic acid, and galactose, separately.
  • transfected cells were re-seeded at IxlO 5 cells per well in 24-well plates. Then cells are fixed and stained for a2,3-sialic acid, a2,6-sialic acid, and galactose following cell culture for 24 hr, 48 hr or 72 hr. Results were analyzed using Acea Flow cytometer system. The results of secreted construct transfections, with recombinant DAS181 treatment as control, are shown in FIGS. 22A-22C for a2,3 (FIG. 22A) and a2,6 (FIG. 22B) sialic acids, and galactose (FIG. 22C).
  • FIGS. 23A-23C The results of transmembrane construct transfections, with secreted DAS 181 transfection as control, are shown in FIGS. 23A-23C for a2,3 (FIG. 23A) and a2,6 (FIG. 23B) sialic acids, and galactose (FIG. 23C). Consistent with the imaging study results, secreted DAS 181 and transmembrane DAS181 transfections led to removal of cell surface a2,3 and a2,6 sialic acids, and exposure of galactose, whereas transfections with secreted and transmembrane DAS 185 or human Neu2 had little effect.
  • Example 14 Secreted DAS181 and transmembrane DAS181 increase tumor cell killing mediated by PBMC and oncolytic virus
  • A549-red parental cells were seeded as controls. The next day, the complete growth medium was removed and replaced with 50 ul of medium with or without oncolytic virus. Freshly isolated PBMC were counted and resuspended at 200,000/ml in A549 complete medium with anti-CD3/anti-CD28/IL2, then 50pl freshly PBMC were added to the cells. The cell growth was monitored by Essen Incucyte up to 5 days based on the counted red objects. As shown in FIG. 24, secreted DAS181 expression sensitized activated PBMC -mediated tumor cells killing and increased oncolytic virus associated PBMC- mediated cell killing at both MOI of 1 and 5. As shown in FIG.
  • transmembrane DAS 181 expression significantly sensitized A549-red cells to activated PBMC killing. A far greater effect was virus was observed at MOI of 5, than at MOI of 1. It is possible that the potency of sialidase activity and oncolytic virus as single agent could be masking the additive effect when they were combined together under certain experimental conditions.
  • This Example demonstrates generation of exemplary oncolytic virus constructs encoding sialidase. Constructs were successfully generated for Endo-Sial-VV, SP-Sial-VV, and TM-Sial-VV.
  • pSEM-1 vectors were created using gene synthesis.
  • the construct comprises of the gene encoding Sialidase, the gene encoding GFP or RFP, and two loxP sites with the same orientation flanking GFP/RFP (pSEM- 1-Sialidase-GFP/RFP).
  • the inserted Sialidase is under the transcriptional control of the F17R late promoter in order to limit the Sialidase expression within tumor tissue.
  • the simplified design of the plasmids is as show in FIG. 26.
  • Vaccinia virus (W) strain WR was used as the parental virus for recombination with Sialidase to create VVs that expresses Sialidase in three different isoforms: i) constrained to the intracellular compartment (Endo-Sial-VV); ii) secreted to the extracellular environment (SP-Sial-VV); or iii) localized at the cell surface (TM-Sial-VV).
  • Sialidase-VVs were generated by insertion of pSEM-1 -TK-Sialidase-GFP, pSEM-1- TK-SP-Sialidase-RFP or pSEM-1 -TK-TM-Sialidase-GFP into the TK gene of VV through homologous recombination. All the viruses were produced and quantified by titration on CV- 1 cells.
  • infectious particles were titrated by plaque assay. Briefly, CV-1 cells seeded in a 12-well plate were infected with serial dilutions of VV, endo-Sial-VV, SP-Sial-VV or TM-Sial-VV. After 48 h of infection, cells were fixed and stained with 20% Ethanol/ 0.1% Crystal Violet and virus plaques were counted. We prepared aliquots of 10 6 of each virus stock in 100 pl of 10 mM Tris-HCl pH 9.0 for shipping. Therefore, all viruses are at 10 7 pfii/ml. 1.2,2 Detection of virus recombination by PCR
  • PCR was performed according to standard protocols to amplify the constructs using each virus stock as the template DNA. To do so, PCR primers were designed to specifically bind to the regions shown in Figure 2. These primers will be able to confirm that: i) the constructs were successfully inserted into VV genome; ii) the constructs maintained their respective modifications during recombination (i.e. secretion and transmembrane domains).
  • the primer sequences used were the following:
  • Example 16 Sialidase- VVs’ are able to infect, replicate in, and lyse tumor cells in vitro.
  • This Example provides results demonstrating that Endo-Sial-VV, SP-Sial-VV, and TM-Sial-VV have comparable infectivity and replication activity in CV-1 and U87 cells, and comparable lytic activity in U87 and A549 cells to parental vaccinia virus, indicating the transgene didn’t impair the VV’s infectivity, replication, and lytic ability.
  • Tumor cells were infected with Sialidase-VV, or parental VV at increasing MOIs. At various time points (24, 48, 72 or 96 hours) post infection, the cells were harvested and subjected to plaque assay and MTS assays to determine virus replication.
  • the replication ability of the virus was not affected by modification with sialidase.
  • CV-1 or U87 cells were plated in 12-well tissue culture plate and infected with Sialidase-VVs or VV at MOIs 0.1 in 2.5% FBS medium for 2 hours followed by culturing in complete medium. At various time points post infection (24, 48, 72, or 96 hours), the cells were harvested and virus replication was determined by plaque assay using CV-1 cells.
  • the lytic activity of the modified vaccinia viruses was comparable to that of parental vaccinia virus in U87 and A549 cells, as shown in FIG. 29 and Tables 7-9 below.
  • GM-CSF/IL4 derived human DC (Astarte, WA) were cultured with VV-U87 tumor cells (ATCC, VA) for 24 hours. DC were collected and stained with antibodies against DC maturation markers CD86, CD80, HLA-ABC, HLA-Dr on DCs were determined by flow cytometry.
  • HLA-Dr-FITC Abeam, MA
