WO2024023740A1 - Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors - Google Patents
Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors Download PDFInfo
- Publication number
- WO2024023740A1 WO2024023740A1 PCT/IB2023/057598 IB2023057598W WO2024023740A1 WO 2024023740 A1 WO2024023740 A1 WO 2024023740A1 IB 2023057598 W IB2023057598 W IB 2023057598W WO 2024023740 A1 WO2024023740 A1 WO 2024023740A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- inhibitor
- acid sequence
- amino acid
- therapeutic combination
- seq
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/768—Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/208—IL-12
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/39541—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/39558—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/5434—IL-12
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70521—CD28, CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70532—B7 molecules, e.g. CD80, CD86
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2818—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2827—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2866—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2881—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD71
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/24011—Poxviridae
- C12N2710/24111—Orthopoxvirus, e.g. vaccinia virus, variola
- C12N2710/24121—Viruses as such, e.g. new isolates, mutants or their genomic sequences
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/24011—Poxviridae
- C12N2710/24111—Orthopoxvirus, e.g. vaccinia virus, variola
- C12N2710/24132—Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/24011—Poxviridae
- C12N2710/24111—Orthopoxvirus, e.g. vaccinia virus, variola
- C12N2710/24141—Use of virus, viral particle or viral elements as a vector
- C12N2710/24143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/24011—Poxviridae
- C12N2710/24111—Orthopoxvirus, e.g. vaccinia virus, variola
- C12N2710/24161—Methods of inactivation or attenuation
- C12N2710/24162—Methods of inactivation or attenuation by genetic engineering
Definitions
- the present disclosure relates to a combination comprising (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12) and (ii) an inhibitor of PD-1/PD-L1.
- IL-12 interleukin- 12
- PD-1/PD-L1 an inhibitor of PD-1/PD-L1.
- Recombinant viruses including e.g., recombinant poxviruses, represent an emerging therapeutic platform for the treatment of cancers because of their advantages over conventional therapeutic modalities, such as chemotherapy.
- recombinant viruses can selectivity replicate in cancer cells, while sparing normal cells and tissues, thus limiting off-target cell killing and toxicities, and thereby offer levels of potency and specificity that are potentially far higher than conventional cancer treatments.
- Recombinant viruses can be engineered to express therapeutic transgenes in cells, for example, transgenes important in cancer biology pathways. Cancer cells are ideal hosts for many viruses because they can have the anti-viral interferon pathway inactivated or can have mutated tumor suppressor genes that enable viral replication to proceed unhindered.
- Interleukin- 12 has been considered as a potential candidate for anti-cancer therapy and has been introduced into viral vectors such as adenoviral vectors for evaluation.
- IL-12 is a cytokine with immune-modulating and anti-angiogenic functions.
- IL- 12 acts as a key regulator of cell-mediated immune responses through the induction of T helper 1 differentiation, and it induces cellular immunity by promoting interferongamma (IFN-y) production, proliferation, and cytolytic activity of natural killer and T cells.
- IFN-y interferongamma
- the multi-functionality of IL-12 has led to investigation of this cytokine as an anticancer agent.
- very modest antitumor effects of IL-12 and unacceptable adverse events in early clinical trials diminished hopes of the successful use of this cytokine.
- Immune checkpoint inhibitors such as PD-1 and PD-L1 targeting molecules have demonstrated some success as cancer therapeutics. However, not all cancers respond to checkpoint inhibitors and some cancers develop resistance to checkpoint inhibitors.
- the present disclosure provides a therapeutic combination comprising (a) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (b) a programmed death protein 1 (PD-1) inhibitor or a programmed cell death ligand 1 (PD-L1) inhibitor.
- the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late or intermediate promoter.
- the poxvirus belongs to the Orthopoxvirus genus.
- the poxvirus belonging to the Orthopoxvirus genus is an oncolytic vaccinia virus.
- the oncolytic vaccinia virus is selected from the group consisting of Copenhagen (Cop), Western Reserve (WR), Elstree, Wyeth, Lister, Tian Tan and LIVP virus strains.
- the genome comprises at least 150 kb, at least about 175 kb, at least about 180 kb, at least about 185 kb, at least about 190 kb, at least about 192 kb, or at least about 194 kb.
- the poxvirus is attenuated. In some aspects, the poxvirus is not NYVAC.
- the late promoter is selected from pAlOL, pAHR, pA13L, pA14L, pA26L, pG7L, and pF17R. In some aspects, the late promoter is selected from pA14L, pA26L, and pF17R. In some aspects, the late promoter is pA14L. In some aspects, the late promoter is pF17R.
- the late promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 11, 13, 22, or 23. In some aspects, the late promoter comprises the nucleotide sequence of SEQ ID NO: 11, 13, 22, or 23.
- the intermediate promoter is selected from pHL, pA12L, pA19L, pA42R, pD13L, pA3L, or pA27L.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, at least 99% identical, or 100% identical to the nucleotide sequence of any one of SEQ ID NO: 25-31.
- the IL-12 is human IL-12.
- the IL-12 is a fusion protein comprising an IL-12 p40 subunit and an IL-12 p35 subunit.
- the IL-12 p40 subunit is N-terminal to the IL-12 p35 subunit.
- the IL-12 p40 subunit comprises the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 17.
- the IL-12 p35 subunit comprises the amino acid sequence of SEQ ID NO: 19 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 19.
- the IL-12 p40 subunit and the IL-12 p35 subunit are fused in a single polypeptide via an amino acid linker.
- the amino acid linker is about 5 to about 10 amino acids in length.
- the amino acid linker is 7 amino acids in length.
- the amino acid linker is a glycine-serine linker.
- the amino acid linker comprises the amino acid sequence of SEQ ID NO: 18.
- the IL-12 comprises the amino acid sequence of SEQ ID NO: 20 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 20.
- the IL-12 p40 subunit and the IL-12 p35 subunit are directly fused in a single polypeptide.
- the heterologous nucleic acid sequence encoding the IL-12 comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 21.
- the heterologous nucleic acid sequence encoding the IL-12 comprises the nucleotide sequence of SEQ ID NO: 21.
- the poxvirus is defective for thymidine kinase (TK) activity.
- the poxvirus lacks a functional J2R gene.
- the poxvirus is defective for ribonucleotide reductase (RR) activity.
- the poxvirus lacks a functional I4L gene.
- the poxvirus lacks a functional F4L gene.
- the heterologous nucleic acid sequence encoding IL-12 is inserted within the J2R locus of the poxvirus genome. In some aspects, the insertion renders the J2R gene nonfunctional, optionally wherein the J2R locus is fully deleted by the insertion.
- the heterologous nucleic acid sequence encoding IL- 12 is inserted within the I4L locus of the poxvirus genome. In some aspects, the insertion renders the I4L gene nonfunctional, optionally wherein I4L locus is not fully deleted by the insertion. In some aspects, the heterologous nucleic acid sequence encoding the IL-12 is inserted within the F4L locus of the poxvirus genome. In some aspects, the insertion renders the F4L gene nonfunctional, optionally wherein F4L locus is not fully deleted by the insertion. In some aspects,
- the poxvirus further comprises in its genome one or more therapeutic genes.
- the one or more therapeutic genes is selected from the group consisting of a suicide gene, an immunomodulatory gene, an anti-angiogenic gene, an immune checkpoint blockade gene, an antibody-coding gene, an extracellular matrix degradation or modulation genes, and a combination thereof.
- the therapeutic combination disclosed herein is capable of lysing one or more cancer cells.
- the recombinant poxvirus is capable of expressing at least 50 ng/mL, at least 100 ng/mL, at least 300 ng/mL, at least 500 ng/mL, at least 1.0 pg/mL, at least 2.0 pg/mL, at least 3.0 pg/mL , at least 4.0 pg/mL, at least 5.0 pg/mL, at least 6.0 pg/mL, at least 7.0 pg/mL, at least 8.0 pg/mL, or about 8.3 pg/mL of the IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10-2.
- MOI multiplicity of infection
- the cancer cells are renal cancer, prostate cancer, breast cancer, bladder cancer, colorectal cancer, lung cancer, liver cancer, gastric cancer, bile duct carcinoma, endometrial cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, glioblastoma, multiple myeloma, or malignant glioma cells.
- the cancer cells are A549, HT29, MIA PaCa-2, A375, RPMI7591, Sk-Mel-5, OVCAR3, OVCAR4, NCLH292, NCI-H460, SW 780, TCCSUP, T24, Huh7, Hep3B, Panel, Hup- T3, DAN-G, MDA-MB-435, HCC38, BT20, SW1417, WiDr, HCT-116, SNU5, NCI- N87, Kato III, A CHN, A 498, PC-3, or MM.1R cells.
- the virus is produced in chicken embryo fibroblasts (CEF), HeLa cells, EB66® cells, Vero cells, HEK 293 cells, PerC6 cells, BHK21 cells, or MRC5 cells. In some aspects of the therapeutic combination disclosed herein, the virus is produced in chicken embryo fibroblasts (CEF).
- CEF chicken embryo fibroblasts
- the recombinant poxvirus is capable of upregulating interferon (fFN)-y.
- the PD-1 inhibitor or PD-L1 inhibitor is an anti-PD-1 antibody or antigen-binding fragment thereof or an anti-PD-Ll antibody or antigen-binding fragment thereof.
- the anti- PD-1 antibody or antigen-binding fragment thereof or an anti-PD-Ll antibody or antigenbinding fragment thereof was produced in a Chinese Hamster Ovary (CHO) cell.
- the PD-1 inhibitor or the PD-L1 inhibitor is a small molecule.
- the PD-1 inhibitor or the PD-L1 inhibitor is a PD-1 inhibitor.
- the PD-1 inhibitor or the PD-L1 inhibitor is a PD-L1 inhibitor.
- the PD-1 inhibitor or PD-L1 inhibitor is selected from the group consisting of: Durvalumab, nivolumab, pembrolizumab, lambrolizumab, MEDI-0680, cemiplimab, JS001, BGB-A317, INCSHR1210, TSR-042, Pidilizumab, GLS-010, STI-1110, AGEN2034, MGA012, IB 1308, AMP-224, BMS-936559, atezolizumab, MPDL3280A, RG7446, avelumab, STI- 1014, CX-072, KN035, and CK-301.
- the PD-1 inhibitor or PD-L1 inhibitor is an anti-PD-Ll antibody or antigen-binding fragment thereof comprising a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 32, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 33, a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 34, a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 35, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 36, and a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 37.
- the anti-PD-Ll antibody or antigen-binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 38 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 39.
- the anti-PD-Ll antibody or antigen-binding fragment thereof further comprises an Fc variant, wherein the Fc variant comprises at least one amino acid substitution selected from the group consisting of 234F, 235F, and 331 S, as numbered by the EU index as set forth in Kabat.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab.
- the present disclosure provides a therapeutic combination comprising (a) recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late or intermediate promoter, and (b) Durvalumab.
- the therapeutic combination is for (i) treating cancer in a subject, (ii) inhibiting the growth of a cancer, or (iii) enhancing a tumor-specific immune response.
- the present disclosure provides a therapeutic combination, wherein the therapeutic combination is a kit of parts comprising (a) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), and (b) a programmed death protein 1 (PD-1) inhibitor or a programmed cell death ligand 1 (PD-L1) inhibitor.
- IL- 12 interleukin- 12
- PD-1 programmed death protein 1
- PD-L1 inhibitor programmed cell death ligand 1
- the present disclosure provides a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a programmed death protein 1 (PD-1) inhibitor or a programmed cell death ligand 1 (PD-L1) inhibitor.
- a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a programmed death protein 1 (PD-1) inhibitor or a programmed cell death ligand 1 (PD-L1) inhibitor.
- the present disclosure provides a method of inhibiting the growth of a cancer in a subject, the method comprising administering to the subject an effective amount of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- the present disclosure provides a method of enhancing a tumor-specific immune response in a subject having cancer, the method comprising administering to the subject an effective amount of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- IL- 12 interleukin- 12
- a PD-1 inhibitor or a PD-L1 inhibitor a PD-L1 inhibitor.
- the heterologous nucleic acid sequence encoding IL- 12 is operably linked to a late promoter or intermediate promoter.
- the poxvirus belongs to the Orthopoxvirus genus.
- the poxvirus belonging to the Orthopoxvirus genus is an oncolytic vaccinia virus.
- the oncolytic vaccinia virus is selected from the group consisting of Copenhagen (Cop), Western Reserve (WR), Elstree, Wyeth, Lister, Tian Tan and LIVP virus strains.
- the genome comprises at least 150 kb, at least about 175 kb, at least about 180 kb, at least about 185 kb, at least about 190 kb, at least about 192 kb, or at least about 194 kb.
- the poxvirus is attenuated.
- the poxvirus is not NYVAC.
- the late promoter is selected from pAlOL, pAHR, pA13L, pA14L, pA26L, pG7L, and pF17R. In some aspects, the late promoter is selected from pA14L, pA26L, and pF17R. In some aspects, the late promoter is pA14L. In some aspects, the late promoter is pF17R.
- the late promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 11, 13, 22, or 23. In some aspects, the late promoter comprises the nucleotide sequence of SEQ ID NO: 11, 13, 22, or 23.
- the intermediate promoter is selected from pHL, pA12L, pA19L, pA42R, pD13L, pA3L, or pA27L.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, at least 99% identical, or 100% identical to the nucleotide sequence of any one of SEQ ID NO: 25-31.
- the IL-12 is human IL-12.
- the IL- 12 is a fusion protein comprising an IL- 12 p40 subunit and an IL- 12 p35 subunit.
- the IL- 12 is a fusion protein comprising an IL- 12 p40 subunit and an IL-12 p35 subunit.
- the IL-12 p40 subunit comprises the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 17.
- the IL-12 p35 subunit comprises the amino acid sequence of SEQ ID NO: 19 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 19.
- the IL-12 p40 subunit and the IL-12 p35 subunit are fused in a single polypeptide via an amino acid linker.
- the amino acid linker is about 5 to about 10 amino acids in length, optionally wherein said amino acid linker is 7 amino acids in length.
- the amino acid linker is a glycine-serine linker.
- the amino acid linker comprises the amino acid sequence of SEQ ID NO: 18.
- the IL-12 comprises the amino acid sequence of SEQ ID NO: 20 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 20.
- the IL-12 p40 subunit and the IL- 12 p35 subunit are directly fused in a single polypeptide.
- the heterologous nucleic acid sequence encoding the IL-12 comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 21, optionally wherein the heterologous nucleic acid sequence encoding the IL-12 comprises the nucleotide sequence of SEQ ID NO: 21.
- the recombinant poxvirus is defective for thymidine kinase (TK) activity. In some aspects, the recombinant poxvirus lacks a functional J2R gene. In some aspects, the recombinant poxvirus is defective for ribonucleotide reductase (RR) activity. In some aspects, the recombinant poxvirus lacks a functional I4L gene. In some aspects, the recombinant poxvirus lacks a functional F4L gene. In some aspects, the heterologous nucleic acid sequence encoding IL-12 is inserted within the J2R locus of the poxvirus genome. In some aspects, the insertion renders the J2R gene nonfunctional, optionally wherein the J2R locus is fully deleted by the insertion.
- the recombinant poxvirus is capable of lysing one or more cancer cells.
- the recombinant poxvirus is capable of expressing at least 50 ng/mL, at least 100 ng/mL, at least 300 ng/mL, at least 500 ng/mL, at least 1.0 pg/mL, at least 2.0 pg/mL, at least 3.0 pg/mL , at least 4.0 pg/mL, at least 5.0 pg/mL, at least 6.0 pg/mL, at least 7.0 pg/mL, at least 8.0 pg/mL, or about 8.3 pg/mL of the IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10-2.
- MOI multiplicity of infection
- the recombinant poxvirus is capable of upregulating interferon (IFN)-y.
- the PD-1 inhibitor or PD-L1 inhibitor is an anti-PD-1 antibody or antigen-binding fragment thereof or an anti-PD-Ll antibody or antigen-binding fragment thereof.
- the anti-PD-1 antibody or antigen-binding fragment thereof or an anti-PD-Ll antibody or antigen-binding fragment thereof was produced in a Chinese Hamster Ovary (CHO) cell.
- the PD-1 inhibitor or the PD-L1 inhibitor is a small molecule.
- the PD-1 inhibitor or the PD-L1 inhibitor is a PD-1 inhibitor.
- the PD-1 inhibitor or the PD- L1 inhibitor is a PD-L1 inhibitor.
- the PD-1 inhibitor or PD-L1 inhibitor is selected from the group consisting of: nivolumab, pembrolizumab, lambrolizumab, MEDI-0680, cemiplimab, JS001, BGB-A317, INCSHR1210, TSR-042, Pidilizumab, GLS-010, STI-1110, AGEN2034, MGA012, IBI308, AMP-224, BMS-936559, atezolizumab, MPDL3280A, RG7446, Durvalumab, avelumab, STI-1014, CX-072, KN035, and CK-301.
- the PD-1 inhibitor or PD-L1 inhibitor is an anti- PD-Ll antibody or antigen-binding fragment thereof comprising a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 32, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 33, a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 34, a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 35, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 36, and a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 37.
- the anti-PD-Ll antibody or antigen-binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 38 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 39.
- the anti-PD-Ll antibody or antigen-binding fragment thereof further comprises an Fc variant, wherein the Fc variant comprises at least one amino acid substitution selected from the group consisting of 234F, 235F, and 33 IS, as numbered by the EU index as set forth in Kabat.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab.
- the cancer is renal cancer, prostate cancer, breast cancer, bladder cancer, colorectal cancer, lung cancer, liver cancer, gastric cancer, bile duct carcinoma, endometrial cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, glioblastoma, multiple myeloma, or malignant glioma.
- the cancer is resistant to an immune checkpoint inhibitor therapy.
- the cancer is resistant to a PD1 inhibitor.
- the cancer is resistant to a PD-L1 inhibitor.
- the effective amount for an individual dose of the recombinant poxvirus comprises from IxlO 3 pfu to IxlO 12 pfu, optionally from IxlO 4 pfu to IxlO 11 pfu, optionally from IxlO 5 pfu to IxlO 10 pfu, optionally from 5xl0 7 pfu to 4xl0 9 pfu.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab, and the effective amount for an individual dose of Durvalumab is 10 mg/kg. In some aspects of the methods disclosed herein, the PD-1 inhibitor or PD- L1 inhibitor is Durvalumab, and the effective amount for an individual dose of Durvalumab is 1500 mg. In some aspects, the Durvalumab is administered in a dose of 10 mg/kg every 2 weeks, 1500 mg every 4 weeks, or 1500 mg every 3 weeks.
- the administering results in an enhanced therapeutic effect compared to treatment with the recombinant poxvirus alone or to treatment with the PD-1 inhibitor or PD-L1 inhibitor alone.
- the subject is human.
- the administration is intratumoral.
- the administration is intravenous.