  • HLA-ABC-PE abl55381, Abeam, MA
  • CD80-FITC abl8279, Abeam, MA
  • CD86- PE ab234226, Abeam, MA
  • FIGS. 30-33 show expression of DC maturation markers HLA-ABC, HLA-DR, CD80, and CD86, respectively. Culturing DCs together with U87 tumor cells infected with SP- Sial-VV or TM-Sial-VV enhanced expression of DC maturation markers compared to that of DC cells cultures with U87 infected with VV or U87 alone.
  • % 100 % x - target cells maximum release - target cells spontaneous release
  • Example 19 Sialidase-VVs inhibit tumor growth in vivo
  • Example 19 provides results demonstrating that Sialidase-VVs significantly inhibit tumor growth in vitro compared to control VV.
  • FIG. 35 shows the tumor size on the right flank.
  • TM-sial-VV significantly inhibited tumor growth compared to control VV.
  • SP-sial VV inhibited tumor growth, albeit to a lesser extent.
  • FIG. 36 shows the tumor size on the left flank. The results indicated that TM-sial-W significantly inhibited tumor growth compared to control VV.
  • FIG. 37 shows that there was no significant difference in mouse body weight for mice treated with the various VVs or PBS control.
  • 2xl0 5 and 2xl0 4 B16-F10 tumor cells were inoculated on the right or left flank of C57 mice.
  • 4xl0 7 pfu VVs were injected intratumorally every other day for 3 doses.
  • Sialidase armed oncolytic vaccinia virus significantly enhances CD8+ and CD4+ T cell infiltration within tumor
  • FIG. 38A shows quantification of the results and p values demonstrating significant enhancement of CD 8+ and CD4+ T cell infiltration by sialidase armed oncolytic vaccinia virus.
  • FIG. 38B shows the FACS plots. The results demonstrated that sialidase armed oncolytic vaccinia virus significantly enhanced CD8+ and CD4+ T cell infiltration within tumor compared to control vaccinia virus.
  • Sialidase armed oncolytic vaccinia virus significantly decreased the ratio of Treg/CD4+ T cells within the tumor
  • Sialidase armed oncolytic vaccinia virus significantly enhances NK and NKT cell infiltration within tumor
  • Sialidase armed oncolytic vaccinia virus significantly enhances NK and NKT cell infiltration within tumor
  • Example 20 Glycoimmune checkpoint and tumor stroma-targeted oncolytic vaccinia virus
  • This example describes the generation of a vvDD-Sial-FAP/CD3, an oncolytic vaccinia vims expressing a membrane-bound sialidase to remove sialic acids from the cell surface glycans, and fibroblast activation protein (FAP)-targeted T cell engager to eliminate glycoimmune checkpoint and tumor stroma, respectively.
  • a vvDD-Sial-FAP/CD3 was designed as an engineered vaccinia vims of Western Reserve (WR) strain with: (1) an insertional dismption of the viral thymidine kinase (TK) gene with the sialidase and FAP/CD3 transgenes.
  • TK viral thymidine kinase
  • TK is an essential enzyme for the pyrimidine synthesis pathway; viral TK gene deletion thus results in selective replication of vims in rapidly dividing cancerous cells with high intracellular nucleotide pools, and (2) a deletion of the vaccinia growth factor (VGF) gene for greater dependence on the cell cycling status of the cancer cells.
  • VVF vaccinia growth factor
  • the vvDD vims has been described (see McCart JA, et al. Systemic cancer therapy with a tumor- selective vaccinia vims mutant lacking thymidine kinase and vaccinia growth factor genes. Cancer Res 2001 ;61 : 8751-8757, the content of which is herein incorporated by reference in its entirety).
  • the nucleic acid constmct used to integrate the sialidase and FAP/CD3 T cell engager into the TK gene of vvDD comprised the nucleic acid sequence shown in SEQ ID NO: 108.
  • results showed that sialidase expressed from wDD-Sial-FAP/CD3 efficiently cleaves the sialic acids from the cell surface and the fusion of Fc on sialidase induced antibody-dependent cell-mediated cytotoxicity (ADCC) pathway activation in T cells using an ADCC reporter assay.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Sialidase expressed from vvDD-Sial-FAP/CD3 efficiently cleaves sialic acids from cells.
  • Human lung adenocarcinoma cells were mock-infected or infected with either vvDD or Sial- FAP/CD3-vvDD at 0.05 pfu/cell.
  • APC allophycocyanin
  • SNA Sambucus Nigra Lectin
  • ADCC reporter assays demonstrate efficacy of wDD-Sial-FAP/CD3
  • A549 human lung adenocarcinoma cells were mock-infected or infected with either wDD or vvDD-Sial-FAP/CD3 at the indicated pfu/cell.
  • effector cells expressing a reporter Jurkat cells stably expressing FcyRIIIa receptor and a nuclear factor of activated T cells (NFAT) response element driving expression of firefly luciferase
  • the NFAT response element-driven luciferase expression acts as an early reporter of ADCC.
  • luciferase substrate was added to wells and luminescence in each well was measured to quantify the luciferase activity in effector cells.
  • test or wDD infected A549 cells did not result in luciferase expression (the lines for mock infection and wDD infection are flat overlapping lines in the graph and cannot be distinguished).
  • the A549 cells infected with vvDD-Sial-FAP/CD3 did induce luciferase expression in the Jurkat reporter effector cells, indicating activation of the ADCC pathway.
  • A549 human lung adenocarcinoma cells were mock-infected or infected with either wDD or vvDD-Sial-FAP/CD3 at 0.05 pfu/cell. About 20 hrs after infection, supernatants were collected and passed through 0.2 mm fdters to generate Conditioned Media. The indicated amounts of Conditioned Media were added to COLO829 (FAP -positive) or A549 (FAP-negative) cells, followed by addition of the T cell receptor (TCR)/CD3 Effector cells, Jurkat cells that express a luciferase reporter driven by Nuclear Factor of Activated T Cells (NFAT) response element.
  • TCR T cell receptor
  • luciferase substrate was added to wells and luminescence in each well was measured to quantify the luciferase activity in effector cells.