- the intravenous administration is via intravenous infusion.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab, and the administration is in a dose of 10 mg/kg every 2 weeks.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab, and the administration is in a dose of 1500 mg every 3 weeks or every 4 weeks.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab, and the administration is in a dose of 1500 mg every 4 weeks. In some aspects, the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab, and the administration is in a dose of 1500 mg every 3 weeks.
- the recombinant poxvirus and/or the PD-1 inhibitor or PD-L1 inhibitor are administered two or more times.
- the PD-1 inhibitor or PD-L1 inhibitor is administered to the patient before administration of the recombinant poxvirus.
- the PD-1 inhibitor or PD-L1 inhibitor is administered to the patient at the same time as the recombinant poxvirus.
- the PD-1 inhibitor or PD-L1 inhibitor and the recombinant poxvirus are administered in separate pharmaceutical compositions.
- the PD-1 inhibitor or PD-L1 inhibitor and the recombinant poxvirus are administered in the same pharmaceutical composition.
- the PD-1 inhibitor or PD-L1 inhibitor is administered to the patient after administration of the recombinant poxvirus
- compositions comprising a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding IL-12 for use in treating cancer in a subject in need thereof, wherein the composition is for administration in combination with a PD-1 inhibitor or a PD-L1 inhibitor, optionally wherein the treating comprises any one of the methods disclosed herein.
- compositions comprising a PD-1 inhibitor or a PD-L1 inhibitor for use in treating cancer in a subject in need thereof, wherein the composition is for administration in combination with a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding IL-12, optionally wherein the treating comprises any one of the methods disclosed herein.
- a pharmaceutical composition comprising a recombinant poxvirus comprising in its genome (i) a heterologous nucleic acid sequence encoding IL- 12 and (ii) a PD-1 inhibitor or a PD-L1 inhibitor, optionally wherein the recombinant poxvirus is the recombinant poxvirus used in any one of the methods disclosed herein and/or wherein the PD-1 inhibitor or PD-L1 inhibitor is the PD-1 inhibitor or PD-L1 inhibitor used in any one of the methods disclosed herein.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab.
- kits comprising a unit dose of (i) a pharmaceutical composition comprising a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor, optionally wherein the recombinant poxvirus is the recombinant poxvirus used in any one of the methods disclosed herein and/or wherein the PD-1 inhibitor or PD-L1 inhibitor is the PD-1 inhibitor or PD-L1 inhibitor used in any one of the methods disclosed herein.
- the PD-1 inhibitor or PD-L1 inhibitor is Durvalumab. BRIEF DESCRIPTION OF THE FIGURES
- FIG. 1 is a schematic representation of plasmid pTG19409.
- FIG. 2 shows luciferase expression from six late promoters after infection/transfection of DF-1 cells.
- FIGS. 3 A and 3B show luciferase expression of after infection of MIA PaCa-2 cells with ten recombinant vaccinia viruses at 6 hours (A) and 24 hours (B).
- FIG. 4 shows luciferase expression after infection of HeLa cells.
- FIG. 5 shows luciferase expression after infection of HCT-116 cells.
- FIG. 6 shows GFP-positive cells after infection of human PBMC at 6 hours
- FIG. 7 shows luciferase expression after infection of human PBMC at 6 hours and 24 hours.
- FIG. 8 shows the sequence of the hIL-12 expression cassette of COPTG19673 (SEQ ID NO:21).
- FIG. 9 is a schematic map of plasmid pTG19673.
- FIG. 10 is a schematic map of plasmid pTG19674.
- FIG. 11 shows the expression of IL-12 in supernatants of A549 cells infected with COPTG19673 and COPTG19674 of primary research stocks measured by ELISA.
- FIGS. 12A-C show the replication of VACV-IL-12 (COPTG19673 and COPTG19674) and unarmed control VACV (VVTG18058) in human tumor cell lines A549 (A), Mi PaCa-2 (B), and HT-29 (C), at 24 hours, 48 hours, and 72 hours postinfection.
- VACV-IL-12 COPTG19673 and COPTG19674
- VVTG18058 unarmed control VACV
- FIG. 13 shows the replication of VACV-IL-12 (COPTG19673 and COPTG19674) and unarmed control VACV (VVTG18058) in production cells (HeLa and CEF) at 72 hours post-infection. Results are mean of three wells.
- FIGS. 14A-C show: the oncolytic activities of COPTG19673, COPTG19674, and empty VACV (VVTG18058), in three different human tumor cell lines A549 (A), MIA PaCa-2 (B), and HT-29 (C) at various MOIs.
- the results shown are means +/- SD of 3 measurements and are represented as percentage of cellular viability (100 % corresponds to mock infected cells).
- FIG. 15 shows the expression level of vIL-12 in supernatants of COPTG19673 and COPTG19674 infected A549, MIA PaCa-2, and HT-29 cells after 3 days of incubation after infection at MOI 0.01. The results are the means and SD of duplicate measurements on three samples.
- FIGS. 16A-F show the IL-12 biological activity after HEK-BlueTM IL-12 cells were incubated with supernatant of COPTG19673-infected tumor cell lines (A-C), and with supernatant of COPTG19674-infected tumor cell lines (D-F), as compared to rhlL- 12, as determined using HEK Blue IL-12 Cell reporter assay.
- the results are represented as mean ⁇ SD of two measurements.
- FIGS. 17A-F show the IL-12 biological activity after NK-92 cells were incubated with supernatant of COPTG19673-infected tumor cell lines (A-C), supernatant of COPTG19674-infected tumor cell lines (D-F), as compared to rhIL-12, as determined using a NK-92 proliferation assay.
- FIG. 18 shows the replication yield of VACVwt, unarmed control VACV (VVTG18058), COPTG19673, and COPTG19674, on human hepatocytes.
- the results are represented as replication yield corresponding to ratio between input/output virus quantities. Results are mean of three wells.
- FIG. 19 shows the replication of VACVwt, VVTG18058 and COPTG19673 and COPTG19674 on Human PBMC.
- the results are represented as replication yield corresponding to ratio between input/output virus quantities.
- the results are mean of three wells.
- FIG. 20 shows the potency of virus-mediated oncolysis in cultured human tumor cells incubated with VACV IL-12 at different MOIs in each of three independent experiments to determine the mean EC50 for cellular lysis. (See also FIG. 29B.)
- FIGS. 21 A-B show virus recovered from tumors by plaque formation assay (PFU of virus per gram of tumor tissue) (A) and intra-tumoral IL- 12 - (ng of IL- 12 per gram of tumor) using a human IL-12-specific ELISA (B).
- PFU plaque formation assay
- B intra-tumoral IL- 12 - (ng of IL- 12 per gram of tumor) using a human IL-12-specific ELISA
- FIGS. 22A-E show the experiment protocol (A), spider plots of tumor growth in C57BL/6 mice engrafted with subcutaneous MC38 colorectal tumors after multi-dose, intra-tumoral administration of VACV expressing murine IL-12 (B-D), and a Kaplan- Meier plot of survival of tumor-bearing mice (E).
- 23A-E show the expression of murine IL-12 in the peripheral blood of mice 4 and 24 hours after intra-tumoral administration of VACV muIL-12 (A), and the expression of cytokines IFNy (B), CXCL10 (C), IL-6 (D) and TNFa (E). *, p ⁇ 0.05, Oneway analysis of variance with Tukey’s post-test.
- FIGS. 24A-H show anti-tumor activity of luciferase-expressing VACV (VACV luc) determined using primary tumors from patients with cancer (PDX: patient derived xenografts) engrafted into immunocompromised NOD/SCID mice.
- PDX patient derived xenografts
- Each tumor tested was either left untreated (open circles, gray line) or was administered le7 VACV luc virus (closed square, black line).
- Each primary patient-derived tumor included only one untreated and one treated tumor, so each open circle and closed square represent a pair but are graphed together within a tumor type for simplicity.
- FIGS. 25A-F show the mRNA expression levels of IL-12RB1 (A), IL-12RB2 (B), NKp46 (NK cells) (C), PD-L1 (D), CXCL9 (E), and CXCL10 (F) genes across mouse stroma isolated from bladder, head and neck, liver, colon, lung, and ovarian cancer patient-derived xenograft models from VACV-Luciferase treated mice 48 hours after day 0 and day 14.
- FIGS. 26A-E show the efficiency of VACV-IL12 infection in human tumors in vitro as measured by detection of IL-12p70 in supernatant from dissociated tumor cell cultures (A), expression of IL-12p70 in supernatant from tumor slice culture in (B), levels of IFNy mRNA (C), levels of IFNy protein (D), and levels of B8R mRNA (E). **** ⁇ 0.0001, one-way ANOVA with Tukey’s correction for multiple comparisons. Each dot is a single slice with 1-4 replicates per condition.
- FIG. 26F is a schematic diagram displaying the timing of VACV infection, transgene production, and immune-activation.
- FIGS. 27A-C show that COPTG19673 replicates selectively in tumor cells relative to normal human cells. There was no observable amplification in PBMCs, and minimal replication in normal hepatocytes (HUCPG and HUCPI) and normal human dermal fibroblasts (NHDF) relative to cancer cells (SW780).
- HUCPG and HUCPI normal hepatocytes
- NHDF normal human dermal fibroblasts
- FIGS. 28A-C show the experimental protocol (A), oncolytic VACV efficacy and overall response across multiple tumor indications (B), and replication kinetics observed 48 hours after the 1 st and 3 rd dose (C).
- CRC colorectal cancer
- HN head and neck squamous cell cancer.
- FIGS. 29A-E show oncolytic activity of VACV-Luc and VACV IL-12 (COPTG1673) across 30 human cancer cell lines, representing 12 tumor indications.
- Figure A shows the oncolytic activity of VACV-Luc in cultured human cancer cell lines.
- Figure B shows oncolytic activity of VACV-IL12 in cultured human cancer cell lines (see also FIG. 20).
- Figure C shows the correlation of VACV Luc and VACV-IL12 oncolysis of human tumor cell lines.
- Figures D and E show VACV-IL12 transgene production and replication in the tumor cell lines 5 days post infection.
- FIGS. 30A-F show anti-tumor efficacy of VACV IL-12 (COPTG1673) in human xenograft tumor models.
- Figures A-C show the mean tumor volume change over time for various doses of VACV-IL12 in comparison to VACV-Luc in mice bearing SW780 tumors, NCI-H292 tumors, and HCT-116 tumors.
- Figures D and E show the amount of virus and recovered intra-tumoral IL-12 over time for each dose in SW780 tumor- engrafted mice.
- Figure F shows human IL12 in mouse plasma over time for each dose level for VACV-IL12 in comparison to VACV-Luc in the SW780 tumor model.
- FIGS. 31A-D show TSC culture gene expression analysis following VACV IL-12 (COPTG1673) infection.
- Figure A shows production of IL-12p70 in VACV-IL12- infected cells in comparison to mock-infected or VACV GFP-infected groups.
- Figures B- D show production of IFN proteins (IFNy in Figure B, IFNa2a in Figure C, and IFNP in Figure D) in VACV-IL12-infected cells in comparison to mock-infected or VACV GFP- infected groups.
- FIGS. 32A-H show that VACV-muIL12 demonstrates similar oncolytic activity to VACV IL-12 (COPTG1673) in human tumor cells and similar IL-12 bioactivity.
- Figure A shows a dose dependent relationship between culture supernatant dilutions from VACV-IL12 infected cells and rat IFNy neutralization.
- Figures B-C show virus ( Figure B) and IL12 transgene (Figure C) production across 3 tumor cell lines for VACV-GFP and VACV-muIL12.
- Figures D-F show the percent survival over the multiplicity of infection for VACV-GFP and VACV-muIL12 in each cancer cell line.
- Figures G-H show the IL12p70 (Figure G) and IFNy (Figure H) plasma levels over time in rats engrafted with F98 rat gliomas and treated intravenously with vehicle, VACV-luc, or three doses of VACV-muIL12.
- FIGS. 33A-E show VACV-muIL12 demonstrates similar oncolytic activity to VACV IL-12 (COPTG1673) in cultured human tumor cells and similar IL-12 bioactivity.
- Figures A-C show percent survival over multiplicity of infection for VACV-LUC, VACV-muIL12, and VACV-huIL12 using cultured SW780 bladder, NCI-H292 lung, and HCT-116 colorectal tumor cell lines.
- Figure D shows IL-12p70 concentrations produced by VACV-Luc, VACV-muIL12, and VACV-huIL12 in human and murine cells for SW780, NCI-H292, and HCT-116 tumor cell lines.
- Figure E shows IL-12 activity for varying concentrations of recombinant human IL- 12 (rIL-12), and IL- 12 measured in cell culture supernatants from cultured SW780 tumor cells treated with VACV-muIL12, VACV-huIL12, or VACV-Luc.
- FIGS. 34A-F show VACV-muIL12 demonstrates similar oncolytic activity to VACV-LUC in murine tumor cells and produces IL-12 with moderate viral replication across tumor cell lines.
- Figures A-D show the percent survival over the multiplicity of infection for VACV-Luc and VACV-muIL12 in murine tumor cell lines.
- Figure E shows the IL-12 concentration produced in multiple murine tumor cell lines for VACV-muIL-12 and VACV-Luc over a period of 5 days.
- Figure F shows the amount of virus recovered in each murine tumor cell line for VACV-luc and VACV-muIL12 on day 5.
- FIGS. 35A-G show VACV-muIL12 enhances the anti-tumor immune response in a murine syngeneic CT26 tumor model.
- Figure A shows the general procedure.
- Figure B shows the survival curve for the vehicle control, VACV-Luc (1X10 7 PFU), and VACV- muIL12 (1X10 7 PFU). ****, p ⁇ 0.0001.
- Figures C-E show the change in tumor volume for the vehicle control group, VACV-Luc, and VACV-muIL12.
- Figure F shows the mouse plasma concentration of IL12p70 at 4 and 24 hours post injection for the vehicle control group, VACV-Luc, and VACV-muIL12.
- FIGS. 36A-C show the tumor volumes over time in CT26 tumor engrafted mice after administration with vehicle (A), VACV-LUC (B), or VACV-muIL12 (C).
- vehicle A
- VACV-LUC B
- VACV-muIL12 C
- the Y axis shows the tumor volume
- the X axis shows the time in days.
- FIGS. 37A-B show the results of ex vivo splenocyte restimulation assays post- VACV dosing and stimulated with either the VACV specific peptide A52L (A) or tumor associated peptide antigen AH-1 (B).
- the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited, elements or method steps. It is contemplated that any aspect discussed in this specification can be implemented with respect to any recombinant virus (e.g., poxvirus), method, system, host cell, expression vector, and/or composition of the present disclosure.
- recombinant virus e.g., poxvirus
- Nucleic acid refers to a polymeric compound that includes covalently linked nucleotides.
- DNA includes, but is not limited to, complementary DNA (cDNA), genomic DNA, plasmid or vector DNA, and synthetic DNA.
- a “gene” refers to an assembly of nucleotides that encodes a gene product, either RNA or protein. Genes include cDNA and genomic DNA molecules.
- a “functional” gene refers to a gene that is capable of expressing an RNA or protein product, wherein the RNA or protein product retains at least one functional activity.
- a “nonfunctional” gene refers to a gene that is not capable of expressing an RNA or protein product that retains any functional activity.
- a nonfunctional gene can refer to a gene that has been completely removed or replaced.
- a nonfunctional gene can also refer to a gene that has been partly removed or replaced, wherein the partial removal or replacement renders the remaining portion of the gene incapable of expressing an active RNA or protein product.
- a “coding sequence” is a nucleic acid sequence that can be transcribed and translated into a polypeptide in a cell in vitro or in vivo when placed under the control of appropriate regulatory sequences.
- “Regulatory sequences” include nucleotide sequences located upstream (5’ non-coding sequences), within, or downstream (3’ non-coding sequences) of a coding sequence, and which influence the transcription, RNA processing or stability, or translation of the associated coding sequence. Regulatory sequences include promoters, translation leader sequences, introns, polyadenylation recognition sequences, RNA processing sites, effector binding sites and stem-loop structures.
- Coding sequences include, but are not limited to, prokaryotic sequences, cDNA from mRNA, genomic DNA sequences, and synthetic DNA sequences. If a coding sequence is intended for expression in a eukaryotic cell, a polyadenylation signal and transcription termination sequence can be located 3’ of the coding sequence.
- ORF Open reading frame
- nucleic acid sequence either DNA, cDNA or RNA, that includes a translation start signal or initiation codon such as an ATG or AUG, and a termination codon and that can be potentially translated into a polypeptide sequence.
- “Homologous recombination” refers to the insertion of a foreign DNA sequence (the “inserted DNA sequence”) into another DNA molecule (the “target DNA sequence”).
- the inserted DNA sequences is targeted to a specific site within the target DNA sequence for homologous recombination.
- the inserted DNA sequence typically contains sufficiently long regions of homology to a sequence of the target DNA sequence to allow complementary binding and incorporation of the inserted DNA sequence into the target DNA sequence. Longer regions of homology, and greater degrees of sequence similarity, generally increase the efficiency of homologous recombination.
- Heterologous describes the relationship of one nucleic acid or amino acid sequence to one or more different nucleic acid or amino acid sequences and indicates that the sequences are not found joined together in the same position, structure, and orientation in nature.
- the joining of heterologous sequences creates a non-naturally occurring juxtaposition of sequences.
- Such joining is the product of engineering performed in the laboratory.
- the products of such joining can be referred to as “recombinant.”
- a linker is a chemical linker.
- the linker comprises one or more amino acids.
- a glycine-serine linker is one which contains both glycine and serine amino acids in any proportion, e.g., GGGS.
- “Operably linked” means that a polynucleotide of interest is linked to a regulatory element in a manner that allows for expression of the polynucleotide sequence.
- the regulatory element is a promoter.
- “Promoter” refers to a nucleic acid sequence that regulates, either directly or indirectly, transcription of a nucleic acid coding sequence to which it is operably linked.
- An “endogenous promoter” is a promoter that is naturally associated with a gene or nucleic acid sequence.
- An endogenous promoter can be obtained, for example, by isolating the 5' non-coding sequences located upstream of the coding segment and/or exon.
- a “recombinant” or “heterologous” promoter is a promoter that is not normally associated with a nucleic acid sequence in its natural environment.
- a “late promoter” is a promoter that is naturally associated with expression of a late gene.
- An “intermediate promoter” is one that controls the expression of a gene that follows early gene expression but is not controlled by a late promoter. Genes expressed early in the viral lifecycle preceding intermediate and late gene expression are termed “early promoters.” The expression of late and intermediate genes controlled by intermediate/late promoters depends on viral replication (as opposed to expression of an early gene whose expression does not depend on viral replication). The timeframe of expression of early, intermediate, and late promoters is a distinguishing factor, as discussed in Yang, et. al. J. Virol., Vol. 85, No. 19, p.
- Baldick disclosed that early-, intermediate-, and late-class mRNAs can be detected in 20, 100, and 140 min, respectively, after synchronous infection of HeLa cells with VACV.
- Yang prepared a genome-wide early, intermediate, and late transcription map which revealed distinctive characteristics of intermediate and late promoters.