  • the conditioned media from cells infected with vvDD-Sial-FAP/CD3 resulted in luciferase expression indicative of ADCC in the FAP -positive, COLO829 cells in a concentration-dependent manner, but resulted in much lower luciferase expression in the FAP-negative A549 cell line, indicating the specific effect of the FAP/CD3 bispecific T cell engager.
  • Conditioned media from mock infected cells or cells infected with wDD did not result in luciferase expression.
  • HCT116 human colon cancer cells mixed with FAP-expressing normal human dermal fibroblasts or FAP -positive HCC1143 human breast cancer cells in the presence of human peripheral blood mononuclear cells.
  • HCT116 cells co-cultured with normal human dermal fibroblasts (NhDF) were mock- infected or infected with wDD or vvDD-Sial-FAP/CD3 at 0.1 pfu/cell.
  • PBMCs peripheral blood mononuclear cells
  • LDH lactate dehydrogenase
  • PBMCs peripheral blood mononuclear cells
  • HCT116 human colon cancer cells co-cultured with normal human dermal fibroblasts (NhDF) or HCC1143 human breast cancer cells were similarly mock-infected or infected with vvDD or vvDD-Sial-FAP/CD3 at 0.1 pfu/cell.
  • PBMCs peripheral blood mononuclear cells
  • infection with vvDD-Sial- FAP/CD3 significantly increased the % of CD69 + cells in the population of CD4 + T cells compared to mock-infected or vvDD infected cells for both the HCT116/NhDF and HCC1143 cells.
  • infection with vvDD-Sial-FAP/CD3 significantly increased the % of CD69 + cells in the population of CD8 + T cells compared to mock-infected or vvDD infected cells for both the HCT116/NhDF and HCC1143 cells.
  • vvDD-Sial- FAP/CD3 induced activation of both CD4 + and CD8 + T cells, as measured by the upregulation of CD69 and CD25 markers and increased granzyme B release, which resulted in enhanced cell killing.
  • wDD-Sial-FAP/CD3 enhanced lymphocyte infiltration in tumor spheroids
  • A549 human lung adenocarcinoma cells co-cultured with cancer-associated fibroblasts (CAFs) were mock-infected or infected with vvDD or vvDD-Sial-FAP/CD3 expressing green or yellow fluorescent protein, respectively, at 0.3 pfu/cell.
  • CAFs cancer-associated fibroblasts
  • the expression of either GFP or YFP was monitor by imaging.
  • Increase in the intensity of the fluorescent proteins indicates the spread of the virus within tumor spheroids.
  • wDD-Sial-FAP/CD3 spread efficiently within the tumor spheroid.
  • PBMCs peripheral blood mononuclear cells
  • SEQ ID NO: 10 A. viscosus nanA sialidase
  • SEQ ID NO: 14 S. mitis nanA_l sialidase
  • SEQ ID NO: 16 S. mitis nanA_3 sialidase
  • SEQ ID NO: 19 S. mitis nanH sialidase
  • SEQ ID NO: 23 A muciniphila sialidase
  • SEQ ID NO: 24 A muciniphila sialidase
  • SEQ ID NO: 26 A viscosus sialidase
  • SEQ ID NO: 28 Construct 1: mIg-K_DAS181 Protein sequence
  • SEQ ID NO: 29 Construct 2: mIg-K_DAS185 Protein sequence METDTLLLWVLLLWVPGSTGDGDHPOATPAPAPDASTELPASMSOAOHLAANTAT DNYRIPAITTAPNGDLLISYDERPKDNGNGGSDAPNPNHIVQRRSTDGGKTWSAPTYI HQGTETGKKVGYSDPSYVVDHQTGTIFNFHVKSYDQGWGGSRGGTDPENRGIIQAE
  • SEQ ID NO: 30 Construct 3: mlg-K Neu2-AR Protein sequence
  • NAVGQDTQEVIVVPHSLPFKVVVISAILALVVLTIISLIILIMLWQKKPR TM Domain
  • SEQ ID NO: 34 Construct 1: mIg-K_DAS181 Nucleotide sequence
  • SEQ ID NO: 37 Construct 4: DAS 181(-AR)_TM Nucleotide sequence atggagacagacacactcctgctatgggtactgctgctctgggttccaggttccactggtgacTATCCA TATGATGTTCCAGATTATGCTGGGGCCACGCCGGCCAGATCTCCCGGGATGGGCG ACCACCCACAGGCAACACCAGCACCTGCCCCAGATGCCTCCACCGAGCTGCCAG CAAGCATGTCCCAGGCACAGCACCTGGCAGCAAATACCGCAACAGACAACTACA GAATCCCCGCCATCACCACAGCCCCAAATGGCGATCTGCTGATCAGCTATGACG
  • SEQ ID NO: 38 Construct 5: DAS185(-AR)_TM Nucleotide sequence atggagacagacacactcctgctatgggtactgctgctctgggttccaggttccactggtgacTATCCATATGATGTTCC AGATTATGCTGGGGCCACGCCGGCCAGATCTCCCGGGATGGGCGACCACCCACA GGCAACACCAGCACCTGCCCCAGATGCCTCCACCGAGCTGCCAGCAAGCATGTC CCAGGCACAGCACCTGGCAGCAAATACCGCAACAGACAACTACAGAATCCCCGC CATCACCACAGCCCCAAATGGCGATCTGCTGATCAGCTATGACGAGCGCCCCAA GGATAACGGAAATGGAGGCTCCGACGCACCAAACCCTAATCACATCGTGCAGCG GAGATCTACCGATGGCGGCAAGACATGGAGCCCCTACCTACATCCACCAGGG CACCGAGACAGGCAAGAAGGTCGGCTACTCTGACCCAAGCTATGTGG
  • SEQ ID NO: 53 Salmonella typhimurium sialidase
  • TK-left (SEQ ID NO: 59) agtgataatcggccccatgttttcaggtaaaagtacagaataattagacgagttagacgttatcaaatagctcaatataaatgcgtgact ataaaatattctaacgataatagatacggaacgggactatggacgcatgataagaataatttttgaagcattggaagcaactaaactatgt gatgtcttggaatcaattacagatttctccgtgataggtatcgatgaaggaggacagttctttccagacattgttgaatt
  • F17R (SEQ ID NO: 61) gaatttcattttgttttttttctatgctataa
  • LoxP (SEQ ID NO: 62) ataacttcgtataatgtatgctatacgaagttat GFP: (SEQ ID NO: 63)
  • TK-right (SEQ ID NO: 64) aattctgtgagcgtatggcaaacgaaggaaaatagttatagtagccgcactcgatgggacatttcaacgtaaaccgtttaataatatttt gaatcttattccattatctgaaatggtggtaaactaactgctgtgtatgtatgaaatgcttttaaggaggcttccttttctaacgattgggtgag gaaaccgagatagaaataaaa
  • SEQ ID NO: 65 Sequence of a portion of a vaccinia virus construct for expressing a sialidase (DAS 181). atgaacggcggacatattcagttgataatcggccccatgttttcaggtaaaagtacagaattaattagacgagttagacgttatcaaatag ctcaatataaatgcgtgactataaaatattctaacgatatagatacggaacgggactatggacgcatgataagaataatttttgaagcatt ggaagcaactaaactatgtgatgtcttggaatcaattacagatttctccgtgataggtatcgatgaaggacagttctttccagacattgttg acattgttg aattagatcgataaaaaattaattaattacccgggtaccacattt
  • SEQ ID NO: 102 exemplary signal peptide sequence
  • SEQ ID NO: 106 Sialidase-IgG Fc-transmembrane domain (with signal peptide sequence) MYRMQLLSCIALSLALVTNSMGDHPQATPAPAPDASTELPASMSQAQHLAANTATD NYRIPAITTAPNGDLLISYDERPKDNGNGGSDAPNPNHIVQRRSTDGGKTWSAPTYIH QGTETGKKVGYSDPSYVVDHQTGTIFNFHVKSYDQGWGGSRGGTDPENRGIIQAEV STSTDNGWTWTHRTITADITKDKPWTARFAASGQGIQIQHGPHAGRLVQQYTIRTAG GAVQAVSVYSDDHGKTWQAGTPIGTGMDENKVVELSDGSLMLNSRASDGSGFRKV AHSTDGGQTWSEPVSDKNLPDSVDNAQIIRAFPNAAPDDPRAKVLLLSHSPNPRPWS RDRGTISMSCDDGASWTTSKVFHEPFVGYTTIAVQSDGSIGLLSEDAHNGADY
  • SEQ ID NO: 108 Sial-FAP/CD3 nucleic acid construct
  • SEQ ID NO: 110 nucleotide sequence encoding Sial-IgG Fc-transmembrane

Abstract

La présente invention concerne des procédés et des compositions pour traiter des cancers (tels que des tumeurs solides) à l'aide d'un virus oncolytique recombinant codant pour une sialidase et un agent de ciblage de cellule immunitaire multispécifique. Dans certains modes de réalisation, l'agent de ciblage de cellule immunitaire multispécifique reconnaît une protéine d'activation des fibroblastes (TAP) et CD3.
PCT/US2021/073203 2020-12-30 2021-12-30 Virus oncolytique codant pour la sialidase et agent de ciblage de cellule immunitaire multispécifique WO2022147480A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063132420P 2020-12-30 2020-12-30
US63/132,420 2020-12-30

Publications (1)

Publication Number Publication Date
WO2022147480A1 true WO2022147480A1 (fr) 2022-07-07

Family

ID=80050568

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2021/073203 WO2022147480A1 (fr) 2020-12-30 2021-12-30 Virus oncolytique codant pour la sialidase et agent de ciblage de cellule immunitaire multispécifique
PCT/US2021/073204 WO2022147481A1 (fr) 2020-12-30 2021-12-30 Polythérapie d'un virus oncolytique délivrant un antigène étranger et cellule immunitaire modifiée exprimant un récepteur chimérique ciblant l'antigène étranger

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2021/073204 WO2022147481A1 (fr) 2020-12-30 2021-12-30 Polythérapie d'un virus oncolytique délivrant un antigène étranger et cellule immunitaire modifiée exprimant un récepteur chimérique ciblant l'antigène étranger

Country Status (2)

Country Link
TW (1) TW202241479A (fr)
WO (2) WO2022147480A1 (fr)