- intermediate promoter and “late promoter” can refer to any of the intermediate and late promoters discussed in Yang, in particular the promoters shown in Fig. 8, therein.
- Exemplary late promoters include but are not limited to: pAlOL, pAl 1R, pA13L, pA14L, pA26L, pG7L, and pF17R.
- Exemplary intermediate promoters include but are not limited to: pHL, pA12L, pA19L, pA42R, pD13L, pA3L, or pA27L.
- Vector refers to a carrier nucleic acid molecule or vehicle that can be introduced into a cell where it can be replicated.
- “Expression vector” refers to a vector containing a nucleic acid sequence encoding at least part of a gene product capable of being transcribed. Expression vectors typically contain one or more control sequences necessary for transcription and/or translation of an operably linked coding sequence. Vectors can be introduced into the desired host cells by known methods, including, but not limited to, transfection, transduction, cell fusion, and lipofection.
- Transfection refers to the introduction of an exogenous nucleic acid molecule into a cell.
- a “transfected” cell includes an exogenous nucleic acid molecule inside the cell, and a “transformed” cell is one in which the exogenous nucleic acid molecule within the cell induces a phenotypic change in the cell.
- peptide refers to a polymeric form of amino acids of any length, which can include coded and non-coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones.
- the start of the protein or polypeptide is known as the “N-terminus” (or aminoterminus, NH2-terminus, N-terminal end, or amine-terminus), referring to the free amine (-NH2) group of the first amino acid residue of the protein or polypeptide.
- the end of the protein or polypeptide is known as the “C-terminus” (or carboxy -terminus, carboxylterminus, C-terminal end, or COOH-terminus), referring to the free carboxyl group (- COOH) of the last amino acid residue of the protein or peptide.
- amino acid refers to a compound including both a carboxyl (- COOH) and amino (-NH2) group. “Amino acid” refers to both natural and unnatural, e.g., synthetic, amino acids.
- Natural amino acids include: Alanine (Ala; A); Arginine (Arg, R); Asparagine (Asn; N); Aspartic acid (Asp; D); Cysteine (Cys; C); Glutamine (Gin; Q); Glutamic acid (Glu; E ); Glycine (Gly; G); Histidine (His; H); Isoleucine (He; I); Leucine (Leu; L); Lysine (Lys; K); Methionine (Met; M); Phenylalanine (Phe; F); Proline (Pro; P); Serine (Ser; S); Threonine (Thr; T); Tryptophan (Trp; W); Tyrosine (Tyr; Y); and Valine (Vai; V).
- amino acid substitution in a polypeptide or protein refers to a polypeptide or protein including one or more substitutions of a wild-type or naturally occurring amino acid with a different amino acid relative to the wild-type or naturally occurring amino acid at that amino acid residue.
- the substituted amino acid can be a synthetic or naturally occurring amino acid.
- the substituted amino acid is a naturally occurring amino acid selected from the group consisting of: A, R, N, D, C, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y, and V. Substitution mutants can be described using an abbreviated system.
- a substitution mutation in which the fifth (5 th ) amino acid residue is substituted can be abbreviated as “X5Y” wherein “X” is the wild-type or naturally occurring amino acid to be replaced, “5” is the amino acid residue position within the amino acid sequence of the protein or polypeptide, and “Y” is the substituted, or nonwild-type or non-naturally occurring, amino acid.
- isolated polypeptide, protein, peptide, or nucleic acid has been removed from its natural environment. It is also to be understood that “isolated” polypeptides, proteins, peptides, or nucleic acids can be formulated with excipients such as diluents or adjuvants and still be considered isolated.
- recombinant when used with reference to a nucleic acid molecule, peptide, polypeptide, or protein means of, or resulting from, a new combination of genetic material that is not known to exist in nature.
- a recombinant molecule can be produced by any well-known technique available in the field of recombinant technology, including, but not limited to, polymerase chain reaction (PCR), gene splicing (e.g., using restriction endonucleases), and solid-phase synthesis of nucleic acid molecules, peptides, or proteins.
- PCR polymerase chain reaction
- gene splicing e.g., using restriction endonucleases
- solid-phase synthesis of nucleic acid molecules, peptides, or proteins solid-phase synthesis of nucleic acid molecules, peptides, or proteins.
- a “poxvirus” refers to a virus of the Poxviridae family, including, e.g., viruses of the Orthopoxvirus genus.
- the “genome” of a recombinant poxvirus as provided herein includes poxviruses genomes containing one or more deletions (removal) of endogenous sequences (genes or nucleotides) and/or addition of one or more heterologous sequences (genes and/or nucleotides).
- the genome of a recombinant poxvirus can refer to the genome of an attenuated poxvirus.
- Oncolytic virus refers to a DNA or RNA virus that preferentially infects and kills cancer cells as compared to normal cells. Oncolytic viruses can kill cancer cells through a number of mechanisms, including direct oncolysis or apoptosis of infected cells, apoptotic death of uninfected cells, and by inducing an immune response against the cancer cells. In direct oncolysis, the virus causes lysis or apoptosis of a host cell as a direct result of replication or infection.
- Oncolytic activity refers to the ability of a virus to preferentially infect and kill cancer cells relative to normal cells. Cancer cell death can be caused by preferential infection, replication in and destruction of the cancer cells (referred to as “direct cytotoxic activity”) and by simulating and amplifying the host anti-cancer immune response, which, in addition to destroying existing cancer cells, can establish lasting immunity. Oncolytic activity can be detected by known methods, including, but not limited to, detecting cell death or apoptosis, inhibition of cell proliferation and/or by detecting a reduction in tumor size. [0109] A virus is considered “cytotoxic” if it reduces cell viability in treated target cells relative to untreated target cells.
- Methods for determining cytotoxicity of viruses include, for example, cytotoxicity assays that measure cell necrosis and/or apoptosis following virus infection, such as MTT (3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide) assays and other related tetrazolium salt based assays (e.g. XTT, MTS or WST), ATP assays, apoptosis assays, such as TUNEL staining of infected cells, DNA fragmentation assays, DNA laddering assays, and cytochrome C release assays.
- Another method for determining cytotoxicity is to monitor tumor size and location before and after treatment. In some cases, it may be desirable to monitor size over several time points for information regarding the increase or decrease in size of a tumor or metastasis.
- Attenuated virus refers to a virus that is not pathogenic and has reduced toxicity towards normal or non-cancerous cells.
- An attenuated virus can be recombinantly modified to be less virulent, or non-virulent in normal tissues. In some aspects, modification does not or only minimally impacts the oncolytic ability of the virus.
- a “pathogenic virus” is a virus that produces disease.
- a recombinant poxvirus provided herein is not a pathogenic virus.
- Replication competent refers to the ability of a virus to replicate in a cell or cell line and produce infectious progeny virions.
- a virus that can produce infectious progeny virions in a cell or cell line is considered “replication competent,” whereas a virus that is not able to produce infectious progeny virus in the cell or cell line is considered “replication defective.”
- Viral replication can be expressed by the ratio of virus produced by an infected cell to the amount used to infect the cell, referred to as the “amplification ratio.”
- An amplification ratio of 1 or more means that the amount of virus produced from the infected cells is the same or more than the amount used to infect the cell, which indicates that replication has taken place, whereas an amplification ratio of less than 1 means that the amount of virus produced from the infected cells is less than the amount used to infect the cell, which indicates a lack of replication in the cell.
- interleukin- 12 refers to a protein comprising a p35 subunit (IL-12A) and a p40 subunit (IL-12B).
- the p35 subunit and the p40 subunit can be expressed as separate proteins that heterodimerize or can be expressed together as a single fusion protein.
- cell proliferative disorder and “proliferative disorder” refer to disorders that are associated with some degree of abnormal cell proliferation.
- a cell proliferative disorder can include cancer.
- Cancer refers to a physiological condition in mammals that is characterized by unregulated cell growth, lack of differentiation, local tissue invasion and/or metastasis. Tumor” refers to an abnormal growth of cells of tissues.
- cancer or “cancerous” refers to a physiological condition in mammals that is characterized by unregulated cell growth, lack of differentiation, local tissue invasion and/or metastasis.
- Tumor refers to an abnormal growth of cells of tissues.
- cancer cancer, “cancerous,” “cell proliferative disorder,” “proliferative disorder,” and “tumor” are not mutually exclusive.
- an “effective amount” refers to an amount that is sufficient to reproducibly produce a detectable result, e.g., in vitro or when administered to a patient.
- a “therapeutically effective amount” refers to an amount sufficient to produce a therapeutically significant change in one or more symptoms of a condition when administered to a patient with the condition. In one aspect, a therapeutically effective amount is sufficient to treat cancer.
- a “PD-1 inhibitor” refers to an agent that decreases the amount or activity of PD- 1.
- a PD-1 inhibitor can be an agent that binds to PD-1 protein and inhibits its interaction with PD-L1.
- a PD-1 inhibitor can, for example, be an antibody or antigenbinding fragment thereof that binds to PD-1, a peptide-based inhibitor, a small molecule, or an antibody drug conjugate.
- a PD-1 inhibitor is an antibody or antigen-binding fragment thereof that binds to PD-1.
- PD-1 inhibitors include but are not limited to: Pembrolizumab (Keytruda), Nivolumab (Opdivo), and Cemiplimab (Libtayo).
- a “PD-L1 inhibitor” refers to an agent that decreases the amount or activity of PD-L1.
- a PD-L1 inhibitor can be an agent that binds to PD-L1 protein and inhibits its interaction with PD-1.
- a PD-L1 inhibitor can, for example, be an antibody or antigen-binding fragment thereof that binds to PD-L1, a peptide-based inhibitor, a small molecule, or an antibody drug conjugate.
- a PD-L1 inhibitor is an antibody or antigen-binding fragment thereof that binds to PD-L1.
- PD-L1 inhibitors include Atezolizumab (Tecentriq), Avelumab (Bavencio), and Durvalumab (Imfinzi).
- antibody and “immunoglobulin” are used interchangeably and refer to an antibody molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing (e.g., a glycoprotein), through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
- a target such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing (e.g., a glycoprotein), through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
- a target such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing (e.g., a glycoprotein)
- antibody encompasses monoclonal antibodies, chimeric antibodies, humanized antibodies, human antibodies, bispecific
- An antibody can be of any the five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g. IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively.
- the different classes of antibodies have different and well known subunit structures and three-dimensional configurations. For the structure and properties of the different classes of antibodies, see, e.g., Basic and Clinical Immunology, 8th Ed., Daniel P. Stites, Abba I. Terr and Tristram G. Parslow (eds.), Appleton & Lange, Norwalk, CT, 1994, page 71 and Chapter 6.
- antibody fragment refers to a portion of an antibody.
- An "antigenbinding fragment” of an antibody refers to a portion of an antibody that binds to an antigen.
- An antigen-binding fragment of an antibody can comprise the antigenic determining regions of an antibody (e.g., the complementarity determining regions (CDRs)).
- CDRs complementarity determining regions
- antigen-binding fragments of antibodies include, but are not limited to Fab, Fab', F(ab')2, and Fv fragments, linear antibodies, and single chain antibodies.
- An antigen-binding fragment of an antibody can be monovalent or multivalent (e.g., bivalent).
- An antigen-binding fragment of an antibody can be derived from any animal species, such as rodents (e.g., mouse, rat, or hamster) and humans or can be artificially produced.
- an “antigen-binding domain” or “antigen-binding region” refers to a monovalent portion of an antibody that binds to an antigen.
- An “antigen-binding domain” can comprise the antigenic determining regions of an antibody (e.g., the complementarity determining regions (CDRs)).
- An antibody or antigen-binding fragment thereof including mono-specific and multi-specific (e.g., bispecific) antibodies or antigenbinding fragments thereof can comprise an antigen-binding domain.
- each antigen binding domain is short, non-contiguous sequences of amino acids that are specifically positioned to form the antigen binding domain as the antibody assumes its three dimensional configuration in an aqueous environment.
- the remainder of the amino acids in the antigen binding domains referred to as "framework" regions, show less inter-molecular variability.
- the framework regions largely adopt a flsheet conformation and the CDRs form loops that connect, and in some cases form part of, the P-sheet structure.
- framework regions act to form a scaffold that provides for positioning the CDRs in correct orientation by inter-chain, non-covalent interactions.
- the antigen binding domain formed by the positioned CDRs defines a surface complementary to the epitope on the immunoreactive antigen. This complementary surface promotes the non-covalent binding of the antibody to its cognate epitope.
- the amino acids comprising the CDRs and the framework regions, respectively can be readily identified for any given heavy or light chain variable region by one of ordinary skill in the art, since they have been precisely defined (see, "Sequences of Proteins of Immunological Interest,” Kabat, E., et al., U.S. Department of Health and Human Services, (1983); and Chothia and Lesk, J. Mol. Biol., 796:901-917 (1987), which are incorporated herein by reference in their entireties).
- Kabat numbering and like terms are recognized in the art and refer to a system of numbering amino acid residues in the heavy and light chain variable regions of an antibody or an antigen-binding fragment thereof.
- CDRs can be determined according to the Kabat numbering system (see, e.g., Kabat EA & Wu TT (1971) Ann NY Acad Sci 190: 382-391 and Kabat EA et a/., (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
- CDRs within an antibody heavy chain molecule are typically present at amino acid positions 31 to 35, which optionally can include one or two additional amino acids, following 35 (referred to in the Kabat numbering scheme as 35A and 35B) (CDR1), amino acid positions 50 to 65 (CDR2), and amino acid positions 95 to 102 (CDR3).
- CDRs within an antibody light chain molecule are typically present at amino acid positions 24 to 34 (CDR1), amino acid positions 50 to 56 (CDR2), and amino acid positions 89 to 97 (CDR3).
- the CDRs of the antibodies described herein have been determined according to the Kabat numbering scheme.
- Chothia refers instead to the location of the structural loops (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987)).
- the end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35 A nor 35B is present, the loop ends at 32; if only 35 A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34).
- the AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody modeling software.
- the term “heavy chain” when used in reference to an antibody can refer to any distinct type, e.g., alpha (a), delta (d), epsilon (e), gamma (g), and mu (m), based on the amino acid sequence of the constant domain, which give rise to IgA, IgD, IgE, IgG, and IgM classes of antibodies, respectively, including subclasses of IgG, e.g., IgGl, IgG2, IgG3, and IgG4.
- Heavy chain amino acid sequences are well known in the art. In specific aspects provided herein, the heavy chain is a human heavy chain.
- the term “light chain” when used in reference to an antibody can refer to any distinct type, e.g., kappa (K) or lambda (X) based on the amino acid sequence of the constant domains. Light chain amino acid sequences are well known in the art. In specific aspects provided herein, the light chain is a human light chain
- variable domains of both the light (VL) and heavy (VH) chain portions determine antigen recognition and specificity.
- the constant domains of the light chain (CL) and the heavy chain (CHI, CH2 or CH3) confer important biological properties such as secretion, transplacental mobility, Fc receptor binding, complement binding, and the like.
- the N-terminal portion is a variable region and at the C- terminal portion is a constant region; the CH3 and CL domains comprise the carboxyterminus of the heavy and light chain, respectively.
- variable region allows the binding molecule to selectively recognize and specifically bind epitopes on antigens. That is, the VL domain and VH domain, or subset of the complementarity determining regions (CDRs), of a binding molecule, e.g., an antibody combine to form the variable region that defines a three dimensional antigen binding site.
- This quaternary binding molecule structure forms the antigen binding site present at the end of each arm of the Y. More specifically, the antigen binding site is defined by three CDRs on each of the VH and VL chains
- a "monoclonal” antibody or antigen-binding fragment thereof refers to a homogeneous antibody or antigen-binding fragment population involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants.
- the term "monoclonal” antibody or antigenbinding fragment thereof encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (such as Fab, Fab', F(ab')2, Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site.
- a “monoclonal” antibody or antigen-binding fragment thereof refers to such antibodies and antigen-binding fragments thereof made in any number of manners including but not limited to by hybridoma, phage selection, recombinant expression, and transgenic animals.
- human antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulins and that do not express endogenous immunoglobulins, as described infra and, for example in, U.S. Pat. No. 5,939,598 by Kucherlapati et al.
- “Patient,” “subject,” and “individual” are used interchangeably and refer to an animal to whom treatment is provided and includes human and non-human animals, including, for example, primates, cows, pigs, sheep, goats, dogs, cats, rabbits, and rodents, and non-mammals such as chickens, amphibians, and reptiles.
- the subject is human.
- the subject is a human with cancer.
- the subject is an experimental animal or animal disease model.
- treat refers to therapeutic treatment wherein the object is to reduce or eliminate one or more symptoms.
- Beneficial or desired results include, but are not limited to, elimination of symptoms, alleviation of symptoms, diminishing of the extent of condition, stabilized (e.g., not worsening) state of condition, delay or slowing of progression of the condition. Treating cancer can include inducing cell death in a cancer cell or cells within a tumor.
- Tumor progression refers to stages of a tumor, including tumorigenesis, tumor growth and proliferation, invasion, and metastasis.
- “Inhibiting tumor progression” refers to inhibiting the development, growth, proliferation, or spreading of a tumor, including, for example: inhibition or reduction of tumor growth; reduction in the number of cancer cells; reduction in tumor size; inhibition or reduction of cancer cell infiltration into adjacent peripheral organs and/or tissues; inhibition or reduction of metastasis; increase in the length of survival of a patient or patient population following treatment; and/or decreased mortality of a patient or patient population at a given timepoint following treatment.
- combination can include a fixed combination in one dosage unit form, separate dosage units, or a kit of parts or instructions for the combined administration where the recombinant poxvirus and the PD- 1/PD-L1 inhibitor can be administered independently at the same time or separately within time intervals.
- a combined pharmaceutical composition can be adapted for simultaneous, separate, or sequential administration. Accordingly, administration "in combination with" one or more further therapeutic agents includes simultaneous (concurrent) or consecutive administration in any order.
- a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late or intermediate promoter.
- IL- 12 interleukin- 12
- Such recombinant poxviruses are especially advantageous in that they exhibit cell-specificity to preferentially kill cancer cells while minimizing any detrimental impact on healthy, non-cancerous cells.
- the poxvirus belongs to the Orthopoxvirus genus.
- the poxvirus belonging to the Orthopoxvirus genus is a vaccinia virus.
- the poxvirus belonging to the Orthopoxvirus genus is an oncolytic vaccinia virus.
- the oncolytic vaccinia virus is selected from the group consisting of Western Reserve (WR), Elstree, Wyeth, Lister, Tian Tan, LIVP and Copenhagen (Cop) strains.
- the oncolytic vaccinia virus is selected from the Copenhagen (Cop) strain.
- the genome of the recombinant poxvirus comprises at least 150 kilobases (kb), at least 175 kb, at least 180 kb, at least 185 kb, at least 190 kb, at least 192 kb, or at least 194 kb. In some aspects of the recombinant poxvirus provided herein, the genome of the recombinant poxvirus comprises about 150 kb to 200 kb.