Citations (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5116742A (en) 1986-12-03 1992-05-26 University Patents, Inc. RNA ribozyme restriction endoribonucleases and methods
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US5716613A (en) 1988-03-21 1998-02-10 Chiron Viagene, Inc. Recombinant retroviruses
WO1998010088A1 (fr) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Procede inductible de production de virus adeno-associes recombines au moyen de la polymerase t7
US5830755A (en) 1995-03-27 1998-11-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services T-cell receptors and their use in therapeutic and diagnostic methods
WO1999007409A1 (fr) 1997-08-04 1999-02-18 Societe De Conseils De Recherches Et D'applications Scientifiques (S.C.R.A.S.) Produit comprenant au moins un arn double brin en association avec au moins un agent anti-viral
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
WO2000001846A2 (fr) 1998-07-03 2000-01-13 Devgen N.V. Caracterisation d'une fonction de gene par inhibition d'arn double brin
WO2000044895A1 (fr) 1999-01-30 2000-08-03 Roland Kreutzer Methode et medicament destines a inhiber l'expression d'un gene donne
WO2000044914A1 (fr) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition et methode destinees a l'attenuation in vivo et in vitro de l'expression genique utilisant de l'arn double brin
WO2001029058A1 (fr) 1999-10-15 2001-04-26 University Of Massachusetts Genes de voies d'interference d'arn en tant qu'outils d'interference genetique ciblee
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
US6428968B1 (en) 1999-03-15 2002-08-06 The Trustees Of The University Of Pennsylvania Combined therapy with a chemotherapeutic agent and an oncolytic virus for killing tumor cells in a subject
WO2002077029A2 (fr) 2000-11-07 2002-10-03 City Of Hope Cellules immunitaires specifiques a cd19 redirigees
US6632670B1 (en) 1995-09-08 2003-10-14 Genzyme Corporation AAV vectors for gene therapy
US6635472B1 (en) 1997-08-15 2003-10-21 Rubicon Laboratory, Inc. Retrovirus and viral vectors
US6639139B2 (en) 2001-01-25 2003-10-28 Richard Muller Color-coded melody text and method of teaching
US6653103B2 (en) 2001-03-30 2003-11-25 Wisconsin Alumni Research Foundation Inhibition of nucleocytoplasmic transport by vesicular stomatitis virus M protein-like polypeptides
US20040009604A1 (en) 2002-03-27 2004-01-15 Xiaoliu Zhang Potent oncolytic herpes simplex virus for cancer therapy
EP1385466A2 (fr) 2001-05-11 2004-02-04 Wellstat Biologics Corporation Traitement par virus oncolytique
US6689871B1 (en) 1999-02-26 2004-02-10 Amersham Health As Process for production diphtheria toxin
US6723316B2 (en) 1999-12-22 2004-04-20 Onyx Pharmaceuticals, Inc. Herpes simplex virus-1 Glycoprotein C mutants for treating unwanted hyperproliferative cell growth
EP1520175A2 (fr) 2002-06-28 2005-04-06 Oncolytics Biotech, Inc. Virus oncolytiques en tant qu'agents de phenotypage destines a des neoplasmes
US6897045B2 (en) 1998-05-20 2005-05-24 University Of Iowa Research Foundation Adeno-associated virus vectors
US20050220818A1 (en) 2002-06-21 2005-10-06 Wellstat Biologics Corporation Administration of therapeutic viruses
US20050260601A1 (en) 2002-09-09 2005-11-24 Whitt Michael A Recombinant mutants of rhabdovirus and methods of use thereof
EP1606411A1 (fr) 2003-03-27 2005-12-21 Ottawa Health Research Institute Virus mutant de la stomatite vesiculaire et son utilisation
US7001765B2 (en) 1996-03-06 2006-02-21 Medigene Ag Adeno-associated virus vector for boosting immunogenicity of cells
US20060039894A1 (en) 2004-06-24 2006-02-23 Ian Mohr Avirulent oncolytic herpes simplex virus strains engineered to counter the innate host response
US7033826B2 (en) 1995-06-23 2006-04-25 Aventis Pharma S.A. Recombinant adenoviruses, use thereof for preparing AAVS, complementary cell line, and pharmaceutical compositions containing said adenoviruses
US7153510B1 (en) 1995-05-04 2006-12-26 Yale University Recombinant vesiculoviruses and their uses
US7172893B2 (en) 1998-11-10 2007-02-06 University Of North Carolina At Chapel Hill Virus vectors and methods of making and administering the same
US20070098743A1 (en) 2003-03-27 2007-05-03 Bell John C Mutant vesicular stomatitis viruses and uses thereof
US20070110720A1 (en) 2003-11-17 2007-05-17 Brown Susanne M Vectors, mutant viruses and methods for generating mutant viruses
US7238526B2 (en) 1997-09-19 2007-07-03 The Trustees Of The University Of Pennsylvania Methods and cell line useful for production of recombinant adeno-associated viruses
US7241447B1 (en) 1999-10-07 2007-07-10 University Of Iowa Research Foundation Adeno-associated virus vectors and uses thereof
US20090010889A1 (en) 2003-11-17 2009-01-08 Crusade Laboratories Limited Treatment Using Herpes Simplex Virus
US7537924B2 (en) 2000-01-21 2009-05-26 Biovex Limited Virus strains
US7550296B2 (en) 2004-12-01 2009-06-23 Bayer Schering Pharma Ag Generation of replication competent viruses for therapeutic use
US7556928B2 (en) 2001-05-26 2009-07-07 One Cell Systems, Inc. Method of screening a population of cells
US20090215147A1 (en) 2005-06-23 2009-08-27 Baylor College Of Medicine Use of Mutant Herpes Simplex Virus-2 for Cancer Therapy
US20090274728A1 (en) 2003-11-17 2009-11-05 Crusade Laboratories Limited Mutant Viruses
US20090285860A1 (en) 2008-04-04 2009-11-19 The General Hospital Corporation Cancer immunotherapy
US7645448B2 (en) 2002-11-22 2010-01-12 Nexbio, Inc. Class of therapeutic protein based molecules
US7662627B2 (en) 1994-06-06 2010-02-16 Nationwide Children's Hospital, Inc. Adeno-associated virus materials and methods
US20100092515A1 (en) 2007-02-16 2010-04-15 Crusade Laboratories Limited Herpes simplex viruses and methods of viral replication
US20100113567A1 (en) 2001-07-11 2010-05-06 University Of Miami Recombinant VSV For The Treatment of Tumor Cells
US20100172877A1 (en) 2009-01-08 2010-07-08 Yale University Compositions and methods of use of an oncolytic vesicular stomatitis virus
US20100178684A1 (en) 2006-12-21 2010-07-15 Woo Savio L C Transgenic oncolytic viruses and uses thereof
US7811814B2 (en) 2004-05-18 2010-10-12 Children's Memorial Hospital Tetracycline-regulated adeno-associated viral (AAV) vectors for gene delivery to the nervous system
US7906111B2 (en) 2003-09-30 2011-03-15 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clades, sequences, vectors containing same, and uses therefor
US7927585B2 (en) 2002-04-30 2011-04-19 University Of Florida Research Foundation, Inc. Production of recombinant AAV virions
US7943374B2 (en) 2005-08-21 2011-05-17 Markus Hildinger Super-size adeno-associated viral vector harboring a recombinant genome larger than 5.7 kb
US7968340B2 (en) 1999-07-15 2011-06-28 Medigene Ag Structural protein of adeno-associated virus with modified antigenicity, its production and its use
US20110177032A1 (en) 2008-05-29 2011-07-21 The General Hospital Corporation D/B/A Massachusetts General Hospital Use on oncolytic herpes viruses for killing cancer stem cells
US8007780B2 (en) 2004-06-03 2011-08-30 Genzyme Corporation AAV vectors for gene delivery to the lung
US20110212530A1 (en) 2005-06-01 2011-09-01 California Institute Of Technology Method of targeted gene delivery using viral vectors
WO2015142675A2 (fr) 2014-03-15 2015-09-24 Novartis Ag Traitement du cancer au moyen d'un récepteur antigénique chimérique
WO2016187349A1 (fr) 2015-05-18 2016-11-24 Tcr2, Inc. Compositions et méthodes de reprogrammation de tcr au moyen de protéines de fusion
US9700602B2 (en) 2012-06-28 2017-07-11 Ansun Biopharma, Inc. Microparticle formulations for delivery to the lower and central respiratory tract and methods of manufacture
US9765342B2 (en) 2012-04-11 2017-09-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
WO2018049261A1 (fr) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Virus oncolytique exprimant des modulateurs du point de contrôle immunitaire
WO2018098365A2 (fr) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions et méthodes de reprogrammation de tcr au moyen de protéines de fusion
US10351828B2 (en) 2013-03-15 2019-07-16 Ansun Biopharma, Inc. Method of protein purification from E. coli
WO2020018996A2 (fr) * 2018-07-20 2020-01-23 Ansun Biopharma, Inc. Administration de sialidase à des cellules cancéreuses, des cellules immunitaires et au microenvironnement tumoral
WO2020086423A1 (fr) 2018-10-22 2020-04-30 Icell Kealex Therapeutics Virus de la vaccine mutants et leur utilisation
WO2020097269A1 (fr) 2018-11-06 2020-05-14 Calidi Biotherapeutics, Inc. Systèmes améliorés pour thérapie virale oncolytique à médiation cellulaire