- the recombinant poxvirus is attenuated.
- the recombinant poxvirus comprises a late promoter selected from pAlOL, pAl 1R, pA13L, pA14L, pA26L, pG7L, and pF17R.
- the late promoter is selected from pA14L, pA26L, and pF17R.
- the late promoter is pA14L or pF17R.
- the late promoter is pA14L.
- the late promoter is pF17R.
- the recombinant poxvirus comprises an intermediate promoter selected from pHL, pA12L, pA19L, pA42R, pD13L, pA3L, or pA27L.
- the late promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 11. In some aspects, the late promoter comprises the nucleotide sequence of SEQ ID NO: 11.
- the late promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 22. In some aspects, the late promoter comprises the nucleotide sequence of SEQ ID NO: 22.
- the late promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 13. In some aspects, the late promoter comprises the nucleotide sequence of SEQ ID NO: 13.
- the late promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 23. In some aspects, the late promoter comprises the nucleotide sequence of SEQ ID NO: 23.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 25. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 25.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 26. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 26.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 27. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 27.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 28. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 28.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 29. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 29.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 30. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 30.
- the intermediate promoter comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 31. In some aspects, the intermediate promoter comprises the nucleotide sequence of SEQ ID NO: 31.
- the recombinant poxviruses provided herein comprise in their genome a heterologous nucleic acid sequence encoding IL-12.
- the IL-12 can be human IL-12.
- the IL-12 can be murine IL-12.
- the IL-12 is a fusion protein comprising an IL-12 p40 subunit and an IL-12 p35 subunit.
- the IL-12 p40 subunit can be N-terminal to the IL- 12 p35 subunit.
- the IL- 12 p40 subunit can be C-terminal to the IL- 12 p35 unit.
- the IL- 12 p40 subunit and the IL- 12 p35 subunit can be directly fused (i.e., without a linker) or can be fused via a linker.
- the linker can be, e.g., a chemical linker or an amino acid linker.
- An amino acid linker can be a glycine-serine linker. In some aspects, the linker is about 5 to about 10 amino acids in length. In some aspects, the linker is 7 amino acids in length. In some aspects, the linker comprises the amino acid sequence of SEQ ID NO: 18.
- Widely used vaccinia virus vectors include highly attenuated strains, such as New York Vaccinia virus (NYVAC).
- NYVAC New York Vaccinia virus
- the recombinant poxvirus is not NYVAC.
- the IL-12 p40 subunit comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 17.
- the IL-12 p40 subunit the amino acid sequence of SEQ ID NO: 17.
- the IL-12 p35 subunit comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 19. In some aspects of the recombinant poxvirus disclosed herein, the IL-12 p35 subunit the amino acid sequence of SEQ ID NO: 19.
- the IL-12 p40 subunit comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 17, and the IL-12 p35 subunit comprises an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 19.
- the IL- 12 p40 subunit the amino acid sequence of SEQ ID NO: 17 and the IL- 12 p35 subunit the amino acid sequence of SEQ ID NO: 19.
- the heterologous nucleic acid sequence encoding IL-12 comprises a nucleotide sequence at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, or at least 99% identical to the nucleotide sequence of SEQ ID NO: 21.
- the heterologous nucleic acid sequence encoding IL-12 comprises the nucleotide sequence of SEQ ID NO: 21.
- the heterologous nucleic acid sequence encodes for an IL- 12 amino acid sequence of SEQ ID NO: 20 or an amino acid sequence that is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the amino acid sequence of SEQ ID NO: 20.
- the poxvirus is defective for thymidine kinase (TK).
- TK thymidine kinase
- the poxvirus lacks a functional J2R gene
- the poxvirus is defective for ribonucleotide reductase (RR) activities.
- the poxvirus lacks a functional I4L gene.
- the poxvirus lacks a functional F4L gene.
- the poxvirus lacks a functional I4L gene and lacks a functional F4L gene.
- the poxvirus is defective for thymidine kinase (TK) and/or ribonucleotide reductase (RR) activities.
- the poxvirus lacks a functional J2R gene and lacks a functional I4L gene.
- the poxvirus lacks a functional J2R gene and lacks a functional F4L gene.
- the poxvirus lacks a functional J2R gene, lacks a functional I4L gene, and lacks a functional F4L gene.
- the heterologous nucleic acid sequence encoding IL- 12 is inserted within the J2R locus of the poxvirus genome.
- the insertion renders the J2R gene nonfunctional.
- the JR2 locus is fully deleted by the insertion. In some aspects, the JR2 locus is not fully deleted by the insertion.
- the heterologous nucleic acid sequence encoding IL- 12 is inserted within the I4L locus of the poxvirus genome.
- the insertion renders the I4L gene nonfunctional.
- the I4L locus is fully deleted by the insertion. In some aspects, the I4L locus is not fully deleted by the insertion.
- the heterologous nucleic acid sequence encoding IL-12 is inserted within the F4L locus of the poxvirus genome.
- the insertion renders the F4L gene nonfunctional.
- the F4L locus is fully deleted by the insertion. In some aspects, the F4L locus is not fully deleted by the insertion.
- the poxvirus in addition to encoding IL-12, further encodes one or more therapeutic genes.
- the one or more therapeutic genes is selected from the group consisting of a suicide gene, an immunomodulatory gene, an anti-angiogenic gene, an immune checkpoint blockade gene, an antibody-coding genes, an extracellular matrix degradation or modulation gene, or a combination thereof.
- the recombinant poxvirus is capable of lysis of one or more cancer cells.
- the recombinant poxvirus is capable of expressing at least 50 ng/mL, at least 100 ng/mL, at least 300 ng/mL, at least 500 ng/mL, at least 1.0 pg/mL, at least 2.0 pg/mL, at least 3.0 pg/mL, at least 4.0 pg/mL, at least 5.0 pg/mL, at least 6.0 pg/mL, at least 7.0 pg/mL, at least 8.0 pg/mL, or about 8.3 pg/mL of IL-12 in cancer cells (e.g., A549 cells) 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 .
- MOI multiplicity of infection
- the recombinant poxvirus is capable of expressing about 50 ng/mL to about 50 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 1 pg/mL to about 50 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 2 pg/mL to about 50 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 .
- the recombinant poxvirus is capable of expressing about 3 pg/mL to about 50 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 1 pg/mL to about 40 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 2 pg/mL to about 40 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 .
- the recombinant poxvirus is capable of expressing about 3 pg/mL to about 40 pg/mL of IL- 12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 1 pg/mL to about 30 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 2 pg/mL to about 30 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 .
- the recombinant poxvirus is capable of expressing about 3 pg/mL to about 30 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 1 pg/mL to about 25 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 2 pg/mL to about 25 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 . In some aspects, the recombinant poxvirus is capable of expressing about 3 pg/mL to about 25 pg/mL of IL-12 in cancer cells 72 hours after infection with a multiplicity of infection (MOI) of 10' 2 .
- cancer cells capable of being lysed by a recombinant poxvirus provided herein and/or capable of expressing 11-12 from a recombinant poxvirus provided herein include but are not limited to: renal cancer, prostate cancer, breast cancer, bladder cancer, colorectal cancer, lung cancer, liver cancer, gastric cancer, bile duct carcinoma, endometrial cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, glioblastoma, multiple myeloma, or malignant glioma cells.
- such cancer cells are A549, HT29, or MIA PaCa-2 cells.
- the virus is produced in chicken embryo fibroblasts (CEF), HeLa cells, EB66® cells, Vero cells, HEK 293 cells, PerC6 cells, BHK21 cells, or MRC5 cells. In some aspects of the recombinant poxvirus disclosed herein, the virus is produced in chicken embryo fibroblasts (CEF).
- CEF chicken embryo fibroblasts
- the recombinant poxvirus is capable of upregulating interferon (IFN)-Y
- the recombinant poxvirus disclosed herein is produced in a suitable host cell line or in suitable producer cells using conventional techniques including culturing the transfected or infected host cell under suitable conditions so as to allow the production and recovery of infectious poxviral particles.
- a method for producing a recombinant poxvirus comprises the steps of: a) obtaining or preparing producer cells; b) infecting the obtained or prepared producer cells with the recombinant poxvirus; c) culturing the infected producer cells under suitable conditions so as to allow the production of the recombinant poxvirus.
- such a method further comprises d) recovering the produced recombinant poxvirus from the culture of said producer cells. In some aspects, such a method further comprises e) purifying said recovered recombinant poxvirus.
- the producer cells are chicken embryo fibroblasts (CEF), HeLa, EB66®, Vero, HEK 293, PerC6, BHK21, or MRC5 cells. In some aspects, the producer cells are chicken embryo fibroblasts (CEF). Also provided herein are recombinant poxviruses produced by such methods.
- the producer cells can be cultured in step a) in an appropriate medium which can, if needed, be supplemented with serum and/or suitable growth factor(s) or not (e.g. a chemically defined medium free from animal-or human-derived products can be used).
- An appropriate medium can be selected by those skilled in the art, depending on the producer cells. Such media are commercially available.
- Producer cells are cultivated at a temperature between +30°C and +38°C (e.g., at approximately 37°C) for between 1 and 8 days before infection. If needed, several passages over 1 to 8 days can be made in order to increase the total number of cells.
- compositions comprising the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL- 12 is operably linked to a late promoter or intermediate promoter are also provided herein.
- the disclosure provides a pharmaceutical composition comprising the recombinant poxvirus described herein and a pharmaceutically acceptable carrier.
- the disclosure provides a composition comprising a therapeutically effective amount of said recombinant poxvirus and a pharmaceutically acceptable carrier.
- a therapeutically effective amount for an individual dose of the recombinant poxvirus described herein comprises from IxlO 3 pfu to IxlO 12 pfu. In some aspects, a therapeutically effective amount for an individual dose of the recombinant poxvirus described herein comprises from IxlO 4 pfu to IxlO 11 pfu. In some aspects, a therapeutically effective amount for an individual dose of the recombinant poxvirus described herein comprises from IxlO 5 pfu to IxlO 10 pfu. In some aspects, a therapeutically effective amount for an individual dose of the recombinant poxvirus described herein 5xl0 7 pfu to 4xl0 9 pfu.
- the disclosure provides a pharmaceutical composition recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter for use for treating or preventing a proliferative disease such as a cancer.
- IL- 12 interleukin- 12
- the cancer is selected from the group consisting of renal cancer, prostate cancer, breast cancer, bladder cancer, colorectal cancer, lung cancer, liver cancer, gastric cancer, bile duct carcinoma, endometrial cancer, pancreatic cancer, ovarian cancer, head and neck cancer, melanoma, glioblastoma, multiple myeloma, and malignant glioma.
- PD-1 is a key immune checkpoint receptor expressed by activated T and B cells and mediates immunosuppression.
- PD-1 is a member of the CD28 family of receptors, which includes CD28, CTLA-4, ICOS, PD-1, and BTLA.
- Two cell surface glycoprotein ligands for PD-1 have been identified, Programmed Death Ligand- 1 (PD-L1) and Programmed Death Ligand-2 (PD-L2), that are expressed on antigen-presenting cells as well as many human cancers and have been shown to downregulate T cell activation and cytokine secretion upon binding to PD-1. Inhibition of the PD-1/PD-L1 interaction mediates potent antitumor activity in preclinical models (U.S. Patent Nos.
- the FDA has approved six immune checkpoint inhibitors for the PD-1/PD-L1 pathway, including three for PD-1 (pembrolizumab, nivolumab and cemiplimab) and three for PD-L1 (atezolizumab, avelumab and durvalumab) (Ai et al., Drug Des Devel Ther., 14: 3625-3649, 2020).
- any one of the PD-L1 inhibitors and/or PD-1 inhibitors known in the art for the treatment of one or more cancers are suitable for inclusion in the therapeutic combinations and methods described herein.
- the inhibitor is a PD-1 inhibitor.
- a PD-1 inhibitor is an agent that binds to PD-1 protein and inhibits its interaction with PD-L1.
- a PD-1 inhibitor is an antibody or antigen-binding fragment thereof that binds to PD-1.
- a PD-1 inhibitor is a peptide-based inhibitor.
- a PD-1 inhibitor is a small molecule.
- a PD-1 inhibitor is an antibody drug conjugate comprising an antibody or antigen-binding fragment thereof that binds to PD-1.
- the inhibitor is a PD-L1 inhibitor.
- a PD-L1 inhibitor is an agent that binds to PD-L1 protein and inhibits its interaction with PD-1 and/or CD80.
- a PD-L1 inhibitor is an antibody or antigen-binding fragment thereof that binds to PD-L1.
- a PD-L1 inhibitor is a peptide- based inhibitor.
- a PD-L1 inhibitor is a small molecule.
- a PD-L1 inhibitor is an antibody drug conjugate comprising an antibody or antigenbinding fragment thereof that binds to PD-L1.
- the anti-PD-1 therapy is an antibody selected from nivolumab (also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538), pembrolizumab (Merck; also known as KEYTRUDA®, lambrolizumab, and MK-3475; see WO2008/156712), PDR001 (Novartis; see WO 2015/112900), MEDI- 0680 (AstraZeneca; also known as AMP-514; see WO 2012/145493), cemiplimab (Regeneron; also known as REGN-2810; see WO 2015/112800), JS001 (TAIZHOU JUNSHI PHARMA; see Si-Yang Liu et al., J.
- nivolumab also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538
- pembrolizumab Merck; also known as
- the anti-PD-1 therapy is the PD-1 antagonist AMP-224, which is a recombinant fusion protein comprised of the extracellular domain of the PD-1 ligand programmed cell death ligand 2 (PD-L2) and the Fc region of human IgG.
- AMP -224 is discussed in U.S. Publ. No. 2013/0017199. The contents of each of these references are incorporated by reference herein in their entirety.
- the anti-PD-Ll therapy is an antibody selected from durvalumab (AstraZeneca; also known as IMFINZITM, MEDI-4736; see WO 2011/066389), BMS- 936559 (also known as 12A4, MDX-1105; see, e.g., U.S. Patent No. 7,943,743 and WO 2013/173223), atezolizumab (Roche; also known as TECENTRIQ®; MPDL3280A, RG7446; see US 8,217,149; see, also, Herbst et al.
- avelumab Pfizer; also known as BAVENCIO®, MSB-0010718C; see WO 2013/079174), STI-1014 (Sorrento; see WO2013/181634), CX-072 (Cytomx; see W02016/149201), KN035 (3D Med/Alphamab; see Zhang et al., Cell Discov.
- the PD-L1 inhibitor is an anti-PD-Ll antibody or antigen-binding fragment thereof.
- the anti-PD-Ll antibody or antigen-binding fragment thereof comprises the CDR sequences provided in Table 3 (i.e., SEQ ID NOs: 32-37).
- the anti-PD-Ll antibody or antigen-binding fragment thereof comprises a VH comprising the amino acid sequence of SEQ ID NO:38. In some aspects, the anti-PD-Ll antibody or antigen-binding fragment thereof comprises a VL comprising the amino acid sequence of SEQ ID NO: 39. In some aspects, the anti-PD-Ll antibody or antigen-binding fragment thereof comprises a VH comprising the amino acid sequence of SEQ ID NO: 38 and a VL comprising the amino acid sequence of SEQ ID NO: 39. In some aspects, the anti-PD-Ll antibody or antigen-binding fragment thereof comprises an IgGl heavy chain (e.g., a human IgGl heavy chain).
- IgGl heavy chain e.g., a human IgGl heavy chain
- the anti-PD-Ll antibody or antigen-binding fragment thereof comprises a kappa light chain (e.g., a human kappa light chain).
- the anti-PD-Ll antibody is a human IgGl kappa monoclonal antibody comprising a VH comprising the amino acid sequence of SEQ ID NO: 38 and a VL comprising the amino acid sequence of SEQ ID NO: 39.
- the anti-PD-Ll antibody or antigen-binding fragment thereof was produced in a Chinese Hamster Ovary (CHO) cell.
- the anti-PD-Ll antibody is a human IgGl kappa monoclonal antibody comprising a VH comprising the amino acid sequence of SEQ ID NO: 38 and a VL comprising the amino acid sequence of SEQ ID NO: 39 produced in a CHO cell.
- the anti-PD-Ll antibody comprises a variant Fc comprising at least one amino acid substitution selected from the group consisting of 234F, 235F, and 331 S, as numbered by the EU index as set forth in Kabat.
- the anti-PD-Ll antibody comprises a variant Fc comprising the amino acid substitutions 234F, 235F, and 33 IS, as numbered by the EU index as set forth in Kabat.
- the anti-PD-Ll antibody is a human IgGl kappa monoclonal antibody comprising a variant Fc comprising the amino acid substitutions 234F, 235F, and 331 S, as numbered by the EU index as set forth in Kabat, a VH comprising the amino acid sequence of SEQ ID NO: 38, and a VL comprising the amino acid sequence of SEQ ID NO: 39.
- the anti-PD-Ll antibody is a human IgGl kappa monoclonal antibody comprising a variant Fc comprising the amino acid substitutions 234F, 235F, and 331 S, as numbered by the EU index as set forth in Kabat, a VH comprising the amino acid sequence of SEQ ID NO: 38, and a VL comprising the amino acid sequence of SEQ ID NO: 39 produced in a CHO cell.
- the anti-PD-Ll antibody or antigen-binding fragment thereof is provided in a 10 mL solution, wherein each mL of the solution comprises the antibody or antigen-binding fragment thereof (e.g., durvalumab), 50 mg, L-histidine (2 mg), L- histidine hydrochloride monohydrate (2.7 mg), a,a-trehalose dihydrate (104 mg), Polysorbate 80 (0.2 mg), and Water for Injection, USP.
- the antibody or antigen-binding fragment thereof e.g., durvalumab
- 50 mg e.g., L-histidine (2 mg), L- histidine hydrochloride monohydrate (2.7 mg), a,a-trehalose dihydrate (104 mg), Polysorbate 80 (0.2 mg), and Water for Injection, USP.
- the solution can comprise, e.g., 120 mg of the antibody or antigen-binding fragment thereof (e.g., durvalumab) in 2.4 mLs or 500 mg of the antibody or antigen-binding fragment thereof (e.g., durvalumab) in 10 mLs.
- the antibody or antigen-binding fragment thereof e.g., durvalumab
- 500 mg of the antibody or antigen-binding fragment thereof e.g., durvalumab
- the PD-L1 inhibitor is Durvalumab (Imfinzi®, AstraZeneca).
- Durvalumab is a human monoclonal antibody that selectively binds PD-L1 and blocks the binding of PD- L1 to the PD-1 and CD80 receptors, as disclosed in U.S. Patent No. 9,493,565, which is incorporated by reference herein in its entirety.
- the fragment crystallizable (Fc) domain of durvalumab contains a triple mutation in the constant domain of the IgGl heavy chain that reduces binding to the complement component Clq and the Fey receptors responsible for mediating antibody-dependent cell-mediated cytotoxicity (ADCC). Kits
- kits comprising a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL- 12 is operably linked to a late promoter or intermediate promoter, which is described herein, or a pharmaceutical composition comprising the recombinant poxvirus described herein.
- a kit comprises a unit dose of such a recombinant virus or pharmaceutical composition.
- a kit comprising one or more containers filled with one or more of the ingredients of the compositions described herein, such as the recombinant poxvirus described herein, optionally with instructions for use.
- kits described herein comprising one or more containers filled with one or more of the ingredients of the compositions described herein, such as the recombinant poxvirus described herein, and one or more PD-1 or PD-L1 inhibitors described herein, optionally with instructions for use.
- the kits described herein comprise a recombinant poxvirus described herein and an anti-PDl antibody (e.g., Pembrolizumab or Nivolumab).
- the kits described herein comprise a recombinant poxvirus described herein and an anti-PD-Ll antibody (e.g., Durvalumab).
- a method of inducing apoptosis of a cancer cell comprising contacting the cancer cell with the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL- 12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein, under conditions to induce apoptosis.
- IL- 12 interleukin- 12
- the cancer cell can include, but is not limited to: a renal cancer cell, prostate cancer cell, breast cancer cell, bladder cancer cell, colorectal cancer cell, lung cancer cell, liver cancer cell, gastric cancer cell, bile duct carcinoma cell, endometrial cancer cell, pancreatic cancer cell, ovarian cancer cell, head and neck cancer cell, melanoma cell, glioblastoma cell, multiple myeloma cell, or malignant glioma cell.
- a renal cancer cell prostate cancer cell, breast cancer cell, bladder cancer cell, colorectal cancer cell, lung cancer cell, liver cancer cell, gastric cancer cell, bile duct carcinoma cell, endometrial cancer cell, pancreatic cancer cell, ovarian cancer cell, head and neck cancer cell, melanoma cell, glioblastoma cell, multiple myeloma cell, or malignant glioma cell.
- Also provided herein is a method of inhibiting growth or promoting death of a cancer cell, the method comprising contacting the cancer cell with the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein, under conditions to inhibit growth or promote cancer cell death.
- IL-12 interleukin- 12
- the cancer cell can include, but is not limited to: a renal cancer cell, prostate cancer cell, breast cancer cell, bladder cancer cell, colorectal cancer cell, lung cancer cell, liver cancer cell, gastric cancer cell, bile duct carcinoma cell, endometrial cancer cell, pancreatic cancer cell, ovarian cancer cell, head and neck cancer cell, melanoma cell, glioblastoma cell, multiple myeloma cell, or malignant glioma cell.
- a renal cancer cell prostate cancer cell, breast cancer cell, bladder cancer cell, colorectal cancer cell, lung cancer cell, liver cancer cell, gastric cancer cell, bile duct carcinoma cell, endometrial cancer cell, pancreatic cancer cell, ovarian cancer cell, head and neck cancer cell, melanoma cell, glioblastoma cell, multiple myeloma cell, or malignant glioma cell.
- the method of inducing apoptosis of a cancer cell is performed in vitro. In some aspects of the present disclosure, the method of inhibiting growth or promoting death of a cancer cell, is performed in vitro. In some aspects of the present disclosure, the method of inducing apoptosis of a cancer cell is performed in vivo. In some aspects of the present disclosure, the method of inhibiting growth or promoting death of a cancer cell, is performed in vivo.
- Also provided herein is a method of treating cancer in a subject, the method comprising administering the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein, to the subject in an amount effective to treat cancer.
- IL- 12 interleukin- 12
- Also provided herein is a method of reducing an amount of cancer cells in a subject, the method comprising administering the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12), wherein the heterologous nucleic acid sequence encoding IL- 12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein, to the subject to reduce the amount of cancer cells in said subject.
- IL- 12 interleukin- 12
- Also provided herein is a method of eliciting an anti-cancer immune response in a subject, the method comprising contacting a cancer cell with the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein, in an amount effective to elicit the anti-cancer immune response.
- the anti-cancer immune response comprises activation of an innate or adaptive immune response against the cancer.
- the anti-cancer immune response comprises activation of an innate immune response against the cancer. In some aspects, the anti-cancer immune response comprises activation of an adaptive immune response against the cancer. In some aspects, the anticancer immune response comprises activation of an innate and an adaptive immune response against the cancer.
- the administering comprises systemic administration.
- the systemic administration is selected from subcutaneous, intramuscular, oral, intravenous, intranasal, transdermal, subcutaneous, and intramuscular administration.
- the recombinant poxvirus is administered two or more times.
- the administering comprises local administration.
- the local administration comprises intratumoral administration.
- the recombinant poxvirus is administered two or more times.
- the present disclosure provides a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a programmed death protein 1 (PD-1) inhibitor or a programmed cell death ligand 1 (PD-L1) inhibitor.
- a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a programmed death protein 1 (PD-1) inhibitor or a programmed cell death ligand 1 (PD-L1) inhibitor.
- the present disclosure provides a method of inhibiting the growth of a cancer in a subject, the method comprising administering to the subject an effective amount of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- the present disclosure provides a method of enhancing a tumor-specific immune response in a subject having cancer, the method comprising administering to the subject an effective amount of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) and (ii) a PD-1 inhibitor or a PD-L1 inhibitor.
- an immune checkpoint inhibitor in combination with the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) described herein, results in an enhanced therapeutic effect (e.g., a more significant reduction in tumor growth, an increase in tumor infiltration by lymphocytes, an increase in the length of progression free survival, etc.) than is observed after treatment with the recombinant poxvirus alone or after treatment with the immune checkpoint inhibitor alone.
- the enhanced therapeutic effect may, for instance, be an enhancement of the tumor-specific immune response. This may be characterized, for instance, by an increase in IFN-gamma production and/or an increase in the number of IFN-gamma secreting cells.
- IL- 12 interleukin- 12
- administration of the recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL- 12) described herein, or compositions thereof, to a subject suffering from a cancer that is resistant (or partially resistant) or insensitive (or partially insensitive) to treatment with one or more immune checkpoint inhibitors results in treatment of the cancer (e.g., reduction in tumor growth, an increase in the length of progression free survival, etc.).
- the cancer is resistant (or partially resistant) or insensitive (or partially insensitive) to treatment with a PD1 inhibitor.
- the PD-1 inhibitor or PD-L1 inhibitor is administered to the patient before administration of the recombinant poxvirus. In some aspects, the PD-1 inhibitor or PD-L1 inhibitor is administered to the patient at the same time as the recombinant poxvirus. In some aspects, the PD-1 inhibitor or PD-L1 inhibitor is administered to the patient after administration of the recombinant poxvirus.
- the disclosure provides a use of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein and (ii) a PD-1 inhibitor or PD-L1 inhibitor or a pharmaceutical composition comprising a PD-1 inhibitor or a PD-L1 inhibitor in the method of inducing apoptosis of a cancer cell described herein.
- IL-12 interleukin- 12
- the disclosure provides a use of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein and (ii) a PD-1 inhibitor or PD-L1 inhibitor or a pharmaceutical composition comprising a PD-1 inhibitor or a PD-L1 inhibitor in the method of inhibiting growth or promoting death of a cancer cell described herein.
- IL-12 interleukin- 12
- the disclosure provides a use of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein and (ii) a PD-1 inhibitor or PD-L1 inhibitor or a pharmaceutical composition comprising a PD-1 inhibitor or a PD-L1 inhibitor in the method of treating cancer in a subject described herein.
- IL-12 interleukin- 12
- the disclosure provides a use of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein and (ii) a PD-1 inhibitor or PD-L1 inhibitor or a pharmaceutical composition comprising a PD-1 inhibitor or a PD-L1 inhibitor in the method of reducing an amount of cancer cells in a subject described herein.
- IL-12 interleukin- 12
- the disclosure provides a use of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein and (ii) a PD-1 inhibitor or PD-L1 inhibitor or a pharmaceutical composition comprising a PD-1 inhibitor or a PD-L1 inhibitor in the method of reducing an amount of cancer cells in a subject described herein.
- IL-12 interleukin- 12
- the disclosure provides a use of (i) a recombinant poxvirus comprising in its genome a heterologous nucleic acid sequence encoding interleukin- 12 (IL-12), wherein the heterologous nucleic acid sequence encoding IL-12 is operably linked to a late promoter or intermediate promoter, or a pharmaceutical composition comprising the recombinant poxvirus described herein and (ii) a PD-1 inhibitor or PD-L1 inhibitor or a pharmaceutical composition comprising a PD-1 inhibitor or a PD-L1 inhibitor in the method of eliciting an anti-cancer immune response in a subject described herein.
- IL-12 interleukin- 12
- COPTG19673 and COPTG19674 vectors which are recombinant Vaccinia Viruses (VACV) expressing human IL-12 (hIL-12) under the control of two different promoters, pF17R and pA14L, respectively, is described below. Also described are the experiments and analysis related to the choice of the pF17R and pA14L promoters.
- VACV Vaccinia Viruses
- the COPTG19673 and COPTG19674 vectors encode human IL-12 (hIL-12) as a fusion of p40 and p35 subunits linked by a glycine-serine (GS)-linker.
- hIL-12 human IL-12
- GS glycine-serine
- the IL- 12 transgene was vectorized in a vaccinia virus Copenhagen strain having both the thymidine kinase gene (J2R) and the ribonucleotide reductase gene (I4E) deleted as in W02009/065546, which is herein incorporated by reference in its entirety. These two deletions restrict the replication of the virus into highly proliferative cells (containing high concentrations of nucleotides) such as tumor cells.
- J2R thymidine kinase gene
- I4E ribonucleotide reductase gene
- transgene expression which is directly dependent on the replication of virus genome, is thus limited to tumor cells (see, e.g., Foloppe et al., 2019, Mol Ther Oncolytics 14: 1-14; and Kleinpeter et al., 2016, Oncoimmunology 5:el220467).
- An expression cassette containing the promoter and the IL- 12 transgene was inserted in the J2R locus of the double deleted vaccinia virus Copenhagen strain.
- In vitro characterization of oncolytic vaccinia viruses COPTG19673 and COPTG19674 expressing interleukin- 12 is also described below.
- VVTG18058 empty VACV, VACV control, or unarmed control VACV is a
- VVTG18058 was used as an unarmed control virus. VVTG18058 was produced in chicken embryo fibroblasts (CEF). Titration was performed by plaque assay on Vero cells.
- COPTG19104 is a Vaccinia virus (Copenhagen strain) expressing the fluorescent protein mCherry under the control of the pH5R promoter at the J2R locus. It has the I4L gene deleted. It was used as the starting parental virus for generation of recombinant virus.
- VACVwt (also named COPwt) is a wild type Vaccinia virus (Copenhagen strain) with no deletion. VACVwt was produced in CEF.
- CEF Chicken embryo fibroblasts
- Vero cells The Cercopithecus aethiops (African green) kidney cell line Vero (ATCC® CCL-81TM) was grown in DMEM (Gibco) 4.5 g/1 glucose supplemented with 10 % FBS, 2 mM L-Glutamine and containing gentamicin at a final concentration of 40 mg/L. Growth conditions were at 37 °C - 5 % CO2.
- the human lung carcinoma cell line A549 (ATCC® CCL-185TM) was grown in DMEM (Gibco) 4.5 g/1 glucose supplemented with 10 % FBS, 2 mM L-Glutamine and containing gentamicin at a final concentration of 40 mg/L. Growth conditions were at 37 °C - 5 % CO2.
- the human cervix tumor cell line HeLa (ATCC® CCL-2TM) was grown at 37 °C, 5 % CO2 in DMEM (Gibco) supplemented with 10 % FBS, and 40 mg/L of gentamicin.
- the human pancreatic tumor cell line MIA PaCa-2 (ATCC® CCL-1420TM) was grown at 37 °C, 5 % CO2 in DMEM (ATCC®) supplemented with 10 % FBS and containing gentamicin at a final concentration of 40 mg/L.
- the human colorectal tumor cell line HCT116 (ATCC® CCL-247TM) was grown at 37 °C, 5 % CO2 in Me Coy’s 5A (ATCC®) supplemented with 10 % FBS and containing gentamicin at a final concentration of 40 mg/L.
- the human colorectal carcinoma cell line HT-29 (ATCC® HTB-38) was grown in McCoy’s 5A (Gibco) supplemented with 10 % FBS and containing gentamicin at a final concentration of 40 mg/L.
- HEK-BlueTM IL-12 Cells (InvivoGen, ref hkb-il 12) were grown in DMEM (Gibco) supplemented with 10 % inactivated FBS, 100 pg/ml NormocinTM (InvivoGen), HEK BlueTM Selection (Invivogen), and containing penicillin and streptomycin at final concentrations of 100 U/mL and 100 pg/mL, respectively.
- the natural killer cell line NK-92 (ATCC® CRL-2407TM) was grown in Alpha Minimum Essential medium (Gibco) with IX Glutamax (Gibco), 1.5 g/L sodium bicarbonate (Gibco), 0.2 mM inositol (Sigma), 0.1 mM Pmercaptoethanol (Sigma), 0.02 mM folic acid (Sigma), 150 U/ml recombinant IL-2, and 25 % fetal bovine serum.
- the continuous cell line of chicken embryo fibroblasts DF-1 (ATCC® CRL- 12203TM) was grown at 39 °C, 10 % CO2 in DMEM (Gibco) supplemented with 10 % FBS, 2 mM L-glutamine and 40 mg/L of gentamicin.
- PBMC peripheral blood mononuclear cells
- Escherichia Coli DH5a strain (Genotype: F- ⁇ b801acZAM I 5 A(lacZYA-argF) U169 recAl endAl hsdR17(rk-, mk+) phoA supE44 thi-1 gyrA96 relAl -; Invitrogen, 1826312) was used for cloning and plasmid amplification in LB media supplemented with 100 pg/mL ampicillin. DNA cloning
- Example 1 Comparative evaluation of vaccinia virus promoters
- Reporter transfer plasmids were designed to assess the strength of the different poxviral promoters to be evaluated.
- the reporter gene encoded for a fusion between the Renilla luciferase (RLuc) and the Aequorea green fluorescent protein (GFP) separated by a linker (Ala)5-Thr (GenBank: ABZ79968.1).
- the different poxviral promoters were inserted upstream of the RLuc-GFP fusion (SEQ ID NO: 1).
- the expression cassettes were inserted in a poxvirus transfer plasmid designed to allow insertion of the nucleotide sequence into the J2R locus of the vaccinia virus genome by homologous recombination. This plasmid originates from the plasmid pUC18 into which were cloned the flanking sequences Left Arm (L arm) and Right arm (R arm) surrounding the J2R locus.
- SEQ ID NO: 1 Nucleic acid sequence of RLuc/GFP ATGACAAGCAAGGTGTACGACCCCGAGCAGCGGAAGCGGATGATTACAGGACCT CAGTGGTGGGCCAGATGCAAGCAGATGAACGTGCTGGACAGCTTCATCAACTAC TACGACAGCGAGAAGCACGCCGAGAACGCCGTGATCTTCCTGCATGGAAATGCC GCCAGCAGCTACCTTTGGAGACACGTGGTGCCTCACATCGAGCCTGTGGCCAGGT GCATCATCCCTGACCTGATCGGCATGGGCAAGAGCGGCAAGTCTGGCAACGGCA GCTACAGACTGCTGGACCACTACAAGTACCTGACCGCTTGGTTTGAGCTGCTGAA CCTGCCTAAGAAGATCATCTTCGTCGGCCACGATTGGGGCGCCTGTCTGGCCTTT CACTACAGCTACGAGCACCAGGACAAGATCAAGGCCATCGTGCACGCCGAAAGC GTGGTGGATGATCGAGCTGGGACGAGTGGCCCGACATCGAG
- RLuc-GFP A synthetic fragment named “RLuc-GFP” containing the fusion gene coding for the RLuc-GFP protein was generated synthetically. It was inserted by homologous recombination in a set of transfer plasmids containing poxviral early/late promoter, restricted by vz/II, giving rise to the plasmids described in Table 4.
- FIG. 1 A schematic representation of the plasmid pTG19409 containing the RLuc-GFP encoding gene under expression control of the pH5R promoter is shown in FIG. 1.
- the sequence coding for the RLuc-GFP fusion was cloned just downstream of the promoters using their natural start codon. Synthetic fragments containing the late promoters and the beginning of the RLuc-GFP fusion were generated synthetically and were inserted in pTG19409 restricted by Smal- MscI by homologous recombination, giving rise to the plasmids described in Table 5.
- reporter transfer plasmids were first tested in transient infection/transfection expression studies to identify the best candidates. In a second phase some of them were used to generate recombinant poxvirus as described below.
- the reporter transfer plasmids using the RLuc-GFP reporter allows for assessment of cell infection and concomitant precise measurement of the expression level of the reporter.
- VACV promotors can be used according to their strength or their expression timing during poxvirus infection (early, intermediate or late).
- Some poxviral promoters like p7.5K or pH5R have both early and late elements, allowing the transgene to be expressed early after virus infection and late after the viral genome replication, respectively.
- the transgene expression could be driven by a late promoter that is active only after virus genome replication.
- pAlOL SEQ ID NO: 9
- pAl 1R SEQ ID NO: 10
- pA14L SEQ ID NO: 11
- pA26L SEQ ID NO: 12
- pF17R SEQ ID NO: 13
- pG7L SEQ ID NO: 14
- DF-1 cells were cultured in 24-well culture plates before being infected at MOI 1 by an empty vaccinia virus without any transgene (VVTG18058). After 2 hours, 0.5 ng of the different Renilla reporter plasmids (see Table 2), together with 0.5 ng of a control reporter plasmid encoding Firefly luciferase under the control of pl 1K7.5 and 250 ng of the control plasmid pTG15839 encoding GFP under the control of CMV promoter complexed with 0.625 pL of lipofectamine 2000 (Invitrogen) in opti-MEM culture medium were added to each well. Transfection were performed in triplicate.
- FIG. 2 The results obtained with the six late promoters are illustrated in FIG. 2.
- the ratios of Renilla luciferase/Firefly luciferase (R/F) are reported for each promoter, and the results of two independent experiments are shown.
- the highest level of expression was obtained with the pF17R promoter which was 3 to 4-fold stronger than the pA14L and pAlOL promoters.
- the pAl 1R, pA26L and pG7L promoters were 8 to 10-fold weaker than pF17R promoter.
- Transfer plasmids containing the fusion reporter gene RLuc-GFP under the control of various promoters were constructed.
- Four early/late promoters were tested: p7.5K (SEQ ID NO: 4), pH5R (SEQ ID NO: 2), pl 1K7.5 (SEQ ID NO: 3), and pSE/L (SEQ ID NO: 8); and three early promoters were also evaluated: pB2R (SEQ ID NO: 5), pA35R (SEQ ID NO: 7), and pCl 1R (SEQ ID NO: 6).
- the three late promoters previously tested (pF17R (SEQ ID NO: 13), pA14L (SEQ ID NO: 11), and pA26L (SEQ ID NO: 12) in transient infection-transfection were also tested in a recombinant VACV. See Table 4 and Table 5.