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5994619A (en) 1996-04-01 1999-11-30 University Of Massachusetts, A Public Institution Of Higher Education Of The Commonwealth Of Massachusetts, As Represented By Its Amherst Campus Production of chimeric bovine or porcine animals using cultured inner cell mass cells
AR039067A1 (es) 2001-11-09 2005-02-09 Pfizer Prod Inc Anticuerpos para cd40
AU2008233051B2 (en) 2007-03-30 2014-04-10 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred T lymphocytes
KR20220151611A (ko) * 2020-01-21 2022-11-15 안선 바이오파르마, 아이엔씨. 암 세포, 면역 세포 및 종양 미세환경으로의 시알리다제의 전달

Patent Citations (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US5093246A (en) 1986-12-03 1992-03-03 University Patents, Inc. Rna ribozyme polymerases, dephosphorylases, restriction endoribo-nucleases and methods
US5116742A (en) 1986-12-03 1992-05-26 University Patents, Inc. RNA ribozyme restriction endoribonucleases and methods
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5716613A (en) 1988-03-21 1998-02-10 Chiron Viagene, Inc. Recombinant retroviruses
US5716826A (en) 1988-03-21 1998-02-10 Chiron Viagene, Inc. Recombinant retroviruses
US5851529A (en) 1988-03-21 1998-12-22 Guber; Harry E. Recombinant retroviruses
US7662627B2 (en) 1994-06-06 2010-02-16 Nationwide Children's Hospital, Inc. Adeno-associated virus materials and methods
US5830755A (en) 1995-03-27 1998-11-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services T-cell receptors and their use in therapeutic and diagnostic methods
US7153510B1 (en) 1995-05-04 2006-12-26 Yale University Recombinant vesiculoviruses and their uses
US7033826B2 (en) 1995-06-23 2006-04-25 Aventis Pharma S.A. Recombinant adenoviruses, use thereof for preparing AAVS, complementary cell line, and pharmaceutical compositions containing said adenoviruses
US6632670B1 (en) 1995-09-08 2003-10-14 Genzyme Corporation AAV vectors for gene therapy
US7001765B2 (en) 1996-03-06 2006-02-21 Medigene Ag Adeno-associated virus vector for boosting immunogenicity of cells
WO1998010088A1 (fr) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Procede inductible de production de virus adeno-associes recombines au moyen de la polymerase t7
WO1999007409A1 (fr) 1997-08-04 1999-02-18 Societe De Conseils De Recherches Et D'applications Scientifiques (S.C.R.A.S.) Produit comprenant au moins un arn double brin en association avec au moins un agent anti-viral
US6635472B1 (en) 1997-08-15 2003-10-21 Rubicon Laboratory, Inc. Retrovirus and viral vectors
US7238526B2 (en) 1997-09-19 2007-07-03 The Trustees Of The University Of Pennsylvania Methods and cell line useful for production of recombinant adeno-associated viruses
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US6897045B2 (en) 1998-05-20 2005-05-24 University Of Iowa Research Foundation Adeno-associated virus vectors
WO2000001846A2 (fr) 1998-07-03 2000-01-13 Devgen N.V. Caracterisation d'une fonction de gene par inhibition d'arn double brin
US7172893B2 (en) 1998-11-10 2007-02-06 University Of North Carolina At Chapel Hill Virus vectors and methods of making and administering the same
WO2000044914A1 (fr) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition et methode destinees a l'attenuation in vivo et in vitro de l'expression genique utilisant de l'arn double brin
WO2000044895A1 (fr) 1999-01-30 2000-08-03 Roland Kreutzer Methode et medicament destines a inhiber l'expression d'un gene donne
US6689871B1 (en) 1999-02-26 2004-02-10 Amersham Health As Process for production diphtheria toxin
US6428968B1 (en) 1999-03-15 2002-08-06 The Trustees Of The University Of Pennsylvania Combined therapy with a chemotherapeutic agent and an oncolytic virus for killing tumor cells in a subject
US7968340B2 (en) 1999-07-15 2011-06-28 Medigene Ag Structural protein of adeno-associated virus with modified antigenicity, its production and its use
US7241447B1 (en) 1999-10-07 2007-07-10 University Of Iowa Research Foundation Adeno-associated virus vectors and uses thereof
WO2001029058A1 (fr) 1999-10-15 2001-04-26 University Of Massachusetts Genes de voies d'interference d'arn en tant qu'outils d'interference genetique ciblee
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
US6723316B2 (en) 1999-12-22 2004-04-20 Onyx Pharmaceuticals, Inc. Herpes simplex virus-1 Glycoprotein C mutants for treating unwanted hyperproliferative cell growth
US7537924B2 (en) 2000-01-21 2009-05-26 Biovex Limited Virus strains
WO2002077029A2 (fr) 2000-11-07 2002-10-03 City Of Hope Cellules immunitaires specifiques a cd19 redirigees
US6639139B2 (en) 2001-01-25 2003-10-28 Richard Muller Color-coded melody text and method of teaching
US6653103B2 (en) 2001-03-30 2003-11-25 Wisconsin Alumni Research Foundation Inhibition of nucleocytoplasmic transport by vesicular stomatitis virus M protein-like polypeptides
EP1385466A2 (fr) 2001-05-11 2004-02-04 Wellstat Biologics Corporation Traitement par virus oncolytique
US7556928B2 (en) 2001-05-26 2009-07-07 One Cell Systems, Inc. Method of screening a population of cells