- VACV-RLuc-GFP vectors were generated by homologous recombination in CEF by insertion of the RLuc-GFP expression cassette in the J2R locus of the double deleted vaccinia virus Copenhagen strain under the transcriptional control of different poxviral promoters as described herein. All of these viruses are both defective for thymidine kinase (TK, J2R locus) and ribonucleotide reductase (RR, I4L locus) activities.
- TK thymidine kinase
- RR ribonucleotide reductase
- the recombinant vaccinia viruses were generated by homologous recombination in CEF using COPTG19104 as a starting parental virus, and the transfer plasmid containing the expression cassette to be integrated with flanking sequences (L arm and R arm) surrounding the J2R locus.
- the homologous recombination between the transfer plasmid and parental vaccinia virus (strain Copenhagen) enables the generation of recombinant vaccinia viruses which have lost the mCherry expression cassette and gained the expression cassette resulting in white (non-fluore scent) plaques. More specifically, a F175 flask of CEFs was infected at MOI 0.05 with COPTG19104 for 1 hour at room temperature.
- the viral suspension was then discarded, and the infected cells were incubated for 2 hours in MBE + 5 % FBS at 37 °C + 5 % CO2 before being trypsinated and counted.
- Ten million infected cells were then transfected with 2 pg of I-Scel- restricted transfer plasmid by nucleofection.
- the transfected cells were then transferred into a well of a 6-well plate incubated at 37 °C for 48 h before being frozen. After sonication, serial dilutions of the transfer mixture were used to infect CEF for selection of recombinant virus. Non-fluorescent white plaques were picked and used for a second round of plaque purification.
- Selected non-fluorescent white plaques were picked and amplified in a 6-well plate at 37 °C, 5 % CO2 for 72 h. Amplifications were used for PCR analysis, followed by the selection of the recombinant vaccinia virus.
- the recombinant vaccinia viruses are referenced as following for identification: COPTG19409 (VACV comprising promoter pH5R), COPTG19410 (VACV comprising promoter pl 1K7.5), COPTG19411 (VACV comprising promoter p7.5K), COPTG19412 (VACV comprising promoter pB2R), COPTG19415 (VACV comprising promoter pA14L), COPTG19416 (VACV comprising promoter pA26L), COPTG19417 (VACV comprising promoter pF17R), COPTG19431 (VACV comprising promoter pCl 1R), COPTG19436 (VACV comprising promoter pA35R), and COPTG19437 (VACV comprising promoter pSE/L). Comparative evaluation of vaccinia virus promoters in human tumoral cell lines and in human PBMC
- Human tumoral cells line HeLa, MIA PaCa-2, and HCT116 were infected with the ten recombinant vaccinia viruses previously obtained at a MOI of 0.1 or 1 in 96-well plates as described in the following protocol. After 6 hours and 24 hours, cells were harvested for quantification of luciferase expression and for detection of GFP-expressing cells.
- FACS Fluorescence-activated Cell Sorting
- Renilla luciferase was measured after 6 and 24 hours, and the results obtained in MIA PaCa-2 cells are illustrated in FIG. 3 A and FIG. 3B, respectively.
- the expression was normalized relative to the weakest promoter, pA26L.
- the late promoters pF17R, pA14L, and pA26L gave rise to very low levels of expression.
- the highest level of expression was obtained with the early/late promoter pH5R, followed by the early promoters pB2R and pCl IR and the early/late promoters pSE/L and pl 1K7.5.
- the results obtained in the three cell lines allowed classification of the promoters into three strength groups.
- the weak promoters include the three early promoters (pB2R, pA35R, and pCl 1R) and the late promoter A26L.
- the moderate promoters were 2 to 5- fold more efficient than the weak ones. They correspond to the early/late promoters p7.5K and pH5R and the late pA14L promoter.
- the strongest promoters were 2 to 3-fold more potent than the moderate ones. They correspond to the early/late promoters pl 1K7.5 and pSE/L and the late pF17R promoter.
- Human PBMC were then infected with the ten recombinant vaccinia viruses at a MOI of 1 in 96-well plates. After 6 hours and 24 hours, cells were harvested for quantification of luciferase expression and for detection of GFP-expressing cells.
- the results of flow cytometry analysis are shown in FIG. 6. After 6 hours, about 12 % of cells were detected as GFP-positive cells after infection with the viruses containing early or early/late promoters. In contrast, a very low quantity of cells was detected as GFP-positive after infection with the viruses containing late promoters (less than 4 % for pA26L and less than 1 % for pF17R and pA26L). The percentage of GFP- positive cells decreased at 24 h post infection due to the death of infected cells and absence of replication of the recombinant vaccinia viruses. The percentage of infected cells was then around 5 % for the viruses containing the early and early/late promoters and was negligible for the viruses containing the late promoters.
- Renilla luciferase was measured after 6 and 24 hours, and the results are shown in FIG. 7. The expression was normalized relative to the weakest promoter, pA26L. The levels of luciferase were lower than the level detected after infection of human tumoral cells (about 100-fold lower). Moreover, the expression decreased between 6 and 24 hours due to death of infected cells. Cells infected with the viruses containing the late promoters (pF17R, pA14L, and pA26L) expressed very low levels of luciferase, about 100 to 300- fold less than the expression detected with the early or the early/late promoters.
- the vaccinia virus used in this study was derived from the Copenhagen strain and has both the thymidine kinase gene (J2R) and the ribonucleotide reductase gene (I4E) deleted. These two deletions restrict the replication of the virus into the highly proliferative cells (containing high concentrations of nucleotides) such as tumor cells. These viruses are not able to replicate efficiently in primary human PBMC and, therefore, the late promoters are not active in these cells.
- Example 2 Generation and production of recombinant vaccinia virus encoding for IL-12 by homologous recombination
- the plasmids pTG19673 and pTG19674 contain the human IL-12 gene under the control of the pF17R and pA14L promoters, respectively.
- Endogenous human IL-12 is unique amongst cytokines in being a disulfide-linked heterodimer of two separately encoded subunits (p35 and p40).
- a single chain IL-12 protein was expressed from vaccinia constructs in which the full length p40 subunit was fused, via a G6S linker, to the p35 subunit truncated of its leader sequence (i.e., IL- 12. p40. delta p35) (see Lieschke et al., 1997, Nat Biotechnol. 1997 Jan;15(l):35-40).
- the primary protein structure of the hIL-12 fusion protein contains IL- 12 p40 linked to IL- 12 p35 by a 7 amino acid polypeptide linker, as shown in the sequence (SEQ ID NO: 15) below.
- Fusion IL-12.p40.delta p35 (SEQ ID NO: 15):
- the mature IL-12 fusion protein comprises amino acids 23-532 of SEQ ID NO: 15 as shown below in SEQ ID NO:20.
- SEQ ID NO: 21 is provided below.
- the vaccinia virus transfer plasmids pTG19535 and pTG19537 were designed to allow insertion of a nucleotide sequence into the J2R locus of the vaccinia virus genome by homologous recombination. They originate from the plasmid pUC18, into which the flanking sequences (L arm and R arm) were cloned on either side of the J2R locus.
- the plasmid pTG19535 contains the pF17R promoter, while pTG19537 contains the pA14L promoter.
- a fragment containing the IL- 12 fusion was generated synthetically and inserted in a plasmid by Geneart.
- the corresponding plasmid was restricted by SriaBL and the resulting fragment “hIL-12” was inserted by homologous recombination in pTG19535 (pF17R promoter) or pTG19537 (pA14L promoter) restricted with vz/II, giving rise to pTG19673 (FIG. 9) or pTG19674 (FIG. 10), respectively.
- the expression cassette was inserted between recombination arms, allowing homologous recombination in the J2R locus of vaccinia virus genome. Maxipreparations of both plasmids were produced and analyzed by sequencing the recombination arms, and the expression cassette inserted between them. Alignment of analyzed and theoretical sequences showed 100 % homology for both plasmids.
- sequences of these two promoters, pA14L and pF17R, with the downstream ATA are as follows: pA14L (SEQ ID NO: 22) TTTGTTCATTCGGCGATTTAAAATTTTTATTAGTTAAATA pF 17R (SEQ ID NO: 23) AAAATATAGTAGAATTTCATTTTGTTTTTTTCTATGCTATAAATA Generation of recombinant vaccinia virus by homologous recombination
- COPTG19673 and COPTG19674 were generated by homologous recombination in CEF using COPTG19104 as starting parental virus and the transfer plasmids pTG19673 and pTG19674, encoding the hIL-12 under the control of pF17R and pA14L, respectively.
- COPTG19104 contains the expression cassette of the mCherry in its J2R locus.
- the homologous recombination between the transfer plasmid and parental vaccinia virus enables the generation of recombinant vaccinia viruses (COPTG19673 and COPTG19674) which have lost the mCherry expression cassette and gained the hIL-12 expression cassette (see Example 1 for the method of generation of recombinant vaccinia virus).
- COPTG19673 primary stock Primary research stock of recombinant virus COPTG19673, obtained from homologous recombination between the transfer plasmid pTG19673 and the parental COPTG19104 is hereafter referred as COPTG19673 primary stock.
- COPTG19674 primary stock Primary research stock of recombinant virus COPTG19674, obtained from homologous recombination between the transfer plasmid pTG19674 and the parental COPTG19104 is thereafter referred as COPTG19674 primary stock.
- the expression of IL-12 in supernatants of A549 cells infected with COPTG19673 and COPTG19674 as measured by ELISA is shown in FIG. 11.
- Example 3 In vitro characterization of recombinant oncolytic vaccinia viruses COPTG19673 and COPTG19674 expressing interleukin-12
- VACVwt (also named COPwt) is a wild type Vaccinia virus (Copenhagen strain) with no deletion. VACVwt was produced in CEF. The infection titer determinations in the following assays were performed on plaque essay on Vero cells.
- the replication of COPTG19673 and COPTG19674 was assessed in three tumoral human cell lines (A549, HT-29, and MIA PaCa-2) and compared to the ones of unarmed control VACV VVTG18058 as the benchmark.
- the A549, HT-29, and MIA PaCa-2 tumor cell lines were infected by each virus at MOI 10' 3 . Infected cells were then plated in 6-well plates and incubated at 37 °C in 5% CO2 atmosphere for 24, 48, and 72 hours. The quantity of virus produced in each cell line was determined at each timepoint by plaque assay on Vero cells.
- FIGs. 12A-C show that the replication of COPTG19673, COPTG19674, and VVTG18058 was similar in the three human tumor cell lines.
- COPTG19673 and COPTG19674 were assessed in CEF and in a human cell line (HeLa) (as cells for virus production) and compared to the ones of unarmed control VACV as the benchmark. Seeded HeLa or CEF were infected, in 6-well plates, by each virus at MOI 0.05. Plates were then incubated for 72 hours at 37 °C in CO2 atmosphere. The production of virus, in each cell type, was determined by titration on Vero cells using plaque assay method. FIG. 13 shows that the replication of COPTG19673, COPTG19674, and VVTG18058 are similar (i.e., less than 0.7 log difference at 72 h) in each of the two cells (CEF and HeLa).
- the oncolytic activity of COPTG19673 and COPTG19674 was assessed on three human tumor cell lines (A549, HT-29, and MIA PaCa-2) and compared to the activities of unarmed control VACV (VVTG18058). Oncolytic activity was assessed by quantification of cell viability after 5 days of incubation.
- Tumor cell lines were infected by each virus at 10 different MOI according to the cell lines used (i.e., A549 and HT-29: 3.10' 5 to 1, MIA PaCa-2: 10' 5 to 3.10' 1 ). The infected tumor cells were then incubated in 96-well plates for 5 days at 37 °C in a CO2 atmosphere. Cell viability was determined using cell titer blue cell viability assay according to the protocol provided by the manufacturer.
- Oncolytic activity is representative of the lytic activity of the tested viral samples on tumor cells. Oncolytic activity of each sample is expressed as a percentage of the mock-infected cell viability.
- FIGs. 14A-C show the oncolytic activities of COPTG19673, COPTG19674, and VVTG18058 at different MOI. From these results, the EC50 values (MOI at which 50 % of cells are killed) were calculated for each virus in each cell line. Comparison of EC50 shows that the oncolytic activity of COPTG19673, COPTG19674, and unarmed control VACV are very similar in MIA PaCa-2.
- COPTG19673 showed higher EC50 than the VVTG18058, but at high MOI, both VACV displayed comparable strong oncolytic activity, with less than 20% residual living cells.
- EC50 for both viruses were calculated using GraphPad Prism. As shown,
- EC50 of COPTG19673 is 4.8.10' 3
- EC50 of COPTG19674 is 2.7.10' 3
- EC50 of VVTG18058 is 4.4.10' 3
- EC50 of COPTG19673 is 1.2. IO' 2 ; EC50 of COPTG19674 is 7.6.10' 3 ; EC50 of VVTG18058 is 4.9.10' 3
- vIL-12 The levels of expression of cytokine vhIL-12 were measured in supernatants of 3 infected tumor cell lines after 3 days of infection at MOI 0.01.
- vIL-12 and vhIL-12 refer to IL-12 as expressed by the VACV according to the present disclosure.
- the starting materials for measuring the expression level of vIL-12 were the supernatants recovered from MIA PaCa-2, A549, and HT-29 cells.
- the tumor cell lines were seeded in 6-well plates, infected at MOI 10' 2 , and incubated for 72 hours in 3 mL of adequate medium without fetal bovine serum. The supernatants were collected and then filtered to remove viruses.
- IL-12 concentrations in supernatant of infected tumor cell lines were determined using the DuoSet®ELISA development system Human IL- 12 (R&D Systems ref DY1270-05).
- the three tumor cell lines A549, HT-29, and MIA PaCa-2 were infected at MOI 0.01 by VVTG18058, COPTG19673, or COPTG19674 for 72 hours.
- IL-12 concentration was then measured in supernatants of infected cells by ELISA.
- the supernatant of VVTG18058-infected cells were used as negative controls. The results are shown in FIG. 15, as the means and standard deviations (SD) of duplicate measurements on three samples.
- high levels of expressions of vIL-12 were obtained with concentrations ranging from 0.3 to 8.3 pg/mL.
- IL- 12 produced by the viruses during the transgene expression assays in the “Supernatant preparation” part of this example was evaluated using HEK-BlueTM IL-12 reporter Cells and by a cell proliferation assay using NK-92 cells.
- vhIL-12 produced in the supernatant of human tumor cell lines was assayed using HEK-BlueTM IL- 12 reporter cell.
- HEK-BlueTM IL-12 cells are designed to detect bioactive human and mouse IL-12 by monitoring the activation of the STAT -4 pathway. Binding of IL-12 to the IL-12 receptor on the surface of HEK-BlueTM IL-12 cells triggers a signaling cascade leading to the activation STAT-4 with the subsequent production of secreted alkaline phosphatase (SEAP) marker protein. Detection of SEAP in the supernatant of HEK-BlueTM IL-12 cells can be readily assessed using QUANTLBlueTM.
- SEAP secreted alkaline phosphatase
- HEK-BlueTM IL-12 cells were plated at 5E+04 cells/well into a 96-well flat-bottomed microtiter. Different dilutions of the culture supernatants and rhIL-12 standard (concentration ranging from 10 pg/mL to 100 ng/mL) were added to a 96-well cell culture microplate. The plate was incubated at 37 °C. After 24 hours of incubation, the detection of SEAP in the supernatant of HEK-BlueTM IL-12 cells was determined using QUANTLBlueTM (Invivogen, ref rep-qbs) according to the provider protocol.
- QUANTLBlueTM Invivogen, ref rep-qbs
- vIL-12 produced from the two VACV-IL12, activated the HEK-Blue IL- 12 cell line with better EC50 than rhIL-12. These results demonstrated that IL-12 produced by VACV-infected tumor cells retains its cytokine activity. vIL-12 biological activity on NK-92 cells
- vIL-12 The biological activity of vIL-12 produced in the supernatants of infected human tumor cell lines was assayed using a cytokine dependent cell proliferation assay of the NK-92 cell line. Proliferation of NK-92 cells can be induced by IL-12. The biological activity of vhIL-12 generated by 3 human tumor cell lines infected by COPTG19673 and COPTG19674 was measured and compared to the biological activity of human recombinant hIL-12.
- NK-92 is an interleukin-2 dependent Natural Killer Cell line derived from peripheral blood mononuclear cells. The NK-92 cells also depend on IL-12 to proliferate and can then be used for controlling IL-12 functionality.
- NK-92 cells were plated at 1E+04 cells/well into a 96-well flat-bottomed microtiter. Different dilutions of the culture supernatants and rhIL-12 standard (concentration ranging from 1 pg/mL to 100 ng/mL) were added to a 96 wells cell culture microplate. The plate was incubated at 37 °C. After 48 h of incubation, the CellTiter- Glo® was added, according to the provider protocol. Supernatants of Mock infected cells and unarmed control VACV infected cells were used as negative control.
- FIGs. 17A-E show that all of the 3 different samples tested for both viruses, COPTG19673 and COPTG19674, contained biologically active vhIL-12.
- SN from mock-infected cells and SN from empty VACV-infected cell lines were added as negative controls and did not stimulate proliferation of NK-92.
- the comparison of vhIL-12 biological activity with rhIL-12 was carried out after dosing all samples with the same ELISA.
- the vhIL-12 from the three infected tumor cell lines by both viruses and rhIL-12 induced similar level of proliferation of NK-92.
- the EC50 values were lower for vIL-12 than for rhIL-12.
- the hepatocytes were provided by Biopredic in 6-well plates. Hepatocytes were infected at MOI 10' 3 by each virus and were incubated for 72 hours at 37 °C in CO2 atmosphere. The quantity of virus produced in 72 hours of incubation was determined by virus titration per well by plaque assay in Vero cells. Results are represented as replication yield corresponding to ratio between input/output virus quantities. Results are mean of three wells.
- Human PBMC were chosen as second normal primary cells to evaluate the safety profile of newly generated VACV. Wild type VACV or unarmed VACV generally do not replicate in hPBMC. The presence of cytokines or immunostimulatory molecules expressed by the VACV could activate or stimulate immune cells and consequently modify the replication profile of VACV in these cells. Therefore, the replication of COPTG19673 and COPTG19674 in these cells (i.e., hPBMC) was evaluated and compared to the benchmark VACVwt and VVTG18058. The virus production was measured by plaque assay in Vero cells after infection at MOI 1 and 3 days of incubation.
- the viral replication yield was determined as the ratio between the total infectious particles detected after 72 h of infection (output) and the viral particles used for PBMC infection (input).
- FIG. 19 shows that the two VACV express IL-12, COPTG19673 and COPTG19674, as well as VACVwt and unarmed control VACV, did not replicate in hPBMC. In other words, the vectorization of human IL- 12 did not modify the replication behavior of the VACV on the hPBMC. Additionally FIGS. 27A-C show that COPTG19673 amplification was not observed in human PBMCs, and minimal replication was observed in normal human hepatocytes and dermal fibroblasts, indicating that replication is relatively specific to tumor cells.