US20100113567A1 (en) 2001-07-11 2010-05-06 University Of Miami Recombinant VSV For The Treatment of Tumor Cells
US20040009604A1 (en) 2002-03-27 2004-01-15 Xiaoliu Zhang Potent oncolytic herpes simplex virus for cancer therapy
US7927585B2 (en) 2002-04-30 2011-04-19 University Of Florida Research Foundation, Inc. Production of recombinant AAV virions
US20050220818A1 (en) 2002-06-21 2005-10-06 Wellstat Biologics Corporation Administration of therapeutic viruses
EP1520175A2 (fr) 2002-06-28 2005-04-06 Oncolytics Biotech, Inc. Virus oncolytiques en tant qu'agents de phenotypage destines a des neoplasmes
US20050260601A1 (en) 2002-09-09 2005-11-24 Whitt Michael A Recombinant mutants of rhabdovirus and methods of use thereof
US7645448B2 (en) 2002-11-22 2010-01-12 Nexbio, Inc. Class of therapeutic protein based molecules
US20070098743A1 (en) 2003-03-27 2007-05-03 Bell John C Mutant vesicular stomatitis viruses and uses thereof
EP1606411A1 (fr) 2003-03-27 2005-12-21 Ottawa Health Research Institute Virus mutant de la stomatite vesiculaire et son utilisation
US7731974B2 (en) 2003-03-27 2010-06-08 Ottawa Hospital Research Institute Mutant vesicular stomatitis viruses and uses thereof
US7906111B2 (en) 2003-09-30 2011-03-15 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clades, sequences, vectors containing same, and uses therefor
US20090274728A1 (en) 2003-11-17 2009-11-05 Crusade Laboratories Limited Mutant Viruses
US20110158948A1 (en) 2003-11-17 2011-06-30 Crusade Laboratories Limited Treatment Using Herpes Simplex Virus
US20090010889A1 (en) 2003-11-17 2009-01-08 Crusade Laboratories Limited Treatment Using Herpes Simplex Virus
US20070110720A1 (en) 2003-11-17 2007-05-17 Brown Susanne M Vectors, mutant viruses and methods for generating mutant viruses
US7897146B2 (en) 2003-11-17 2011-03-01 Crusade Laboratories Limited Treatment using herpes simplex virus
US7811814B2 (en) 2004-05-18 2010-10-12 Children's Memorial Hospital Tetracycline-regulated adeno-associated viral (AAV) vectors for gene delivery to the nervous system
US8007780B2 (en) 2004-06-03 2011-08-30 Genzyme Corporation AAV vectors for gene delivery to the lung
US20060039894A1 (en) 2004-06-24 2006-02-23 Ian Mohr Avirulent oncolytic herpes simplex virus strains engineered to counter the innate host response
US7731952B2 (en) 2004-06-24 2010-06-08 New York University Avirulent oncolytic herpes simplex virus strains engineered to counter the innate host response
US7550296B2 (en) 2004-12-01 2009-06-23 Bayer Schering Pharma Ag Generation of replication competent viruses for therapeutic use
US20110212530A1 (en) 2005-06-01 2011-09-01 California Institute Of Technology Method of targeted gene delivery using viral vectors
US20090215147A1 (en) 2005-06-23 2009-08-27 Baylor College Of Medicine Use of Mutant Herpes Simplex Virus-2 for Cancer Therapy
US7943374B2 (en) 2005-08-21 2011-05-17 Markus Hildinger Super-size adeno-associated viral vector harboring a recombinant genome larger than 5.7 kb
US20100178684A1 (en) 2006-12-21 2010-07-15 Woo Savio L C Transgenic oncolytic viruses and uses thereof
US20100092515A1 (en) 2007-02-16 2010-04-15 Crusade Laboratories Limited Herpes simplex viruses and methods of viral replication
US20090285860A1 (en) 2008-04-04 2009-11-19 The General Hospital Corporation Cancer immunotherapy
US20110177032A1 (en) 2008-05-29 2011-07-21 The General Hospital Corporation D/B/A Massachusetts General Hospital Use on oncolytic herpes viruses for killing cancer stem cells
US20100172877A1 (en) 2009-01-08 2010-07-08 Yale University Compositions and methods of use of an oncolytic vesicular stomatitis virus
US9765342B2 (en) 2012-04-11 2017-09-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US9700602B2 (en) 2012-06-28 2017-07-11 Ansun Biopharma, Inc. Microparticle formulations for delivery to the lower and central respiratory tract and methods of manufacture
US10351828B2 (en) 2013-03-15 2019-07-16 Ansun Biopharma, Inc. Method of protein purification from E. coli
WO2015142675A2 (fr) 2014-03-15 2015-09-24 Novartis Ag Traitement du cancer au moyen d'un récepteur antigénique chimérique
WO2016187349A1 (fr) 2015-05-18 2016-11-24 Tcr2, Inc. Compositions et méthodes de reprogrammation de tcr au moyen de protéines de fusion
WO2018049261A1 (fr) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Virus oncolytique exprimant des modulateurs du point de contrôle immunitaire
WO2018098365A2 (fr) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions et méthodes de reprogrammation de tcr au moyen de protéines de fusion
WO2020018996A2 (fr) * 2018-07-20 2020-01-23 Ansun Biopharma, Inc. Administration de sialidase à des cellules cancéreuses, des cellules immunitaires et au microenvironnement tumoral
WO2020086423A1 (fr) 2018-10-22 2020-04-30 Icell Kealex Therapeutics Virus de la vaccine mutants et leur utilisation
WO2020097269A1 (fr) 2018-11-06 2020-05-14 Calidi Biotherapeutics, Inc. Systèmes améliorés pour thérapie virale oncolytique à médiation cellulaire