- Example 4 Potency of COPTG19673 (“VACV IL-12”) across human tumor cell lines
- VACV IL- 12 The VACV expressing IL- 12 COPTG19673 (referred to below as “VACV IL- 12”) was further evaluated.
- the tumor type from which each cell line was derived is indicated below the x-axis.
- the dashed line is arbitrarily set at an MOI of 0.1 PFU, which represents an amount of virus that infects and lyses one in ten cynomolgus monkey Vero cells in the plaque forming assay used to measure PFU.
- Oncolysis mediated by VACV IL-12 was broadly observed across tumor cell lines of differing cancer types, with 27/30 tumor cell lines showing mean EC50 values of 0.1 MOI or lower. Transgene production and virus replication were also evaluated in the tumor cell lines 5 days post infection (MOI 0.004) (FIGS. 29D-E).
- Example 5 Amount of VACV IL12 virus and human IL-12 recovered from human bladder tumors after IV administration to tumor-bearing mice
- Tumors originating from SW780 human bladder cancer cells were engrafted on the flank of immuno-deficient NOD/SCID mice. After administration of a single dose of 10 5 , 10 6 , or 10 7 PFU of VACV IL- 12 by the intravenous route, tumors were removed and analyzed for infiltration of virus by plaque formation assay (PFU of virus per gram of tumor tissue) and the IL-12 transgene produced by virally-infected cells (ng of IL-12 per gram of tumor) using a human IL12-specific ELISA (FIG. 21 A).
- the X-axis indicates time points after viral administration. Asterisks show time points for which no determination of virus and transgene was made.
- mice were treated with a single intravenous dose of VACV-LUC or VACV IL-12 (10 5 , 10 6 and 10 7 PFU) (FIGS. 30A-C).
- VACV-LUC or VACV IL-12 10 5 , 10 6 and 10 7 PFU
- Significant tumor control was observed from administration of a single dose of 10 5 PFU in the NCLH292 and HCT-116 models (p-value ⁇ 0.001).
- Significant tumor control was observed in the SW780 model following a single dose of VACV IL-12 at 10 6 PFU (p-value ⁇ 0.001). Due to the lack of an intact immune system, VACV-LUC and VACV IL-12 demonstrated similar tumor control, indicating that oncolytic activity of VACV is a key contributor of tumor control.
- VACV encoding murine IL-12 was assessed after multi-dose, intra-tumoral administration to C57BL/6 mice engrafted with subcutaneous MC38 colorectal tumors (FIG. 22A).
- Murine IL-12 was chosen as the cytokine encoded by VACV IL- 12 in these experiments because human IL- 12 does not bind to the mouse IL- 12 receptor.
- Tumors were randomized to treatment groups after growing to a median size of approximately 80 mm 3 . A total of 5 doses of le 7 PFU of virus were administered twice weekly over a 14-day period. Blood was sampled at 4 and 24 hours after administration of the first dose, and tumor volumes were recorded thereafter.
- FIGS. 22B-D Tumor growth was assessed among vehicle treated controls, mice administered a VACV containing no transgene (empty VACV), and mice administered VACV encoding muIL-12 (VACV muIL-12).
- Example 7 Expression of murine IL-12 and induction of pro-inflammatory cytokines by VACV muIL-12
- Example 8 Activity of VACV luc in primary human patient-derived tumor xenografts
- Anti -tumor activity of luciferase-expressing VACV was determined using primary tumors from 47 patients with cancer (Table 6) (PDX: patient derived xenografts) engrafted into immunocompromised NOD/SCID mice (FIGS. 24A-H). Each tumor tested was either left untreated (open circles, gray line) or was administered IxlO 7 pfu VACV luc virus (closed square, black line) by the intravenous (IV) route every week (protocol shown in FIG. 28A) for three total doses starting when tumors reached a mean volume of approximately 200 mm 3 .
- Each primary patient-derived tumor included only one untreated and one treated tumor, so each open circle and closed square represent a pair but are graphed together within a tumor type for simplicity.
- Response was determined according to tumor volume relative to untreated controls (complete responders (CR): no measurable tumor; partial responders (PR): >30% decrease in tumor volume; stable disease (DS): less than 100% tumor increase; and progressive disease (PD): progressive growth).
- CR complete responders
- PR partial responders
- DS stable disease
- PD progressive disease
- VACV Luc virus treated animals saw anti-tumor activity relative to untreated controls across tumor types. (FIG. 28B and Table 6).
- Example 9 IL-12R, NK cells, PDL1, CXCL9 and 10 are upregulated upon VACV infection
- Total RNA was isolated from fresh frozen PDX tissue and depleted of ribosomal RNA and globin transcripts. Stranded RNAseq libraries were created, and paired end sequencing was performed.
- Paired-end reads were aligned to mouse genome reference build mm 10 using STAR aligner, and gene-level read counts were generated using Salmon. The read counts were further normalized for sequencing depth and gene length using tximport to generate transcripts per kilobase million (TPM) values for plotting. All plots were generated using R version 4.1. These results demonstrate that IL-12R, NK cells, PDL1, and CXCL9/10 are upregulated upon VACV infection, thereby priming the tumor microenvironment (TME) to elicit potent anti-tumor immunity.
- TPM kilobase million
- Example 10 VACV-IL-12 efficiently infects human tumors in vitro, resulting in IL-12 and early B8R dependent IFN-y blockage.
- VACV-IL12 infection in human tumors in vitro was investigated.
- the potency of VACV IL-12 was assessed across tumor tissue slice cultures (TSCs) derived from melanoma, bladder, colorectal, lung, and ovarian cancer, and dissociated human tumor cells (DTCs) derived from melanoma, head and neck squamous cell, and ovarian cancer patients.
- TSCs tumor tissue slice cultures
- DTCs dissociated human tumor cells derived from melanoma, head and neck squamous cell, and ovarian cancer patients.
- DTCs from different tumor types were thawed in culture media supplemented with IX CTL anti -aggregate wash.
- IL-12 i.e., IL12p70
- concentrations were measured using a custom U-Plex human IL-12p70 assay or U-PLEX mouse IL-12p70 assay (Catalog # K151UAK; K152UAK; Meso Scale Diagnostics), respectively, according to the manufacture’s protocol.
- IL-12 human IL-12 was detected in supernatant from tumor tissue slice culture (TSC) 3 days after infection with VACV IL-12 but not after infection with VACV-GFP or in mock infected tissues (FIG. 26A) and the same was true of DTCs (FIG. 26B).
- TSC tumor tissue slice culture
- FIG. 26A Increased levels of IFNy mRNA were detected in VACV IL-12 treated tissue slice cultures compared to slices treated with VACV-GFP or mock infected, although no difference in IFNy protein expression was detected in the supernatant from the same tissues (FIGs. 26C-D).
- Vaccinia virus encodes several immunomodulatory genes that suppress immune cell activation, such as B8R, which sequesters and neutralizes IFNy.
- B8R immunomodulatory genes that suppress immune cell activation
- VACV-IL-12 infection of TSC resulted in expression of B8R, which has been shown to sequester IFNy in the supernatant.
- Significantly higher B8R levels were detected in TSCs infected with either VACV-GFP or VACV-IL-12 compared to mock infected tissues.
- FIG. 26E each dot is a single slice with 1-4 replicates per condition. Data was analyzed using GraphPad Prism software version 9.0.0.
- a schematic of VACV infection, transgene production, and tumor cell lysis is shown in FIG. 26F.
- VACV IL-12 (COPTG1673) infection resulted in production of IL-12p70 in 19/19 of DTC samples tested, (17984 pg/mL ⁇ SD 49523).
- IL-12 concentrations in the supernatants from the mock-infected (6.15 pg/mL ⁇ SD 9.13) or VACV GFP-treated (4.99 pg/mL ⁇ SD 6.36) (FIGS. 31A-D) DTCs was negligible.
- Example 11 VACV encoding murine IL-12 in an in vivo rat model overcomes the inhibitory effects of B8R, resulting in IFN-y induction
- VACV encoded B8R does not bind murine IFNy; however, B8R binds and neutralizes fFNy from human and rats.
- a competitive ELISA was performed. Recombinant B8R protein or filtered supernatant from VACV infected HeLa cells (MOI 1) were incubated with recombinant IFNy for 1 h at 37 ° C. Next, an ELISA was performed to measure the available rat fFNy.
- VACV-muIL-12 (COPTG1673)
- a syngeneic rat tumor model was utilized to evaluate VACV IL-12 (COPTG1673) in an immunocompetent model.
- the surrogate VACV-muIL12 virus was used because muIL12 has been shown to cross-react in rats.
- VACV-muIL12 effectively replicates and produces muIL-12 transgene in rat tumor cells, as compared to murine tumor cells (Fig 32B-C).
- VACV-muIL12 mediates potent oncolysis of rat tumor cell lines whereas cell killing activity in mouse tumor cell lines is less (Fig 32D-F).
- Rats were implanted with F98 tumor cells and treated with VACV-LUC (1X10 7 PFU) or VACV-muIL12 (IxlO 5 , IxlO 6 or IxlO 7 ) on days 0, 4, and 7 by intravenous administration. Cytokine analysis in the plasma confirmed production of IL12p70, with a modest increase between rats treated with IxlO 5 and IxlO 6 (FIG 32G). Additionally, IFN-y could be detected in the plasma of VACV-muIL12 treated rats, but not the VACV-LUC control groups (Fig 32H). These results indicate that IL 12 encoded by VACV can induce peripheral IFNy production, which is not inhibited by B8R.
- Recombinant rat IFN-y (Ing/ml) (R&D) was incubated with PBS, recombinant B8R (50 ng/ml) (Vendor) or 0.2 um filtered supernatant from HeLa cells infected with VACV IL-12 (MOI 1). The mixes were incubated at 37° C for 1 hour. Since B8R binds IFN-y and prevents antibodies from binding and detection by ELISA, an ELISA was performed to determine the percent recovery of IFN- y, as described below. Percent recovery was calculated relative to IFN-y alone. ELISAs
- Rat interferon-y was measured in the plasma of rats treated with VACV at the indicated time points. Plasma was diluted and measured using the Rat IFN- y DuoSet Elisa, according to the manufacture’s protocol (R&D systems). This demonstrates that wherein the recombinant poxvirus is capable of upregulating interferon (IFN)-y.
- IFN interferon
- Example 12 VACV Expressing IL12 Improves Therapeutic Efficacy In Murine Syngeneic Tumor Models
- VACV- muIL12 demonstrates similar replication, oncolysis and transgene bioactivity as VACV IL-12 in human and mouse tumor cell lines (FIGS. 33A-E and 34A-F).
- Mice bearing subcutaneous CT26 tumors were treated with 5 intratumoral (i.t.) doses (10 7 PFU) of either the vehicle control, VACV-LUC or the surrogate VACV-muIL12 (FIGS. 35A-D).
- Treatment with VACV-LUC did not result in control of the CT26 tumor (0/10 complete responders) and was similar to the vehicle group (FIGS. 35B-D).
- treatment with VACV-muIL12 resulted in regression of the CT26 tumors, with 6/10 complete responders (FIG. 35E).
- VACV-LUC and VACV-muIL12 demonstrated similar oncolytic activity and replication in murine tumor cells (FIG 34A-F). Therefore, it was hypothesized that the observed difference in tumor control could be attributed to the immune-modulating effects of muIL12. Accordingly, animals were treated according to the protocol shown in FIG. 35 A. Evidence of IL 12 production in the sera was detected as early as 4 hours post injection (FIG 35F). Induction of IFNy is an important feature of IL-12 signaling and was detected in VACV-muIL12 treated mice at 4 and 24 hours post injection (FIG 35G). Similar results were observed in the MC38 syngeneic tumor model (FIGS. 22A-E, 23 A- B, 23E). Overall, these results suggest that VACV encoding IL- 12 engages the adaptive immune response to control tumor growth, while the oncolytic activity of VACV-LUC alone is not sufficient to control the tumor burden.
- Example 13 Methods for In Vivo and Ex Vivo Studies
- Cell line derived xenografts were established by subcutaneous (sc) injection of 5 * 10 6 cells/200 pL suspended in PBS into the right flank of 8- to 12-week-old animals. Tumors reached 150-250 mm 3 before randomization. Body weight of animals was measured 2 times per week for 4 weeks, followed by once per week and any weight loss was monitored for welfare purposes.
- cryogenic vials containing tumor cells were thawed and prepared for injection into mice. Thawed cells were washed in RPMI media, counted, and resuspended in cold RPMI at a concentration of 50,000-100,000 viable cells/50 pL. Cell suspensions were mixed with an equal volume of CULTUREXTM Extracellular matrix (ECM) and kept on ice during transport to the vivarium. Cells were prepared for injection by withdrawing ECM-Cell mixture into a chilled 1 ml slip-tip syringe. The filled syringes were kept on ice to avoid the solidification of ECM. Animals were shaved prior to injection.
- CCM Extracellular matrix
- tumor chunk inoculation frozen tumor fragments were thawed and cut into tumor chunks that were approximately 2-3 mm in diameter. Each mouse received a single injection of buprenorphine 30 minutes before tumor chunk implantation. One tumor chunk was loaded into a trocar needle and injected into the right front flank of mice for tumor development. Mice were ear tagged and animals left undisturbed for up to seven days before observing for tumor growth. Tumors were allowed to reach approximately 130-230 mm 3 before assignment to treatment groups. Anticancer effects of VACV Luc were determined by comparison of the growth curves for each treated tumor with that of a size-matched, untreated tumor.
- CR complete response
- PR partial response
- DS stable disease
- PD progressive disease
- F98 tumors were removed from 10 stacks flasks using Trypsin solution and neutralized trypsin by adding DMEM + 10% FBS. Harvested cells were kept on ice from the time of harvest to implantation (not exceeding 3 hrs). F98 cell line was grafted by subcutaneous (SC) injection of 5.0 x io 6 cells suspended in 0.2 mL of PBS into the right flanks of 7 to 9-week-old animals. Tumors were allowed to reach -300-350 mm 3 before randomization.
- SC subcutaneous
- mice were euthanized and blood was collected and tumors and normal tissues were resected and snap-frozen. Frozen tumors and normal tissues were weighed and suspended in ice-cold homogenization buffer (PBS supplemented with lx antibiotic-antimycotic and lx HALTTM protease-phosphatase inhibitor). The tissues with homogenization buffer were transferred to Matrix A tubes and homogenized using the Fast-prep-24 lysis system for 20 seconds at 4 m/second. Tissue homogenates were then subjected to two freeze-thaw cycles, aliquoted and stored at -80° C. Two additional aliquots that were not subjected to freeze thaws were prepared for DNA isolation or cytokine analysis by MSD. Cytokine analysis
- Example 14 Combination therapy with VACV-muIL12 and immune checkpoint inhibitors
- VACV-muIL12 results in significant tumor control.
- mice were implanted with CT26 tumor cells and treated with vehicle, VACV-LUC, or VACV-muIL12.
- vehicle VACV-LUC
- VACV-muIL12 Each treatment group also received a vehicle, an isotype control, or a commercially-available anti-muPD-Ll blocking antibody that prevents the binding of mouse PD-L1 to PD-1. Because the tumors are well controlled following VACV-muIL12 therapy, a significant enhancement in tumor control when delivered in combination with anti-PD-Ll antibodies was not observed (FIGS.
- Anti-tumor immune responses were further characterized by ex vivo splenocyte restimulation assays. Splenocytes were collected on day 7 post-VACV dosing and stimulated with either a tumor associated peptide antigen (AH-1) or a VACV specific peptide (A52L). Combining VACV-muIL12 with anti-PD-Ll resulted in a significant increase in the number of IFN-gamma secreting cells upon AH-1 peptide stimulation (p- value 0.041) (FIG. 37A).
- VACV-LUC alone or in combination did not result in a significant increase in IFN-gamma secreting cells relative to the other treatment groups. Additionally, the VACV-specific immune response was not impacted by IL-12 or anti-PD-Ll (FIG. 37B).
- This example demonstrates that VACV-muIL12 enhances the tumor-specific immune response when combined with a PD-L1 inhibitor in a tumor model resistant to anti-PD-Ll therapy.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Engineering & Computer Science (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Zoology (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Virology (AREA)
- Gastroenterology & Hepatology (AREA)
- Mycology (AREA)
- Biomedical Technology (AREA)
- Wood Science & Technology (AREA)
- Toxicology (AREA)
- Oncology (AREA)
- General Engineering & Computer Science (AREA)
- Biotechnology (AREA)
- Cell Biology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
Claims
Priority Applications (7)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2025504072A JP2025526369A (en) | 2022-07-27 | 2023-07-26 | Combination of recombinant viruses expressing interleukin-12 with PD-1/PD-L1 inhibitors |
| AU2023313118A AU2023313118A1 (en) | 2022-07-27 | 2023-07-26 | Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors |
| CA3262167A CA3262167A1 (en) | 2022-07-27 | 2023-07-26 | Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors |
| EP23754426.7A EP4561599A1 (en) | 2022-07-27 | 2023-07-26 | Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors |
| CN202380068582.4A CN119923267A (en) | 2022-07-27 | 2023-07-26 | Combination of recombinant virus expressing interleukin-12 and PD-1/PD-L1 inhibitor |
| KR1020257005580A KR20250044313A (en) | 2022-07-27 | 2023-07-26 | Combination of a PD-1/PD-L1 inhibitor and a recombinant virus expressing interleukin-12 |
| MX2025001073A MX2025001073A (en) | 2022-07-27 | 2025-01-27 | Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202263369605P | 2022-07-27 | 2022-07-27 | |
| US63/369,605 | 2022-07-27 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024023740A1 true WO2024023740A1 (en) | 2024-02-01 |
Family
ID=87571419
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/IB2023/057598 Ceased WO2024023740A1 (en) | 2022-07-27 | 2023-07-26 | Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors |
Country Status (8)
| Country | Link |
|---|---|
| EP (1) | EP4561599A1 (en) |
| JP (1) | JP2025526369A (en) |
| KR (1) | KR20250044313A (en) |
| CN (1) | CN119923267A (en) |
| AU (1) | AU2023313118A1 (en) |
| CA (1) | CA3262167A1 (en) |
| MX (1) | MX2025001073A (en) |
| WO (1) | WO2024023740A1 (en) |
Citations (32)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5939598A (en) | 1990-01-12 | 1999-08-17 | Abgenix, Inc. | Method of making transgenic mice lacking endogenous heavy chains |
| WO2007147528A1 (en) | 2006-06-20 | 2007-12-27 | Transgene S.A. | Process for producing poxviruses and poxvirus compositions |
| WO2008156712A1 (en) | 2007-06-18 | 2008-12-24 | N. V. Organon | Antibodies to human programmed death receptor pd-1 |
| WO2009065546A1 (en) | 2007-11-19 | 2009-05-28 | Transgene Sa | Poxviral oncolytic vectors |
| US7943743B2 (en) | 2005-07-01 | 2011-05-17 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (PD-L1) |
| WO2011066389A1 (en) | 2009-11-24 | 2011-06-03 | Medimmmune, Limited | Targeted binding agents against b7-h1 |
| US8008449B2 (en) | 2005-05-09 | 2011-08-30 | Medarex, Inc. | Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics |
| US8217149B2 (en) | 2008-12-09 | 2012-07-10 | Genentech, Inc. | Anti-PD-L1 antibodies, compositions and articles of manufacture |
| WO2012145493A1 (en) | 2011-04-20 | 2012-10-26 | Amplimmune, Inc. | Antibodies and other molecules that bind b7-h1 and pd-1 |
| US20130017199A1 (en) | 2009-11-24 | 2013-01-17 | AMPLIMMUNE ,Inc. a corporation | Simultaneous inhibition of pd-l1/pd-l2 |
| WO2013014668A1 (en) | 2011-07-24 | 2013-01-31 | Curetech Ltd. | Variants of humanized immunomodulatory monoclonal antibodies |
| WO2013079174A1 (en) | 2011-11-28 | 2013-06-06 | Merck Patent Gmbh | Anti-pd-l1 antibodies and uses thereof |
| WO2013173223A1 (en) | 2012-05-15 | 2013-11-21 | Bristol-Myers Squibb Company | Cancer immunotherapy by disrupting pd-1/pd-l1 signaling |
| WO2013181634A2 (en) | 2012-05-31 | 2013-12-05 | Sorrento Therapeutics Inc. | Antigen binding proteins that bind pd-l1 |
| WO2014179664A2 (en) | 2013-05-02 | 2014-11-06 | Anaptysbio, Inc. | Antibodies directed against programmed death-1 (pd-1) |
| WO2014194302A2 (en) | 2013-05-31 | 2014-12-04 | Sorrento Therapeutics, Inc. | Antigen binding proteins that bind pd-1 |
| US20150079109A1 (en) | 2013-09-13 | 2015-03-19 | Beigene, Ltd. | Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics |
| WO2015085847A1 (en) | 2013-12-12 | 2015-06-18 | 上海恒瑞医药有限公司 | Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof |
| WO2015112900A1 (en) | 2014-01-24 | 2015-07-30 | Dana-Farber Cancer Institue, Inc. | Antibody molecules to pd-1 and uses thereof |
| WO2015112800A1 (en) | 2014-01-23 | 2015-07-30 | Regeneron Pharmaceuticals, Inc. | Human antibodies to pd-1 |
| WO2016049492A1 (en) * | 2014-09-26 | 2016-03-31 | The Usa, As Represented By The Secretary, Dept. Of Health And Human Services | Virus-based expression vectors and uses thereof |
| WO2016149201A2 (en) | 2015-03-13 | 2016-09-22 | Cytomx Therapeutics, Inc. | Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof |
| WO2017019846A1 (en) | 2015-07-30 | 2017-02-02 | Macrogenics, Inc. | Pd-1-binding molecules and methods use thereof |
| WO2017025016A1 (en) | 2015-08-10 | 2017-02-16 | Innovent Biologics (Suzhou) Co., Ltd. | Pd-1 antibodies |
| WO2017034916A1 (en) | 2015-08-24 | 2017-03-02 | Eli Lilly And Company | Pd-l1 ("programmed death-ligand 1") antibodies |
| WO2017040790A1 (en) | 2015-09-01 | 2017-03-09 | Agenus Inc. | Anti-pd-1 antibodies and methods of use thereof |
| WO2017132825A1 (en) | 2016-02-02 | 2017-08-10 | 华为技术有限公司 | Emission power verification method, user equipment, and base station |
| WO2017133540A1 (en) | 2016-02-02 | 2017-08-10 | Innovent Biologics (Suzhou) Co., Ltd. | Pd-1 antibodies |
| WO2020074902A1 (en) * | 2018-10-10 | 2020-04-16 | Queen Mary University Of London | Oncolytic vaccinia virus with modified b5r gene for the treatment of cancer |
| WO2020124274A1 (en) * | 2018-12-21 | 2020-06-25 | Ottawa Hospital Research Institute | Modified orthopoxvirus vectors |
| WO2021040056A1 (en) * | 2019-08-29 | 2021-03-04 | Astellas Pharma Inc. | Genetically engineered oncolytic vaccinia viruses and methods of uses thereof |
| EP3858369A1 (en) * | 2018-09-26 | 2021-08-04 | Astellas Pharma Inc. | Cancer therapy where oncolytic vaccinia virus and immune checkpoint inhibitor are used in combination, and pharmaceutical composition and combination drug used therein |
-
2023
- 2023-07-26 AU AU2023313118A patent/AU2023313118A1/en active Pending
- 2023-07-26 KR KR1020257005580A patent/KR20250044313A/en active Pending
- 2023-07-26 WO PCT/IB2023/057598 patent/WO2024023740A1/en not_active Ceased
- 2023-07-26 JP JP2025504072A patent/JP2025526369A/en active Pending
- 2023-07-26 CA CA3262167A patent/CA3262167A1/en active Pending
- 2023-07-26 EP EP23754426.7A patent/EP4561599A1/en active Pending
- 2023-07-26 CN CN202380068582.4A patent/CN119923267A/en active Pending
-
2025
- 2025-01-27 MX MX2025001073A patent/MX2025001073A/en unknown
Patent Citations (36)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5939598A (en) | 1990-01-12 | 1999-08-17 | Abgenix, Inc. | Method of making transgenic mice lacking endogenous heavy chains |
| US8008449B2 (en) | 2005-05-09 | 2011-08-30 | Medarex, Inc. | Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics |
| US7943743B2 (en) | 2005-07-01 | 2011-05-17 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (PD-L1) |
| WO2007147528A1 (en) | 2006-06-20 | 2007-12-27 | Transgene S.A. | Process for producing poxviruses and poxvirus compositions |
| WO2008156712A1 (en) | 2007-06-18 | 2008-12-24 | N. V. Organon | Antibodies to human programmed death receptor pd-1 |
| WO2009065546A1 (en) | 2007-11-19 | 2009-05-28 | Transgene Sa | Poxviral oncolytic vectors |
| US8217149B2 (en) | 2008-12-09 | 2012-07-10 | Genentech, Inc. | Anti-PD-L1 antibodies, compositions and articles of manufacture |
| US9493565B2 (en) | 2009-11-24 | 2016-11-15 | Medimmune Limited | Targeted binding agents against B7-H1 |
| WO2011066389A1 (en) | 2009-11-24 | 2011-06-03 | Medimmmune, Limited | Targeted binding agents against b7-h1 |
| US20130017199A1 (en) | 2009-11-24 | 2013-01-17 | AMPLIMMUNE ,Inc. a corporation | Simultaneous inhibition of pd-l1/pd-l2 |
| WO2012145493A1 (en) | 2011-04-20 | 2012-10-26 | Amplimmune, Inc. | Antibodies and other molecules that bind b7-h1 and pd-1 |
| US8686119B2 (en) | 2011-07-24 | 2014-04-01 | Curetech Ltd. | Variants of humanized immunomodulatory monoclonal antibodies |
| WO2013014668A1 (en) | 2011-07-24 | 2013-01-31 | Curetech Ltd. | Variants of humanized immunomodulatory monoclonal antibodies |
| WO2013079174A1 (en) | 2011-11-28 | 2013-06-06 | Merck Patent Gmbh | Anti-pd-l1 antibodies and uses thereof |
| WO2013173223A1 (en) | 2012-05-15 | 2013-11-21 | Bristol-Myers Squibb Company | Cancer immunotherapy by disrupting pd-1/pd-l1 signaling |
| WO2013181634A2 (en) | 2012-05-31 | 2013-12-05 | Sorrento Therapeutics Inc. | Antigen binding proteins that bind pd-l1 |
| WO2014179664A2 (en) | 2013-05-02 | 2014-11-06 | Anaptysbio, Inc. | Antibodies directed against programmed death-1 (pd-1) |
| WO2014194302A2 (en) | 2013-05-31 | 2014-12-04 | Sorrento Therapeutics, Inc. | Antigen binding proteins that bind pd-1 |
| WO2015035606A1 (en) | 2013-09-13 | 2015-03-19 | Beigene, Ltd. | Anti-pd1 antibodies and their use as therapeutics and diagnostics |
| US20150079109A1 (en) | 2013-09-13 | 2015-03-19 | Beigene, Ltd. | Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics |
| WO2015085847A1 (en) | 2013-12-12 | 2015-06-18 | 上海恒瑞医药有限公司 | Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof |
| WO2015112800A1 (en) | 2014-01-23 | 2015-07-30 | Regeneron Pharmaceuticals, Inc. | Human antibodies to pd-1 |
| WO2015112900A1 (en) | 2014-01-24 | 2015-07-30 | Dana-Farber Cancer Institue, Inc. | Antibody molecules to pd-1 and uses thereof |
| WO2016049492A1 (en) * | 2014-09-26 | 2016-03-31 | The Usa, As Represented By The Secretary, Dept. Of Health And Human Services | Virus-based expression vectors and uses thereof |
| WO2016149201A2 (en) | 2015-03-13 | 2016-09-22 | Cytomx Therapeutics, Inc. | Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof |
| WO2017019846A1 (en) | 2015-07-30 | 2017-02-02 | Macrogenics, Inc. | Pd-1-binding molecules and methods use thereof |
| WO2017024465A1 (en) | 2015-08-10 | 2017-02-16 | Innovent Biologics (Suzhou) Co., Ltd. | Pd-1 antibodies |
| WO2017025016A1 (en) | 2015-08-10 | 2017-02-16 | Innovent Biologics (Suzhou) Co., Ltd. | Pd-1 antibodies |
| WO2017034916A1 (en) | 2015-08-24 | 2017-03-02 | Eli Lilly And Company | Pd-l1 ("programmed death-ligand 1") antibodies |
| WO2017040790A1 (en) | 2015-09-01 | 2017-03-09 | Agenus Inc. | Anti-pd-1 antibodies and methods of use thereof |
| WO2017132825A1 (en) | 2016-02-02 | 2017-08-10 | 华为技术有限公司 | Emission power verification method, user equipment, and base station |
| WO2017133540A1 (en) | 2016-02-02 | 2017-08-10 | Innovent Biologics (Suzhou) Co., Ltd. | Pd-1 antibodies |
| EP3858369A1 (en) * | 2018-09-26 | 2021-08-04 | Astellas Pharma Inc. | Cancer therapy where oncolytic vaccinia virus and immune checkpoint inhibitor are used in combination, and pharmaceutical composition and combination drug used therein |
| WO2020074902A1 (en) * | 2018-10-10 | 2020-04-16 | Queen Mary University Of London | Oncolytic vaccinia virus with modified b5r gene for the treatment of cancer |
| WO2020124274A1 (en) * | 2018-12-21 | 2020-06-25 | Ottawa Hospital Research Institute | Modified orthopoxvirus vectors |
| WO2021040056A1 (en) * | 2019-08-29 | 2021-03-04 | Astellas Pharma Inc. | Genetically engineered oncolytic vaccinia viruses and methods of uses thereof |
Non-Patent Citations (23)
| Title |
|---|
| "Basic and Clinical Immunology", vol. 6, 1994, APPLETON & LANGE, NORWALK, pages: 71 |
| AI ET AL., DRUG DES DEVEL THER., vol. 14, 2020, pages 3625 - 3649 |
| ALHARBI NAIF KHALAF: "Poxviral promoters for improving the immunogenicity of MVA delivered vaccines", HUMAN VACCINES & IMMUNOTHERAPEUTICS, vol. 15, no. 1, 6 September 2018 (2018-09-06), US, pages 203 - 209, XP055875123, ISSN: 2164-5515, DOI: 10.1080/21645515.2018.1513439 * |
| BALDICK ET AL., J. VIROL., vol. 67, 1993, pages 3515 - 3527 |
| CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917 |
| FOLOPPE ET AL., MOL THER ONCOLYTICS, vol. 14, 2019, pages 1 - 14 |
| GE YAN ET AL: "Oncolytic vaccinia virus delivering tethered IL -12 enhances antitumor effects with improved safety", JOURNAL FOR IMMUNOTHERAPY OF CANCER, BIOMED CENTRAL, LONDON, GB, vol. 8, no. 1, 1 March 2020 (2020-03-01), pages 1 - 8, XP009529247, ISSN: 2051-1426, [retrieved on 20200324], DOI: 10.1136/JITC-2020-000710 * |
| GORELIK ET AL., AACR:ABSTRACT, April 2016 (2016-04-01), pages 4606 |
| HAMMOND J M ET AL: "A SYNTHETIC VACCINIA VIRUS PROMOTER WITH ENHANCED EARLY AND LATE ACTIVITY", JOURNAL OF VIROLOGICAL METHODS, ELSEVIER BV, NL, vol. 66, no. 1, 1 January 1997 (1997-01-01), pages 135 - 138, XP001086677, ISSN: 0166-0934, DOI: 10.1016/S0166-0934(97)00045-1 * |
| HERBST ET AL., J CLIN ONCOL, vol. 3, 2013, pages 3000 |
| KABAT EA ET AL.: "Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services", NIH PUBLICATION, no. 91-3242, 1991 |
| KABAT EA, WU TT, ANN NY ACAD SCI, vol. 190, 1971, pages 382 - 391 |
| KABAT, E. ET AL.: "Sequences of Proteins of Immunological Interest", U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, 1983 |
| KLEINPETER ET AL., ONCOIMMUNOLOGY, vol. 5, 2016, pages el220467 |
| KNUTSON BRUCE A. ET AL: "Vaccinia Virus Intermediate and Late Promoter Elements Are Targeted by the TATA-Binding Protein", JOURNAL OF VIROLOGY, vol. 80, no. 14, 15 July 2006 (2006-07-15), US, pages 6784 - 6793, XP093094190, ISSN: 0022-538X, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1489061/pdf/2705-05.pdf> DOI: 10.1128/JVI.02705-05 * |
| KUROKAWA CHEYNE ET AL: "835?AZD4820 oncolytic vaccinia virus encoding IL-12 mediates anti-tumor activity through oncolysis and tumor-specific immunity", REGULAR AND YOUNG INVESTIGATOR AWARD ABSTRACTS, 1 November 2022 (2022-11-01), pages A871 - A871, XP093094053, DOI: 10.1136/jitc-2022-SITC2022.0835 * |
| LIESCHKE ET AL., NAT BIOTECHNOL., vol. 15, no. 1, pages 35 - 40 |
| SI-YANG LIU ET AL., J HEMATOL. ONCOL., vol. 70, 2017, pages 136 |
| SI-YANG LIU ET AL., J. HEMATOL. ONCOL., vol. 70, 2017, pages 136 |
| TIAN YAOMEI ET AL: "Engineering strategies to enhance oncolytic viruses in cancer immunotherapy", SIGNAL TRANSDUCTION AND TARGETED THERAPY, vol. 7, no. 1, 6 April 2022 (2022-04-06), XP093094879, Retrieved from the Internet <URL:https://www.nature.com/articles/s41392-022-00951-x> DOI: 10.1038/s41392-022-00951-x * |
| YANG, J. VIROL., no. 19, 2011, pages 9899 - 9908 |
| ZHANG ET AL., CELL DISCOV., vol. 7, March 2017 (2017-03-01), pages 3 |
| ZONG SHENG GUO ET AL: "Vaccinia virus-mediated cancer immunotherapy: cancer vaccines and oncolytics", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 7, no. 1, 9 January 2019 (2019-01-09), XP055677423, DOI: 10.1186/s40425-018-0495-7 * |
Also Published As
| Publication number | Publication date |
|---|---|
| CA3262167A1 (en) | 2024-02-01 |
| CN119923267A (en) | 2025-05-02 |
| MX2025001073A (en) | 2025-05-02 |
| AU2023313118A1 (en) | 2025-03-06 |
| JP2025526369A (en) | 2025-08-13 |
| KR20250044313A (en) | 2025-03-31 |
| EP4561599A1 (en) | 2025-06-04 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7077377B2 (en) | Oncolytic virus for expression of immune checkpoint modulator | |
| JP7038664B2 (en) | Manipulated oncolytic virus | |
| ES2704632T3 (en) | Oncolytic herpes simplex virus and its therapeutic uses | |
| US7429481B2 (en) | Targeting viruses using a modified sindbis glycoprotein | |
| JP7625518B2 (en) | Therapy for treating cancer by intratumoral and/or intravenous administration of recombinant MVA encoding 4-1BBL (CD137L) and/or CD40L | |
| JP2021527694A (en) | Treatment with oncolytic virus | |
| US20230357793A1 (en) | Oncolytic herpes simplex viruses (hsv) expressing immunomodulatory fusion proteins | |
| US20240269203A1 (en) | Oncolytic virus and uses thereof | |
| JP7644111B2 (en) | Recombinant MVA virus for intratumoral and/or intravenous administration to treat cancer - Patents.com | |
| JP7555334B2 (en) | Combination therapy to treat cancer by intravenous administration of recombinant MVA and immune checkpoint antagonist or immune checkpoint agonist | |
| KR20230023032A (en) | recombinant vaccinia virus | |
| WO2022101302A1 (en) | Antibodies conjugated or fused to the receptor-binding domain of the sars-cov-2 spike protein and uses thereof for vaccine purposes | |
| WO2024023740A1 (en) | Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors | |
| AU2023283081A1 (en) | Recombinant virus expressing interleukin-12 | |
| RU2830601C2 (en) | Therapy for treating cancer by intratumoral and/or intravenous administration of recombinant mva coding 4-1bbl (cd137l) and/or cd40l | |
| KR20230105035A (en) | Recombinant vaccinia virus | |
| HK40088967A (en) | Recombinant vaccinia virus |
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: 23754426 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025504072 Country of ref document: JP |
|
| WWE | Wipo information: entry into national phase |
Ref document number: MX/A/2025/001073 Country of ref document: MX |
|
| REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112025001430 Country of ref document: BR |
|
| WWE | Wipo information: entry into national phase |
Ref document number: AU2023313118 Country of ref document: AU |
|
| ENP | Entry into the national phase |
Ref document number: 20257005580 Country of ref document: KR Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025104183 Country of ref document: RU Ref document number: 2023754426 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2023313118 Country of ref document: AU Date of ref document: 20230726 Kind code of ref document: A |
|
| ENP | Entry into the national phase |
Ref document number: 2023754426 Country of ref document: EP Effective date: 20250227 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 202380068582.4 Country of ref document: CN |
|
| WWP | Wipo information: published in national office |
Ref document number: 1020257005580 Country of ref document: KR |
|
| WWP | Wipo information: published in national office |
Ref document number: MX/A/2025/001073 Country of ref document: MX Ref document number: 202380068582.4 Country of ref document: CN |
|
| WWP | Wipo information: published in national office |
Ref document number: 2023754426 Country of ref document: EP |
|
| WWP | Wipo information: published in national office |
Ref document number: 2025104183 Country of ref document: RU |
|
| ENP | Entry into the national phase |
Ref document number: 112025001430 Country of ref document: BR Kind code of ref document: A2 Effective date: 20250124 |