Non-Patent Citations (40)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. NM080741
"GenBank", Database accession no. WP_003789074
"Remington's Pharmaceutical Sciences", 1980
"Sialic Acids Chemistry, Metabolism and Function", 1982, SPRINGER-VERLAG, pages: 233
ADRA ET AL., GENE, vol. 60, 1987, pages 65 - 74
ANONYMOUS: "Frontiers | Delivery and Biosafety of Oncolytic Virotherapy | Oncology", 11 February 2020 (2020-02-11), XP055908419, Retrieved from the Internet <URL:https://www.frontiersin.org/articles/10.3389/fonc.2020.00475/full> [retrieved on 20220404] *
BARTELSZOSTAK, SCIENCE, vol. 261, 1993, pages 1411 - 1418
BELSER J A ET AL: "DAS181, a novel sialidase fusion protein, protects mice from lethal avian influenza h5N1 virus infection", JOURNAL OF INFECTIOUS DISEASES, UNIVERSITY OF CHICAGO PRESS, US, vol. 196, no. 10, 15 November 2007 (2007-11-15), pages 1493 - 1499, XP002518024, ISSN: 0022-1899, DOI: 10.1086/522609 *
BRANDLERTANGY, CIMID, vol. 31, 2008, pages 271
CATTANEO, NAT. REV. MICROBIOL., vol. 6, no. 7, 2008, pages 529 - 540
CHAKRABARTI ET AL., BIOTECHNIQUES, vol. 23, 1997, pages 1094 - 7
CHAKRABARTI ET AL., BIOTECHNIQUES, vol. 23, no. 6, 1997, pages 1094 - 1097
DAVISON ET AL., NUCLEIC ACIDS RES., vol. 18, 1990, pages 4285 - 4286
DAVISONMOSS, J MOL BIOL, vol. 210, 1989, pages 749 - 769
DORER ET AL., ADV. DRUG DELIV. REV., vol. 61, no. 7- 8, 2009, pages 554 - 571
EDGAR, R.C., BMC BIOINFORMATICS, vol. 5, no. 1, 2004, pages 113
EDGAR, R.C., NUCLEIC ACIDS RESEARCH, vol. 32, no. 5, 2004, pages 1792 - 1797
ELBASHIR ET AL., NATURE, vol. 411, 2001, pages 494 - 498
ERBS ET AL., CANCER GENE THER, vol. 15, no. 1, 2008, pages 18 - 28
GUO: "bi and tri -specific", 10 July 2020 (2020-07-10), XP055908427, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7400484/pdf/biomedicines-08-00204.pdf> [retrieved on 20220404] *
HAMMOND ET AL., J. VIROL METHODS, vol. 66, 1997, pages 135 - 8
HASELHOFFAND GERLACH, NATURE, vol. 334, 1988, pages 585 - 591
HAYES, C.: "Cellular immunotherapies for cancer", IR J MED SCI, 2020
HUTVAGNERZAMORE, SCIENCE, vol. 297, 2002, pages 1831 - 2237
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
KELLY ET AL., MOL. THER., vol. 17, no. 3, 2009, pages 409 - 416
KESSELS ET AL.: "Immunotherapy through TCR gene transfer", NAT. IMMUNOL., vol. 2, 2001, pages 957 - 961
KUMARBOYLE, VIROLOGY, vol. 179, 1990, pages 151 - 8
MATVEEVA OLGA V ET AL: "Oncolysis by paramyxoviruses: multiple mechanisms contribute to therapeutic efficiency", MOLECULAR THERAPY - ONCOLYTICS, vol. 2, 1 January 2015 (2015-01-01), pages 15011, XP055908476, ISSN: 2372-7705, DOI: 10.1038/mto.2015.11 *
MCCART JA ET AL.: "Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes", CANCER RES, vol. 61, 2001, pages 8751 - 8757
MCMANUS ET AL., RNA, vol. 8, 2002, pages 842 - 850
MEYER, F AKING, MGELMAN, R A, BIOCHIMICA ET BIOPHYSICA ACTA, vol. 392, 1975, pages 223 - 232
MONTI ET AL., NEUROCHEM RES, vol. 27, 2002, pages 646 - 663
MONTI, EPRETIROSSI, EBALLABIO, ABORSANI G, GENOMICS, vol. 57, 1999, pages 137 - 143
MORDENTI, J.CHAPPELL, W. ET AL.: "Toxicokinetics and New Drug Development", 1989, PERGAMON PRESS, article "The Use of Interspecies Scaling in Toxicokinetics", pages: 42 - 46
NAIK ET AL., EXPERT OPIN. BIOL. THER., vol. 9, no. 9, 2009, pages 1163 - 1176
NICHOLSON IC ET AL., MOL IMMUNOL, 1997
PATEL ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 9431 - 9435
REINHART ET AL., GENE & DEV, vol. 16, 2002, pages 1616 - 1626
ZAMORE ET AL., CELL, vol. 101, 2000, pages 25 - 33

Also Published As

Publication number Publication date
TW202241479A (zh) 2022-11-01
WO2022147481A1 (fr) 2022-07-07

Similar Documents

Publication Publication Date Title
EP3503918B1 (fr) Adénovirus armé avec des éléments bispécifiques de liaison aux cellules t (bite)
US10799536B2 (en) Method of treating multiple myeloma using natural killer cells expressing a chimeric antigen receptor for CD38
JP7280244B2 (ja) 二重特異性t細胞アクチベーターを搭載したアデノウイルス
US20210137839A1 (en) Compositions and methods for membrane protein delivery
US20230149487A1 (en) Delivery of sialidase to cancer cells, immune cells and the tumor microenvironment
US20230210902A1 (en) Sars-cov-2-specific t cells
WO2020019983A1 (fr) Cellule génétiquement modifiée utilisée pour traiter une tumeur
WO2022170219A1 (fr) Traitement adjuvant du cancer
EP4061406A1 (fr) Virus mva recombinants pour l&#39;administration intratumorale et/ou intraveineuse pour le traitement du cancer
WO2022147480A1 (fr) Virus oncolytique codant pour la sialidase et agent de ciblage de cellule immunitaire multispécifique
CN114828861A (zh) 表达多特异性免疫细胞接合器的溶瘤病毒
US20230022654A1 (en) Ror1 targeting chimeric antigen receptor
Kirchhammer Tumor-targeted immunotherapy using an engineered adenoviral vector platform
WO2022148736A1 (fr) Vectorisation de l&#39;anticorps engageant les cellules t muc1
WO2023046777A1 (fr) Nouvelles combinaisons d&#39;anticorps et utilisations associées
EP4065139A1 (fr) Cellules immunitaires pour l&#39;administration de sialidase à des cellules cancéreuses, à des cellules immunitaires et au microenvironnement tumoral
NZ791667A (en) Adenovirus armed with bispecific T-cell activator

Legal Events

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

Ref document number: 21848520

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC