CN117222436A - Modified oncolytic virus, compositions comprising modified oncolytic virus and uses thereof - Google Patents

Modified oncolytic virus, compositions comprising modified oncolytic virus and uses thereof Download PDF

Info

Publication number
CN117222436A
CN117222436A CN202180083706.7A CN202180083706A CN117222436A CN 117222436 A CN117222436 A CN 117222436A CN 202180083706 A CN202180083706 A CN 202180083706A CN 117222436 A CN117222436 A CN 117222436A
Authority
CN
China
Prior art keywords
oncolytic virus
modified oncolytic
sequence
acid sequence
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.)
Pending
Application number
CN202180083706.7A
Other languages
Chinese (zh)
Inventor
梁旻
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Jinsi Yuanchuang Biotechnology Co ltd
Shanghai Jinsi Biotechnology Co ltd
Original Assignee
Shanghai Jinsi Yuanchuang Biotechnology Co ltd
Shanghai Jinsi Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Jinsi Yuanchuang Biotechnology Co ltd, Shanghai Jinsi Biotechnology Co ltd filed Critical Shanghai Jinsi Yuanchuang Biotechnology Co ltd
Publication of CN117222436A publication Critical patent/CN117222436A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4747Apoptosis related proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24161Methods of inactivation or attenuation
    • C12N2710/24162Methods of inactivation or attenuation by genetic engineering

Landscapes

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

Abstract

The present invention provides a modified oncolytic virus having a first heterologous polynucleotide encoding a first molecule capable of inhibiting the interaction between PD-1 and PD-L1 and a second heterologous polynucleotide encoding a second molecule capable of inhibiting TGF-beta signaling. The invention also provides a pharmaceutical composition comprising a modified oncolytic virus and uses of the pharmaceutical composition, when in use, to administer the modified oncolytic virus or pharmaceutical composition to a subject. The modified oncolytic virus can selectively replicate in tumor cells, and the replication rate of the modified oncolytic virus is obviously higher than that of non-tumor cells in the tumor cells, so that the efficacy of the oncolytic virus is enhanced, and metastatic tumor cells are better cleared.

Description

Modified oncolytic virus, compositions comprising modified oncolytic virus and uses thereof
Technical Field
The invention relates to the technical field of tumor treatment, in particular to a modified oncolytic virus, a composition containing the modified oncolytic virus and application thereof.
Background
Over 1400 ten thousand people worldwide are diagnosed with tumors each year. Despite many advances in medical research, tumors account for about 16% of all deaths.
Malignant tumors are often resistant to conventional therapies and represent a rather difficult therapeutic challenge. For example, micrometastases can be established at a very early stage of primary tumor development. Thus, at the time of diagnosis, many tumor patients already have micrometastases. Tumor-reactive T cells can locate and destroy micrometastases and protect surrounding healthy tissue. However, naturally occurring anti-malignant T cell responses are often insufficient to cause regression of primary or metastatic tumors.
Oncolytic viruses have shown potential as antitumor agents. Unlike conventional gene therapy, oncolytic viruses are able to spread through tumor tissue by viral replication and concomitant cell lysis. However, oncolytic viruses alone are not sufficient to treat primary or metastatic tumors.
Thus, there is a particular urgent need to enhance the efficacy of oncolytic viruses and to clear metastatic tumor cells.
Disclosure of Invention
In one aspect, the disclosure relates to a modified oncolytic virus comprising a viral genome having a first heterologous polynucleotide encoding a first molecule capable of inhibiting an interaction between PD-1 and PD-L1 and a second heterologous polynucleotide encoding a second molecule capable of inhibiting TGF- β signaling.
In certain embodiments, the oncolytic virus is selected from the group consisting of vaccinia virus, adenovirus, reovirus, measles virus, herpes simplex virus, semliki forest virus, venezuelan equine encephalitis virus, parvovirus, chicken anaemia virus, measles virus, coxsackie virus, vesicular stomatitis virus, saikovirus, malabar virus, newcastle disease virus, and myxoma virus. In certain embodiments, the oncolytic virus is a vaccinia virus.
In certain embodiments, the modified oncolytic virus is an attenuated virus and can replicate in tumor cells.
In certain embodiments, the viral genome comprises at least one deletion or disruption, such that the virus is capable of selective replication in tumor cells. In certain embodiments, the deletion or disruption is located in an Open Reading Frame (ORF) encoding at least a portion of an enzyme that is both critical for viral replication and preferentially expressed in tumor cells over non-tumor cells. In certain embodiments, the enzyme is a kinase. In certain embodiments, the enzyme is thymidine kinase.
In certain embodiments, the oncolytic virus is derived from a Western Reserve strain.
In certain embodiments, the first heterologous polynucleotide and the second heterologous polynucleotide are inserted in place of the deletion.
In certain embodiments, the first heterologous polynucleotide and the second heterologous polynucleotide are configured such that they are expressed in the same or different phases of the replication cycle of the modified oncolytic virus.
In certain embodiments, the first molecule is a first fusion protein and the second molecule is a second fusion protein. In certain embodiments, the first heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: first promoter-polynucleotide encoding first fusion protein-first stop codon, and second heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: second promoter-polynucleotide encoding a second fusion protein-second stop codon.
In certain embodiments, the first fusion protein expressed from the first heterologous polynucleotide and the second fusion protein expressed from the second heterologous polynucleotide are expressed as separate proteins. In certain embodiments, the first heterologous polynucleotide is immediately upstream or immediately downstream of the second heterologous polynucleotide. In certain embodiments, wherein the first promoter is capable of driving expression of the first fusion protein and the second promoter is capable of driving expression of the second fusion protein, wherein the first promoter and the second promoter are in a head-to-head orientation.
In certain embodiments, the first promoter and the second promoter are the same or different. In certain embodiments, the first promoter and the second promoter are both early and late promoters. In certain embodiments, the early and late promoters are pSE/L.
In certain embodiments, the first stop codon and the second stop codon are the same or different.
In certain embodiments, the first fusion protein comprises a PD-1 extracellular domain (PD-1 ECD). In certain embodiments, the first fusion protein further comprises a first immunoglobulin Fc region. In certain embodiments, the first immunoglobulin Fc region is a first human IgG1 Fc region.
In certain embodiments, the PD-1ECD is operably linked to the first immunoglobulin Fc region at the C-terminus of the PD-1 ECD.
In certain embodiments, the first fusion protein further comprises a signal peptide.
In certain embodiments, the signal peptide is operably linked to the PD-1ECD at the C-terminus of the signal peptide. In certain embodiments, the signal peptide is a CD33 signal peptide.
In certain embodiments, the PD-1ECD comprises the amino acid sequence of SEQ ID NO. 1 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO. 7 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the first human IgG1 Fc region comprises the amino acid sequence of SEQ ID NO. 3 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO. 9 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the CD33 signal peptide comprises the amino acid sequence of SEQ ID NO. 4 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID NO. 11 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the first fusion protein comprises the amino acid sequence of SEQ ID NO. 5 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID NO. 13 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the first molecule is an anti-PD-1 antibody and the second molecule is a second fusion protein.
In certain embodiments, an anti-PD-1 antibody comprises: HCDR1 having the amino acid sequence of SEQ ID No. 23 or a homologous sequence thereof having at least 80% sequence identity, HCDR2 having the amino acid sequence of SEQ ID No. 24 or a homologous sequence thereof having at least 80% sequence identity, HCDR3 having the amino acid sequence of SEQ ID No. 25 or a homologous sequence thereof having at least 80% sequence identity, LCDR1 having the amino acid sequence of SEQ ID No. 26 or a homologous sequence thereof having at least 80% sequence identity, LCDR2 having the amino acid sequence of SEQ ID No. 27 or a homologous sequence thereof having at least 80% sequence identity and LCDR3 having the amino acid sequence of SEQ ID No. 28 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, HCDR1 is encoded by the nucleic acid sequence of SEQ ID NO. 31 or a homologous sequence thereof having at least 80% sequence identity, HCDR2 is encoded by the nucleic acid sequence of SEQ ID NO. 32 or a homologous sequence thereof having at least 80% sequence identity, HCDR3 is encoded by the nucleic acid sequence of SEQ ID NO. 33 or a homologous sequence thereof having at least 80% sequence identity, LCDR1 is encoded by the nucleic acid sequence of SEQ ID NO. 34 or a homologous sequence thereof having at least 80% sequence identity, LCDR2 is encoded by the nucleic acid sequence of SEQ ID NO. 35 or a homologous sequence thereof having at least 80% sequence identity, and LCDR3 is encoded by the nucleic acid sequence of SEQ ID NO. 36 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, an anti-PD-1 antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID NO. 29 or a homologous sequence thereof having at least 80% sequence identity and a light chain variable region having the amino acid sequence of SEQ ID NO. 30 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the anti-PD-1 antibody heavy chain variable region is encoded by the nucleic acid sequence of SEQ ID NO. 37 or a homologous sequence thereof having at least 80% sequence identity, and the anti-PD-1 antibody light chain variable region is encoded by the nucleic acid sequence of SEQ ID NO. 38 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, an anti-PD-1 antibody comprises a full length heavy chain having the amino acid sequence of SEQ ID NO. 21 or a homologous sequence thereof having at least 80% sequence identity and a full length light chain having the amino acid sequence of SEQ ID NO. 20 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the full length heavy chain of the anti-PD-1 antibody is encoded by the nucleic acid sequence of SEQ ID NO. 39 or a homologous sequence thereof having at least 80% sequence identity, and the full length light chain of the anti-PD-1 antibody is encoded by the nucleic acid sequence of SEQ ID NO. 40 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the first heterologous polynucleotide further comprises a third heterologous polynucleotide and a fourth heterologous polynucleotide, wherein the third heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: third promoter-polynucleotide encoding heavy chain of anti-PD-1 antibody-third stop codon, and wherein the fourth heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: fourth promoter-polynucleotide encoding anti-PD-1 antibody light chain-fourth stop codon; and the second heterologous polynucleotide further comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: second promoter-polynucleotide encoding a second fusion protein-second stop codon.
In certain embodiments, the third heterologous polynucleotide is immediately upstream or immediately downstream of the fourth heterologous polynucleotide. In certain embodiments, the first heterologous polynucleotide is immediately upstream or immediately downstream of the second heterologous polynucleotide.
In certain embodiments, the third promoter is capable of driving expression of the anti-PD-1 antibody heavy chain and the fourth promoter is capable of driving expression of the anti-PD-1 antibody light chain, wherein the third promoter and the fourth promoter are in a head-to-head orientation.
In certain embodiments, the anti-PD-1 antibody expressed from the first heterologous polynucleotide and the second fusion protein expressed from the second heterologous polynucleotide are expressed as separate proteins.
In certain embodiments, the second promoter, the third promoter, and the fourth promoter are the same or different. In certain embodiments, the second promoter, the third promoter, and the fourth promoter are all early and late promoters. In certain embodiments, the early and late promoters are pSE/L.
In certain embodiments, the second stop codon, the third stop codon, and the fourth stop codon are the same or different.
In certain embodiments, the second fusion protein comprises a TGF- β receptor II extracellular domain (TGFBRII ECD). In certain embodiments, the second fusion protein further comprises a second immunoglobulin Fc region. In certain embodiments, the immunoglobulin Fc region is a second human IgG1 Fc region.
In certain embodiments, the TGFBRII ECD is operably linked to the second immunoglobulin Fc region at the C-terminus of the TGFBRII ECD.
In certain embodiments, the second fusion protein further comprises a signal peptide. In certain embodiments, the signal peptide is operably linked to TGFBRII ECD at the C-terminus of the signal peptide. In certain embodiments, the signal peptide is a CD33 signal peptide.
In certain embodiments, the TGFBRII ECD comprises the amino acid sequence of SEQ ID NO. 2 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO. 8 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the second human IgG1 Fc region comprises the amino acid sequence of SEQ ID NO. 3 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO. 10 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the CD33 signal peptide comprises the amino acid sequence of SEQ ID NO. 4 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID NO. 12 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the second fusion protein comprises the amino acid sequence of SEQ ID NO. 6 or a homologous sequence thereof having at least 80% sequence identity. In certain embodiments, the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID NO. 14 or a homologous sequence thereof having at least 80% sequence identity.
In certain embodiments, the first fusion protein and the second fusion protein are capable of forming a dimer. In certain embodiments, the dimer is formed by association of a first immunoglobulin Fc region with a second immunoglobulin Fc region.
In certain embodiments, the modified oncolytic viruses provided herein have the nucleic acid sequence of SEQ ID NO. 17 or SEQ ID NO. 22.
In one aspect, the invention provides a pharmaceutical composition comprising a modified oncolytic virus provided herein and a pharmaceutically acceptable carrier.
In one aspect, the invention provides the use of a pharmaceutical composition for the treatment of a tumor, in which an effective amount of a modified oncolytic virus or pharmaceutical composition is administered to a subject, wherein the subject is a human, the route of administration is topical, and the route of administration is intratumoral injection;
Wherein the tumor is a solid tumor, which is melanoma, non-small cell lung cancer, renal cell carcinoma, hodgkin's lymphoma, head and neck squamous cell carcinoma, bladder cancer, colorectal cancer, triple negative breast cancer, hepatocellular carcinoma, pancreatic cancer, ovarian cancer, colon cancer, pharyngeal squamous cell carcinoma, or ovarian teratoma.
Compared with the prior art, the invention has the following beneficial effects:
the present invention introduces the first heterologous polynucleotide and the second heterologous polynucleotide into the modified oncolytic virus such that the oncolytic virus is modified, the modified oncolytic virus being capable of selectively replicating in tumor cells where the replication rate of the modified oncolytic virus is significantly higher than the replication rate in non-tumor cells, thereby enhancing the efficacy of the oncolytic virus and better clearing metastatic tumor cells.
Drawings
FIG. 1 shows a WR-GO-001 vaccinia virus construct comprising the insertion of nucleic acid sequences encoding human PD-1G22-170ECD Fc fusion protein and TGF-beta RII 23-166ECD Fc fusion protein.
FIG. 2 shows a WR-GO-002 vaccinia virus construct comprising the insertion of a nucleic acid sequence encoding the TGF-beta RII 23-166ECD Fc fusion protein.
FIG. 3 shows a WR-GO-003 vaccinia virus construct comprising the insertion of a nucleic acid sequence encoding a human PD-1G22-170ECD Fc fusion protein.
FIG. 4 shows a WR-GO-004 vaccinia virus construct comprising an insertion of nucleic acid sequences encoding anti-PD-1 antibodies and TGF-beta RII 23-166ECD Fc fusion proteins.
FIGS. 5A-5C show the expression of human PD-1Fc chimeric proteins, human TGF-beta receptor II Fc chimeric proteins and anti-human PD-1 antibodies in both supernatant and intracellular samples of WR-GO-002, WR-GO-003 or WR-GO-004 infected Hela cells measured using ELISA methods.
FIGS. 6A-6H show cytotoxicity of WR-GO-001, WR-GO-002, WR-GO-003 and WR-GO-004 in MC38 and CT26 cell lines.
FIGS. 7A to 7D show cytotoxicity assays of WR-GO-003 in human tumor cell lines (FaDu, PA1, PANC-1) and human normal cell lines (HUVEC).
FIGS. 8A to 8D show cytotoxicity assays of WR-GO-004 in human tumor cell lines (FaDu, PA1, PANC-1) and human normal cell lines (HUVEC).
Fig. 9A and 9B show the fold change in VP of WR-GO-003 or WR-GO-004 in FaDu.
Detailed Description
The present invention will be described in further detail with reference to examples and drawings, but embodiments of the present invention are not limited thereto.
In one aspect, the disclosure relates to a modified oncolytic virus comprising a viral genome having a first heterologous polynucleotide encoding a first molecule capable of inhibiting an interaction between PD-1 and PD-L1 and a second heterologous polynucleotide encoding a second molecule capable of inhibiting TGF- β signaling.
Oncolytic viruses
The term "oncolytic virus" as used in this patent refers to a virus that is capable of selectively replicating and slowing the growth of or inducing death of tumor cells in vitro or in vivo with minimal or no impact on normal cells. In certain embodiments, the oncolytic virus contains a viral genome packaged into a viral particle (or virion) and is infectious (i.e., capable of infecting and entering a host cell or subject). In certain embodiments, the oncolytic virus may be a DNA virus or an RNA virus, and may be in any suitable form, such as a DNA virus vector, an RNA virus vector, or a viral particle.
As used in this patent, the term "selective replication" refers to the replication rate of oncolytic viruses in tumor cells being significantly higher than the replication rate in non-tumor cells (e.g., healthy cells). In certain embodiments, the oncolytic virus exhibits a lysis rate in tumor cells that is at least 50%, 60%, 70%, 80%, 90%, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 50-fold, 100-fold, or 1000-fold higher than in non-tumor cells (e.g., healthy cells).
In certain embodiments, the oncolytic viruses of the present disclosure can selectively replicate in liver tumor cells (e.g., hepal-6 cells, hep3B cells, 7402 cells, and 7721 cells), breast tumor cells (e.g., MCF-7 cells), tongue tumor cells (e.g., TCa8113 cells), adenoid cystic tumor cells (e.g., ACC-M cells), prostate tumor cells (e.g., LNCaP cells), human embryonic kidney cells (e.g., HEK293 cells), lung tumor cells (e.g., a549 cells), or cervical tumor cells (e.g., hela cells).
Oncolytic viruses of the present disclosure may be derived from poxviruses (e.g., vaccinia virus), adenoviruses (e.g., delta-24-RGD, ICOVIR-5, ICOVIR-7, onyx-015, coloAdl, H101, and AD 5/3-D24-GMCSF), reoviruses (e.g., reolysin), measles viruses, herpes simplex viruses (e.g., HSV, oncoveX GMCSF), newcastle viruses (e.g., 73-T PV701 and HDV-HUJ strains, and those described in Phungsab et al, 2001, cancer Lett., vol.172:1, pages 27-36; lorence et al, 2007, curr Drug Targets, volume 7, page 2: 157-167; and Freeman et al, 2006, mol. Ther, volume 13, page 1: 221-228), retrovirus (e.g., influenza virus), myxoma virus, rhabdovirus (e.g., vesicular stomatitis virus; those described in Stojdl et al, 2000, nat. Med., volume 6, page 7: 821-825; and Stojdl et al, 2003, cancer Cell, volume 4, pages 263-275), picornaviruses (e.g., seca valley virus; SW-001 and NTX-010), coxsackie virus or parvovirus.
In certain embodiments, the oncolytic viruses of the present disclosure are derived from poxviruses. The term "poxvirus" as used in this patent refers to a virus belonging to the subfamily poxviridae. In certain embodiments, the poxvirus is a virus belonging to the subfamily vertebrate poxviridae. In certain embodiments, the poxvirus is a virus belonging to the subfamily orthopoxvirus. The sequences of the genomes of various poxviruses (e.g., vaccinia virus, canary pox virus, murine poxvirus, myxoma virus) are available in the art and in specialized databases such as Genbank (accession numbers nc_006998, nc_003663, nc_005309, nc_004105, nc_001132, respectively).
In certain embodiments, the oncolytic viruses of the present disclosure are derived from vaccinia virus. Vaccinia virus is a member of the poxviridae family, characterized by a double stranded DNA genome of about 190kb, which encodes a number of viral enzymes and factors that enable replication of the virus independent of host cell mechanisms. In certain embodiments, the vaccinia virus of the present disclosure is derived from an Elstree strain, a Copenhagen strain, a Western Reserve strain, or a Wyeth strain. In certain embodiments, a vaccinia virus of the disclosure is a Western Reserve (WR) strain. The Western Reserve strain is well characterized and its complete sequence is available on the NCBI website under accession number AY243312 (www.ncbi.nlm.nih.gov).
The term "modified oncolytic virus" as used in this patent refers to oncolytic viruses that have been modified by the introduction of a heterologous nucleic acid or protein or by altering the native nucleic acid or protein. In certain embodiments, the modified oncolytic viruses provided herein are genetically altered by the deletion and/or addition of nucleic acid sequences. In certain embodiments, the modified oncolytic viruses provided herein comprise a deletion of the Thymidine Kinase (TK) gene. In certain embodiments, the modified oncolytic viruses provided herein comprise the addition of a nucleic acid sequence encoding an anti-human PD-1 antibody, a PD-1ECD-IgG1 Fc fusion protein, and/or a TGFBRII ECD-IgG1 Fc fusion protein.
In certain embodiments, the modified oncolytic viruses of the present disclosure are attenuated viruses. In certain embodiments, the modified oncolytic virus has reduced toxicity (e.g., at least 90%, 80%, 70%, 60%, 50%) or undetectable toxicity compared to its wild-type counterpart in a normal cell (e.g., a healthy cell).
The modified oncolytic viruses of the present disclosure may be derived from any oncolytic virus known in the art that is oncolytic due to its propensity to selectively replicate and kill tumor cells compared to non-tumor cells. Oncolytic viruses may be naturally oncolytic or may be made oncolytic by genetic engineering, such as by modifying one or more genes to increase tumor selectivity and/or preferential replication in tumor cells. Examples of such genes for modification include those involved in DNA replication, nucleic acid metabolism, host tropism, surface attachment, toxicity, host cell lysis, and viral transmission (see, e.g., kim et al, 2001, nat. Med., volume 7: page 781; wong et al, 2010, viruses, volume 2: pages 78-106).
In certain embodiments, the viral genome of the modified oncolytic viruses of the present disclosure comprises at least one deletion or disruption, such that the virus is capable of selective replication in tumor cells. For example, a deletion or disruption may reduce the expression or function of an enzyme critical for viral replication such that the ability of the virus to replicate in the absence of such an enzyme is reduced. In some embodiments, viral replication is dependent on the presence and/or level of such enzymes in the cell, the higher the level of enzymes, the more replication capacity or replication rate the virus.
In certain embodiments, the deletion or disruption is in an Open Reading Frame (ORF). The term "open reading frame" or "ORF" or "coding sequence" as used in this patent refers to a DNA sequence capable of translation into an amino acid sequence. The ORF typically begins with a start codon (e.g., ATG), followed by an amino acid encoding codon, and ends with a stop codon (e.g., TGA, TAA, TAG).
In certain embodiments, the ORF encodes at least a portion of an enzyme that is critical for replication of the virus and is preferentially expressed in tumor cells over non-tumor cells. The term "expression" as used in this patent refers to a process in which a protein or peptide sequence is produced from its coding DNA or RNA sequence. In certain embodiments, the enzyme is a kinase.
In certain embodiments, the deletion in the ORF constitutes 100%, more than 99%, more than 98%, more than 95%, more than 90%, more than 85%, more than 80%, more than 75%, more than 70%, more than 65%, more than 60%, more than 55%, more than 50%, more than 45%, more than 40%, more than 35%, more than 30%, more than 25%, more than 20%, more than 15%, or more than 10% of the full length of the ORF. In certain embodiments, the deletions in the ORF constitute at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 300, 500, 800, 1000, 1200, 1500, 1800, 2000, 2200, 2400, 2500 or more nucleotides (optionally contiguous).
In certain embodiments, the ORF for Thymidine Kinase (TK) is deleted or disrupted. TK is involved in the synthesis of deoxyribonucleotides. TK is necessary for the replication of the virus in normal cells, as these cells usually have low concentrations of nucleotides, whereas it is not necessary in tumor cells containing high concentrations of nucleotides. In poxviruses, the thymidine kinase encoding gene is located at the locus J2R. In certain embodiments, TK is completely absent.
In certain embodiments, the ORF of Ribonucleotide Reductase (RR) is deleted or disrupted. RR catalyzes the reduction of ribonucleotides to deoxyribonucleotides, a key step in DNA biosynthesis. The viral enzyme consists of two heterologous subunits, designated R1 and R2, encoded by the I4L locus and the F4L locus, respectively. The sequences of the I4L gene and F4L gene and their positions in the genomes of various poxviruses can be obtained in public databases, for example by GenBank accession numbers DQ437594, DQ437593, DQ377804, AH015635, AY313847, AY313848, nc_003391, nc_003389, nc_003310, M-35027, AY243312, DQ011157, DQ011156, DQ011155, DQ011154, DQ011153, Y16780, X71982, AF438165, U60315, AF410153, AF380138, U86916, L22579, nc_006998, DQ121394 and nc_008291. In the context of the present invention, the I4L gene (encoding the R1 large subunit) or the F4L gene (encoding the R2 small subunit) or both may be deleted or disrupted.
In certain embodiments, the modified oncolytic virus viral genome further comprises additional deletions or disruptions that further increase the tumor specificity of the virus. In certain embodiments, the additional deletion or disruption is located in an ORF encoding at least a portion of a tumor specific protein that is preferentially or specifically expressed in tumor cells. A representative example of a tumor-specific protein is VGF. VGF is a secreted protein expressed early after infection of cells by a virus, whose function appears to be important for the transmission of the virus in normal cells. Another example is the A56R gene encoding hemagglutinin (Zhang et al, 2007, cancer Res., volume 67: pages 10038-10046). Another example is the F2L gene encoding viral dUTPase, which is involved in both maintaining the accuracy of DNA replication and providing a precursor for TMP production by thymidylate synthase (Broyles et al, 1993, virol. Vol.195:863-865). The sequence of the vaccinia virus F2L gene is available in GenBank under accession number M25392.
The term "fused" or "fused" when used with respect to an amino acid sequence (e.g., a peptide, polypeptide, or protein) refers to the combination of two or more amino acid sequences into a single amino acid sequence that does not occur naturally, e.g., by chemical bonding or recombinant means. The fusion amino acid sequence may be produced by genetic recombination of two encoding polynucleotide sequences and may be expressed by introducing a construct comprising the recombinant polynucleotide into a host cell.
The term "heterologous" as used in this patent means that the sequence is not endogenous to the wild-type virus.
The term "encode" or "encode … …" as used in this patent refers to a polypeptide that is capable of transcription into mRNA and/or translation into a peptide or protein.
PD-1 extracellular Domain (PD-1 ECD)
In certain embodiments, the first molecule is a first fusion protein and the second molecule is a second fusion protein. In certain embodiments, the first fusion protein is capable of binding PD-L1. In certain embodiments, the first fusion protein is capable of blocking PD-L1 function in a tumor cell.
The term "PD-1" as used in this patent refers to a programmed cell death protein that belongs to the immunoglobulin superfamily and serves as a co-inhibitory receptor for down-regulation of the immune system. PD-1 is a member of the CD28/CTLA-4 family and has two known ligands, including PD-L1 and PD-L2. Representative amino acid sequences of human PD-1 are identified in GenBank accession numbers: np_005009.2, and a representative nucleic acid sequence encoding human PD-1 is disclosed in GenBank accession No.: shown under NM 005018.3.
The term "PD-1" as used in this patent is intended to include any form of PD-1 that retains useful activity, e.g., 1) naturally unprocessed PD-1 molecules, "full length" PD-1 chains, or naturally occurring variants of PD-1, including, e.g., splice variants or allelic variants; 2) Any form of PD-1 resulting from processing in a cell; or 3) full length forms, fragment forms (e.g., truncated forms, extracellular domain/transmembrane domain) or modified forms (e.g., mutant forms, glycosylated/pegylated forms, his-tag/immunofluorescent fusion forms) of PD-1 subunits produced by recombinant methods. Preferably, the PD-1 polypeptides as described in this patent, as well as protein complexes or fusion proteins comprising said polypeptides, are soluble.
Ligands for PD-1: PD-L1 (also known as B7-H1/CD 274) is expressed on antigen presenting cells and tumor cells. Binding of PD-L1 to PD-1, such as B7 to CD28, inhibits T cell activation and counteracts T cell stimulation signals. PD-L1 is not expressed by normal epithelial tissue, but it is abnormally expressed in a wide range of human cancers. In this context, activation of PD-L1 signaling may promote cancer progression by disabling host anti-tumor responses. Its expression on tumor cells is associated with a poor prognosis in renal cell carcinoma, lung carcinoma, breast carcinoma, pancreatic carcinoma, ovarian carcinoma, urothelial carcinoma, gastric carcinoma, esophageal carcinoma, colon carcinoma, pharyngeal squamous cell carcinoma, ovarian teratoma and hepatocellular carcinoma. Representative amino acid sequence of human PD-L1 is identified in NCBI accession number: np_054862.1, and a representative nucleic acid sequence encoding human PD-L1 is disclosed under NCBI accession No.: shown under NM 014143.3.
Binding of the PD-1 ECDs of the present disclosure to PD-L1 inhibits PD-L1 function, such as reducing the activity of PD-L1 by at least 5%, 10%, 20%, 40%, 50%, 80%, 90%, 95% or more.
The activity or function (e.g., of PD-L1) may be reduced due to, for example, inhibition of binding between the functional protein and its ligand (e.g., binding between PD-1 and PD-L1), inhibition of its biological activation (e.g., activation of PD-L1), and/or reduction of the level (e.g., PD-L1 level).
In certain embodiments, the first fusion protein comprises a PD-1 protein truncation capable of specifically binding to PD-L1. In certain embodiments, the PD-1 protein truncations comprise the extracellular domain of a PD-1 protein (PD-1 ECD). In certain embodiments, the PD-1ECD is residues 22-170 of the PD-1 protein ECD (PD-1 ECD G22-170) having the amino acid sequence of SEQ ID NO. 1, corresponding to the nucleic acid sequence of SEQ ID NO. 7.
PD-1ECD G22-170 amino acid sequence (SEQ ID NO: 1)
PD-1ECD G22-170 nucleic acid sequence (SEQ ID NO: 7)
As used in this patent, the term "specific binding" or "specifically binding" refers to a non-random binding reaction between two molecules, such as, for example, a non-random binding reaction between an antibody and an antigen. In certain embodiments, the antibodies or antigen binding fragments provided herein specifically bind to human and/or monkey PD-1 with a binding affinity (KD) of 10-6M (e.g., 5X10-7M, 2X10-7M, 5X10-8M, 2X10-8M, 5X10-9M, 2X10-9M, 10-10M). KD as used in this patent refers to the ratio of dissociation rate to association rate (koff/kon) which can be determined using surface plasmon resonance methods, for example using an instrument such as Biacore.
The term "identity" as used in this patent with respect to an amino acid sequence (or nucleic acid sequence) refers to the percentage of amino acid (or nucleic acid) residues in a candidate sequence that are identical to amino acid (or nucleic acid) residues in a reference sequence after aligning the sequences and introducing gaps, if necessary, to reach the maximum number of identical amino acids (or nucleic acids). Conservative substitutions of amino acid residues are not considered to be identical residues. To determine the percent amino acid (or Nucleic acid) sequence identity, an alignment can be accomplished, for example, using a publicly available tool such as BLASTN, BLASTp (available on the website of the National Center for Biotechnology Information (NCBI), see also Altschul S.F. et al, J.mol.biol., volume 215: pages 403-410 (1990); stephen F. Et al, nucleic Acids Res., volume 25: pages 3389-3402 (1997)), clustalW2 (available on the website of the European Bioinformatics institute, see also Higgins D.G. et al, methods in Enzymology, volume 266: pages 383-402 (1996); larkin M.A. et al, bioinformation (Oxford, england), volume 23: pages 21: 2947-2948 (2007)), and ALIGN or Megalign (DNASTAR) software. The person skilled in the art may use default parameters provided by the tool or may customize parameters suitable for alignment, such as by selecting an appropriate algorithm.
In certain embodiments, the PD-1ECD has the amino acid sequence of SEQ ID NO. 1 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the PD-1ECD is encoded by the nucleic acid sequence of SEQ ID NO. 7 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
anti-PD-1 antibodies
In certain embodiments, the first molecule is an anti-PD-1 antibody and the second molecule is a second fusion protein.
The term "antibody" as used in this patent includes any immunoglobulin, monoclonal antibody, polyclonal antibody, multispecific antibody, or bispecific (bivalent) antibody that binds a particular antigen. A natural intact antibody comprises two heavy chains and two light chains. Each heavy chain consists of a variable region and a first, second and third constant region, while each light chain consists of a variable region and a constant region. Mammalian heavy chains are classified as α, δ, ε, γ and p, while mammalian light chains are classified as λ or κ. The antibody has a "Y" shape, and the backbone of Y consists of a second constant region and a third constant region of two heavy chains bonded together via disulfide bonds. Each arm of Y comprises a variable region and a first constant region of a single heavy chain joined to a variable region and a constant region of a single light chain, wherein the first constant region of the heavy chain is linked to the second constant region by a hinge region. The variable regions of the light and heavy chains are responsible for antigen binding specificity. The variable region in both chains typically contains three highly variable loops, known as Complementarity Determining Regions (CDRs) (light (L) chain CDRs, including LCDR1, LCDR2 and LCDR3, and heavy (H) chain CDRs, including HCDR1, HCDR2 and HCDR 3). The CDR boundaries of antibodies and antigen binding fragments disclosed in this patent can be defined or divided according to the convention of Rabat, chothia or Al-Lazikani (see for details Al-Lazikani, B., chothia, C., lesk, A.M., J.Mol.Biol., vol.273, p.927 (1997)), chothia, C., et Al, J Mol biol., vol.186, vol.3, pp.651-663 (12, 5, 1985), chothia, C., and Lesk, A.M., J.Mol.Biol., vol.196, p.901 (1987)), chothia, C., et Al, nature, vol.342, 6252, pp.877-883 (21-28, 1989), rabat E.A. et Al, national Institutes of Health, bethesda, md. (1991)). The three CDRs are inserted between flanking stretches called Framework Regions (FR), which are more highly conserved than the CDRs and form a scaffold to support the structure of the variable region. The constant regions of the heavy and light chains are not associated with antigen binding specificity, but exhibit various effector functions. Antibodies are classified according to the amino acid sequence of the heavy chain constant region of the antibody. The five main classes or isotypes of antibodies are IgA, igD, igE, igG and IgM, characterized by the presence of α, δ, ε, γ and μ heavy chains, respectively. Several of the main antibody classes are divided into subclasses, such as IgG1 (gamma 1 heavy chain), igG2 (gamma 2 heavy chain), igG3 (gamma 3 heavy chain), igG4 (gamma 4 heavy chain), igA1 (alpha 1 heavy chain) or IgA2 (alpha 2 heavy chain).
The term "antigen-binding fragment" as used in this patent refers to an antibody fragment formed from a portion of an antibody that comprises one or more CDRs but does not comprise the complete antibody structure. Examples of antigen binding fragments include, but are not limited to, fab ', F (ab') 2, fv fragments, single chain antibody molecules (scFv), scFv dimers, camelbody single domain antibodies, and nanobodies. The antigen binding fragment is capable of binding to the same antigen to which the parent antibody binds.
The term "antigen-binding fragment" as used in this patent refers to an antibody fragment formed from a portion of an antibody that comprises one or more CDRs but does not comprise the complete antibody structure. Examples of antigen binding fragments include, but are not limited to, fab ', F (ab') 2, fv fragments, single chain antibody molecules (scFv), scFv dimers, camelbody single domain antibodies, and nanobodies. The antigen binding fragment is capable of binding to the same antigen to which the parent antibody binds.
The term "Fab" as used in this patent refers to the portion of an antibody consisting of a single light chain (both variable and constant regions) that is bound by disulfide bonds to the variable and first constant regions of a single heavy chain.
The term "Fab'" as used in this patent refers to a Fab fragment comprising a portion of a hinge region.
The term "F (ab ') 2" as used in this patent refers to the dimer of Fab'.
The term "Fv" as used in this patent refers to an Fv fragment consisting of a variable region of a single light chain and a variable region of a single heavy chain.
The term "single chain Fv antibody" or "scFv" as used in this patent refers to an engineered antibody consisting of a light chain variable region and a heavy chain variable region linked to each other either directly or through a peptide linker sequence (see, e.g., huston JS et al, proc Natl Acad Sci USA, vol. 85: p. 5879 (1988)).
The term "scFv dimer" as used in this patent refers to a polymer formed from two scFv.
The term "camelized single domain antibody", also known as "heavy chain antibody" or "HCAb" (heavy chain-only antibody), refers to an antibody that contains two heavy chain variable regions but no light chain (see, e.g., riechmann l. And Muyldermans s., J Immunol Methods, volume 231, stages 1-2, pages 25-38 (12, 10, 1999), muyldermans s., J biotechnol, volume 74, stages 4, pages 277-302 (6, 2001), WO94/04678, WO94/25591, and U.S. Pat. No. 6,005,079). Heavy chain antibodies were originally derived from camelidae (camel, dromedary and llama). In spite of the lack of light chains, camelized antibodies have a true antigen binding repertoire (see Hamers-Casterman C. Et al., nature, volume 363, 6428: pages 446-448 (1993); nguyen VK. et al, "Heavy-chain antibodies in Camelidae; a case of evolutionary innovation", immunogenetics, volume 54, page 1: pages 39-47 (2002); and Nguyen VK. et al, immunology, volume 109, page 1: pages 93-101 (2003), which are incorporated by reference in their entirety into this patent).
The term "nanobody" as used in this patent refers to an antibody consisting of a heavy chain variable region and two constant regions CH2 and CH3 from a chain antibody.
In certain embodiments, the antibodies provided herein are fully human, humanized, chimeric, mouse, or rabbit antibodies. In certain embodiments, the antibodies provided herein are polyclonal, monoclonal, or recombinant antibodies. In certain embodiments, the antibodies provided herein are monospecific antibodies, bispecific antibodies, or multispecific antibodies. In certain embodiments, the antibodies provided herein may be further labeled. In certain embodiments, the antibody or antigen-binding fragment thereof is a fully human antibody, which is optionally produced by a transgenic rat, e.g., a transgenic rat in which expression of endogenous rat immunoglobulin genes is inactivated and carries a recombinant human immunoglobulin locus with a J locus deletion and a C- κ mutation, and which may also be expressed by an engineered cell (e.g., CHO cell).
The term "fully human" as used in this patent with respect to an antibody or antigen binding fragment refers to an amino acid sequence of an antibody or antigen binding fragment that corresponds to an amino acid sequence of an antibody produced by a human or human immune cell, or an amino acid sequence derived from a non-human source such as a transgenic non-human animal utilizing a human antibody repertoire, or other human antibody coding sequence.
The term "humanized" as used in this patent with respect to an antibody or antigen binding fragment refers to an antibody or antigen binding fragment that comprises CDRs derived from a non-human animal, human-derived FR regions, and, when applicable, human-derived constant regions. Humanized antibodies or antigen binding fragments may be used as human therapeutics in certain embodiments because of their reduced immunogenicity. In certain embodiments, the non-human animal is a mammal, e.g., a mouse, rat, rabbit, goat, sheep, guinea pig, or hamster. In certain embodiments, the humanized antibody or antigen binding fragment consists of substantially all human sequences except the human CDR sequences.
The term "chimeric" as used herein with respect to an antibody or antigen-binding fragment refers to an antibody or antigen-binding fragment in which a portion of the heavy and/or light chain is derived from one species and the remainder of the heavy and/or light chain is derived from a different species. In certain embodiments, chimeric antibodies may comprise constant regions derived from humans and variable regions derived from non-human species, such as from mice or rabbits.
The term "conservative substitution" as used in this patent with respect to an amino acid sequence refers to the substitution of an amino acid residue with a different amino acid residue having a side chain with similar physiochemical properties. For example, conservative substitutions may be made between amino acid residues with hydrophobic side chains (e.g., met, ala, val, leu and Ile), between residues with neutral hydrophilic side chains (e.g., cys, ser, thr, asn and gin), between residues with acidic side chains (e.g., asp, glu), between amino acids with basic side chains (e.g., his, lys, and Arg), or between residues with aromatic side chains (e.g., trp, tyr, and Phe). As known in the art, conservative substitutions typically do not cause a significant change in the conformational structure of the protein, and thus may preserve the biological activity of the protein.
The CDR sequences of anti-PD-1 antibodies are shown in Table 1. Heavy chain variable region sequences and light chain variable region sequences are also provided in table 2 below.
TABLE 1 CDR amino acid sequences and nucleotide sequences
TABLE 2 amino acid and nucleotide sequences of the variable regions
/>
In certain embodiments, an anti-PD-1 antibody comprises:
HCDR1 having the amino acid sequence of SEQ ID No. 23 or a homologous sequence having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity;
HCDR2 having the amino acid sequence of SEQ ID No. 24 or a homologous sequence having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity;
HCDR3 having the amino acid sequence of SEQ ID No. 25 or a homologous sequence having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity;
LCDR1 having the amino acid sequence of SEQ ID No. 26 or a homologous sequence having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity;
LCDR2 having the amino acid sequence of SEQ ID No. 27 or a homologous sequence having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity; and
LCDR3 having the amino acid sequence of SEQ ID No. 28 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, HCDR1 is encoded by the nucleic acid sequence of SEQ ID NO. 31 or a homologous sequence thereof having at least 80% sequence identity,
HCDR2 is encoded by the nucleic acid sequence of SEQ ID NO. 32 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity,
HCDR3 is encoded by the nucleic acid sequence of SEQ ID NO. 33 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity,
LCDR1 is encoded by the nucleic acid sequence of SEQ ID NO. 34 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity,
LCDR2 is encoded by the nucleic acid sequence of SEQ ID NO. 35 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity, and
LCDR3 is encoded by the nucleic acid sequence of SEQ ID NO:36 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, an anti-PD-1 antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID No. 29 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity and a light chain variable region having the amino acid sequence of SEQ ID No. 30 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the anti-PD-1 antibody heavy chain variable region is encoded by the nucleic acid sequence of SEQ ID NO. 37 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity, and the anti-PD-1 antibody light chain variable region is encoded by the nucleic acid sequence of SEQ ID NO. 38 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, an anti-PD-1 antibody comprises a full length heavy chain having the amino acid sequence of SEQ ID NO. 21 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity and a full length light chain having the amino acid sequence of SEQ ID NO. 20 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the anti-PD-1 antibodies and fragments thereof provided herein further comprise an immunoglobulin constant region. In some embodiments, the immunoglobulin constant region comprises a heavy chain constant region and/or a light chain constant region. The heavy chain constant region comprises a CH1 region, a hinge region, and/or a CH2-CH3 region. In certain embodiments, the heavy chain constant region comprises an Fc region. In certain embodiments, the light chain constant region comprises ck or cλ.
In some embodiments, the anti-PD-1 antibodies and antigen-binding fragments thereof provided herein have an immunoglobulin (Ig), optionally a human Ig, optionally a constant region of human IgG. In certain embodiments, the anti-PD-1 antibodies and antigen-binding fragments thereof provided herein comprise a constant region of an IgG1 isotype that induces ADCC or CDC, or a constant region of an IgG4 or IgG2 isotype with reduced or depleted effector function. Effector function can be assessed using various assays such as Fc receptor binding assays, C1q binding assays, and cell lysis assays.
Table 3 shows the full length sequence of the anti-PD-1 antibodies provided by this patent.
TABLE 3 full-length amino acid and nucleotide sequences of anti-PD-1 antibodies
/>
/>
In certain embodiments, the full length heavy chain of an anti-PD-1 antibody is encoded by the nucleic acid sequence of SEQ ID NO. 39 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity, and the full length light chain of an anti-PD-1 antibody is encoded by the nucleic acid sequence of SEQ ID NO. 40 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
The binding of the anti-PD-1 antibodies and antigen-binding fragments thereof provided herein to PD-1 inhibits the interaction between PD-1 and PD-L1, thereby reducing the activity of PD-L1, e.g., by at least 5%, 10%, 20%, 40%, 50%, 80%, 90%, 95% or more.
The activity or function (e.g., of PD-L1) may be reduced due to, for example, inhibition of binding between a functional protein and its ligand (e.g., binding between an anti-PD-1 antibody and PD-1), inhibition of its biological activation (e.g., activation of PD-L1), and/or reduction of the level (e.g., PD-L1 level).
TGF-beta receptor extracellular domain (TGFBRII ECD)
In certain embodiments, the second molecule is a second fusion protein. In certain embodiments, the second fusion protein comprises a TGF- β receptor II extracellular domain (TGFBRII ECD).
The "transforming growth factor-beta" or "TGF-beta" superfamily is a protein composed of extracellular cytokines present in most human cells. TGF-beta refers to any protein in the TGF-beta family of proteins having any full length, native amino acid sequence of any TGF-beta from a subject (e.g., human), including latent forms and associative or non-associative complexes of precursor and mature TGF-beta. TGF-. Beta.has been shown to play a major role in the regulation of immune responses primarily through its inhibitory function on cells of the immune system. TGF- β inhibits antigen-specific T cell proliferation, at least by decreasing cell cycle rate, as opposed to inducing apoptosis. In particular, TGF- β acts on Cytotoxic T Lymphocytes (CTLs) to specifically inhibit expression of at least five cytolytic gene products important for CTL-mediated tumor cytotoxicity: perforin, granzyme a, granzyme B, fas ligand and interferon gamma (Thomas and Massague, cancer Cell, 11 month 2005, 8/5: pages 369-380). All about 40 TGF- β superfamily ligands share the same overall architecture, and each region of the protein has general features. Such TGF-beta referred to in this patent is understood to refer to any of the presently divided forms, including TGF-beta 1, TGF-beta 2, TGF-beta 3 isoforms and latent forms thereof, and to future divided human TGF-beta species, including polypeptides derived from and at least about 75%, preferably at least about 80%, more preferably at least about 85%, still more preferably at least about 90%, even more preferably at least about 95% homologous to any known TGF-beta sequence. The terms "transforming growth factor-beta", "transforming growth factor beta", "tgfβ", "TGF-beta" and "TGFB" are used interchangeably in this disclosure.
As used herein, the term "human TGF- β1" refers to a TGF- β1 protein encoded by a human TGFB1 gene (e.g., a wild-type human TGFB1 gene). An exemplary wild-type human tgfβ1 protein is provided under GenBank accession number np_ 000651.3. As used herein, the term "human TGF- β2" refers to a TGF- β2 protein encoded by a human TGFB2 gene (e.g., a wild-type human TGFB2 gene). Exemplary wild-type human TGF- β2 proteins are provided under GenBank accession numbers NP-001129071.1 and NP-003229.1. As used herein, the term "human TGF- β3" refers to a TGF- β3 protein encoded by a human TGFB3 gene (e.g., a wild-type human TGFB3 gene). Exemplary wild-type human TGF-beta 3 proteins are provided under GenBank accession numbers NP-003230.1, NP-001316868.1, and NP-001316867.1.
As used herein, the term "TGF- β receptor type II" or "TGFBRII" refers to the transforming growth factor- β receptor II (TGFBRII) protein family. Members of the TGFBRII family are typically transmembrane proteins, consisting of a ligand-binding extracellular domain with a cysteine-rich region, a transmembrane domain, and a cytoplasmic domain with predicted serine/threonine kinase activity. The nucleic acid sequence encoding the TGFBRII isoform a precursor protein is shown under Genbank reference sequence nm_001024847.2 and the amino acids sequenced for human TGFBRII are shown under Genbank accession No. np_ 001020018.1. The nucleic acid sequence encoding the TGFBRII isoform B precursor protein is shown under Genbank reference sequence nm_003242.6 and the amino acids sequenced for human TGFBRII are shown under Genbank accession No. np_ 003233.4. The nucleic acid sequence encoding the TGFBRII isoform X1 protein is shown under Genbank reference sequence xm_011534043.2 and the amino acids sequenced from human TGFBRII are shown under Genbank accession number xp_ 011532345.1. The nucleic acid sequence encoding the TGFBRII isoform X2 protein is shown under Genbank reference sequence xm_011534045.3 and the amino acids sequenced from human TGFBRII are shown under Genbank accession number xp_ 011532347.1. The nucleic acid sequence encoding the TGFBRII isoform X2 protein is shown under Genbank reference sequence xm_017007106.1 and the amino acids sequenced from human TGFBRII are shown under Genbank accession number xp_ 016862595.1. The expressed TGFBRII polypeptide lacks a signal sequence.
The term "TGF- β receptor type II" or "TGFBRII" as used in this patent is intended to include any form of TGF- β receptor type II that retains useful activity, e.g., 1) naturally unprocessed TGF- β receptor type II molecules, "full length" TGF- β receptor type II chains, or naturally occurring variants of TGF- β receptor type II, including, e.g., splice variants or allelic variants; 2) Any form of TGF- β receptor type II that results from processing in a cell; or 3) full length forms, fragment forms (e.g., truncated forms, extracellular domain/transmembrane domain) or modified forms (e.g., mutant forms, glycosylated/pegylated forms, his tag/immunofluorescent fusion forms) of TGF-beta receptor type II subunits produced by recombinant methods. Preferably, a TGF-beta type II polypeptide as described in this patent is soluble in a protein complex or fusion protein comprising the polypeptide.
Binding of TGFBRII of the present disclosure to TGF- β inhibits TGF- β function, such as reducing activity of TGF- β by at least 5%, 10%, 20%, 40%, 50%, 80%, 90%, 95% or more.
The activity or function (e.g., of TGF- β) may be reduced due to, for example, inhibition of binding between a functional protein and its ligand (e.g., binding between TGFBRII and TGF- β), inhibition of its biological activation (e.g., activation of TGF- β), and/or reduction of the level (e.g., of TGF- β).
In certain embodiments, the second fusion protein comprises a TGFBRII protein truncation capable of specifically binding to TGF- β. In certain embodiments, the TGFBRII protein truncates an extracellular domain comprising TGFBRII protein (TGFBRII ECD). In certain embodiments, TGFBRII ECD is residues 23-166 of TGFBRII ECD protein (TGFBRII ECD G23-166) having the amino acid sequence of SEQ ID NO. 2, corresponding to the nucleic acid sequence of SEQ ID NO. 8.
The amino acid sequence of TGFBRII ECD G23-166 (SEQ ID NO: 2)
TGFBRII ECD G23-166 nucleic acid sequence (SEQ ID NO: 8)
In certain embodiments, the TGFBRII ECD has the amino acid sequence of SEQ ID NO. 2 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the TGFBRII ECD is encoded by the nucleic acid sequence of SEQ ID NO. 8 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
Immunoglobulin Fc region
In certain embodiments, the first fusion protein further comprises an immunoglobulin Fc region.
In certain embodiments, the second fusion protein further comprises an immunoglobulin Fc region.
As used herein, an immunoglobulin "Fc" region refers to a portion of an antibody that consists of a second constant region and a third constant region of a first heavy chain that are bound to the second constant region and the third constant region of a second heavy chain via disulfide bonding. The Fc portion of antibodies is responsible for various effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), but does not play a role in antigen binding.
In certain embodiments, the first fusion protein comprises a first immunoglobulin Fc region.
In certain embodiments, the second fusion protein comprises a second immunoglobulin Fc region.
In certain embodiments, the first immunoglobulin Fc region or the second immunoglobulin Fc region comprises one or more amino acid substitutions that improve pH-dependent binding to neonatal Fc receptor (FcRn). This variant may have an extended pharmacokinetic half-life because it binds FcRn at acidic pH, which allows it to escape degradation in lysosomes, and then translocate and be released from the cell.
In certain embodiments, the first immunoglobulin Fc region or the second immunoglobulin Fc region comprises one or more amino acid substitutions that alter Antibody Dependent Cellular Cytotoxicity (ADCC). Certain amino acid residues at the CH2 domain of the Fc region may be substituted to provide enhanced ADCC activity. Alternatively or in addition, the carbohydrate structure on the antibody may be altered to enhance ADCC activity. In certain embodiments, the first immunoglobulin Fc region or the second immunoglobulin Fc region comprises one or more amino acid substitutions that alter Complement Dependent Cytotoxicity (CDC), e.g., by increasing or decreasing C1q binding and/or CDC.
When the first heterologous polynucleotide encodes a first fusion protein and the second heterologous polynucleotide encodes a second fusion protein, the first immunoglobulin Fc region and the second immunoglobulin Fc region are capable of binding into dimers, for example, via formation of a knob-to-socket structure, hydrophobic interactions, electrostatic interactions, hydrophilic interactions, or increased flexibility.
In certain embodiments, the first immunoglobulin Fc region or the second immunoglobulin Fc region comprises one or more amino acid substitutions in the interface of the Fc regions to facilitate/promote heterodimerization. Such modifications include introducing a protuberance into a first Fc polypeptide and introducing a cavity into a second Fc polypeptide (or introducing a cavity into a first Fc polypeptide and introducing a protuberance into a second Fc polypeptide), wherein the protuberance may be positioned in the cavity so as to facilitate interaction of the first Fc polypeptide and the second Fc polypeptide to form a heterodimer or complex.
The PD-1ECD of the present disclosure is operably linked to the first immunoglobulin Fc region at the C-terminus of the PD-1 ECD. The TGFBRII ECD of the present disclosure is operably linked to a second immunoglobulin Fc region at the C-terminus of the TGFBRII ECD.
The PD-1ECD or TGFBRII ECD of the present disclosure may be directly or indirectly linked to the first immunoglobulin Fc region or the second immunoglobulin Fc region, respectively. In certain embodiments, the PD-1ECD is linked to the first immunoglobulin Fc region through a linker. In certain embodiments, the TGFBRII ECD is linked to the second immunoglobulin Fc region through a linker. In certain embodiments, the linker is a polypeptide linker. In certain embodiments, the linker is a bifunctional cross-linking agent, such as disuccinimidyl glutarate or 1-ethyl-3- [ 3-dimethylaminopropyl ] carbodiimide hydrochloride, which may be linked at one end to the amino group of the first polypeptide and at the other end to the carboxy terminus of the second polypeptide. The skilled artisan can select an appropriate linker from among those known in the art, provided that the linked fusion protein retains sufficient biological activity.
In certain embodiments, the first immunoglobulin Fc region or the second immunoglobulin Fc region is derived from a human IgG Fc region. In certain embodiments, the first immunoglobulin Fc region and the second immunoglobulin Fc region are identical. In certain embodiments, the human IgG Fc region is a human IgG1, igG2, igG3, or IgG4 Fc region.
In certain embodiments, the first immunoglobulin Fc region and the second immunoglobulin Fc region are human IgG1Fc regions and have the amino acid sequence of SEQ ID NO:3 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
Amino acid sequence of human IgG1Fc region (SEQ ID NO: 3)
In certain embodiments, the human IgG1Fc region is encoded by the nucleic acid sequence of SEQ ID NO 9 or 10 or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
Nucleic acid sequence of human IgG1Fc region (SEQ ID NO: 9)
The nucleic acid sequence of the Fc region of human IgG1 (SEQ ID NO: 10),
In certain embodiments, the PD-1ECD is operably linked to a human IgG1Fc region at the C-terminus of the PD-1 ECD. In certain embodiments, the human IgG1Fc region is encoded by the nucleic acid sequence of SEQ ID NO. 9.
In certain embodiments, the TGFBRII ECD is operably linked to a human IgG1 Fc region at the C-terminus of the TGFBRII ECD. In certain embodiments, the human IgG1 Fc region is encoded by the nucleic acid sequence of SEQ ID NO. 10.
The term "operably linked" refers to two or more biological sequences of interest being juxtaposed with or without a spacer or linker in a manner such that they are in a relationship that allows them to function in the intended manner. When used with respect to a polypeptide, it is intended to mean that the polypeptide sequences are linked in a manner that allows the linked product to have the intended biological function. The term may also be used with respect to polynucleotides. For example, when a polynucleotide encoding a polypeptide is operably linked to a regulatory sequence (e.g., a promoter, enhancer, silencer sequence, etc.), it is intended to mean that the polynucleotide sequences are linked in a manner that allows the polypeptide to regulate expression from the polynucleotide.
Signal peptides
Transport of the protein may be mediated by a signal peptide located at the amino terminus of the protein itself. The signal peptide consists of about ten to twenty hydrophobic amino acids, which target nascent proteins from the ribosome to specific membrane-bound compartments such as the Endoplasmic Reticulum (ER). The ER-targeting proteins may be carried out either by the secretory pathway or may reside in any secretory organelle such as the ER, golgi apparatus or lysosomes. Proteins transported by the secretory pathway are either secreted into the extracellular space or remain in the plasma membrane. Secreted proteins are typically synthesized as inactive precursors that are activated by post-translational processing events during transport through the secretory pathway. These events include glycosylation, phosphorylation, proteolysis, and removal of signal peptides by signal peptidases. Other events that may occur during protein transport include chaperone-dependent unfolding and folding of nascent proteins and interactions of proteins with receptors or pore complexes.
In certain embodiments, the first fusion protein further comprises a signal peptide. In certain embodiments, the signal peptide of the first fusion protein is operably linked to the PD-1ECD at the C-terminus of the signal peptide.
In certain embodiments, the second fusion protein further comprises a signal peptide. In certain embodiments, the signal peptide of the second fusion protein is operably linked to TGFBRII ECD at the C-terminus of the signal peptide.
In certain embodiments, the signal peptide of the first fusion protein is a CD33 signal peptide having the amino acid sequence of SEQ ID No. 4 (MPLLLLLPLLWAGALAM) or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the signal peptide of the second fusion protein is a CD33 signal peptide having the amino acid sequence of SEQ ID No. 4 (MPLLLLLPLLWAGALAM) or a homologous sequence thereof having at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the CD33 signal peptide consists of SEQ ID NO. 11
(ATGCCACTGCTCCTCCTGCTGCCACTGCTCTGGGCCGGCGCCCTCG CTATG)
Or SEQ ID NO. 12
(ATGCCTCTGCTGCTGCTGCTGCCTCTGCTGTGGGCCGGCGCCCTGG CCAT G) or a homologous sequence encoding at least 80%, 85%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In certain embodiments, the CD33 signal peptide is operably linked to TGFBRII ECD at the C-terminus of the CD33 signal peptide. In certain embodiments, the CD33 signal peptide is encoded by the nucleic acid sequence of SEQ ID NO. 11.
In certain embodiments, the CD33 signal peptide is operably linked to TGFBRII ECD at the C-terminus of the CD33 signal peptide. In certain embodiments, the CD33 signal peptide is encoded by the nucleic acid sequence of SEQ ID NO. 12.
Polynucleotide
In certain embodiments, the modified oncolytic viruses of the present disclosure comprise a viral genome having a first heterologous polynucleotide encoding a first fusion protein capable of binding to PD-L1 and a second heterologous polynucleotide encoding a second fusion protein capable of binding to TGF- β.
The term "polynucleotide" or "nucleic acid" as used in this patent refers to ribonucleic acid (RNA), deoxyribonucleic acid (DNA), or mixed ribonucleic acid-deoxyribonucleic acid such as DNA-RNA hybrids. The polynucleotide or nucleic acid may be single-or double-stranded DNA or RNA or DNA-RNA hybrids. The polynucleotide or nucleic acid may be linear or circular. In certain embodiments, wherein when the virus is a DNA virus, both the first heterologous polynucleotide and the second heterologous polynucleotide are DNA, or when the virus is an RNA virus, both the first heterologous polynucleotide and the second heterologous polynucleotide are RNA. In certain embodiments, the first heterologous polynucleotide and the second heterologous polynucleotide are both double stranded DNA. The polynucleotides of the present disclosure are double stranded DNA and the nucleic acid sequences are represented by coding sequences such as those shown in SEQ ID NOS 7-19, 22 and 31-40.
The first heterologous polynucleotide and the second heterologous polynucleotide can be introduced into the modified oncolytic virus using conventional methods known in the art, for example synthesized by Polymerase Chain Reaction (PCR) and ligated to a viral genome having compatible restriction ends. See, for example, sambrook et al, molecular Cloning: ALaboratory Manual (Cold Spring Harbor Laboratory, N.Y. (1989)), which manual is incorporated by reference in its entirety.
In certain embodiments, the first heterologous polynucleotide and the second heterologous polynucleotide are introduced in place of the deletion in the ORF. In certain embodiments, the first heterologous polynucleotide is immediately upstream or immediately downstream of the second heterologous polynucleotide. The term "immediately upstream or immediately downstream" as used in this patent means that the first heterologous polynucleotide and the second heterologous polynucleotide are positioned sufficiently close on the viral genome that they are no more than 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides apart from each other. For example, if the 3 'end of an upstream polynucleotide is separated from the 5' end of a downstream polynucleotide by no more than 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, the 3 'end of the upstream polynucleotide is immediately adjacent to the 5' end of the downstream polynucleotide. In certain embodiments, there is no ORF between the first heterologous polynucleotide and the second heterologous polynucleotide. In certain embodiments, a restriction site exists between the first heterologous polynucleotide and the second heterologous polynucleotide.
In certain embodiments, the first heterologous polynucleotide encodes a first molecule that is a first fusion protein and the second heterologous polynucleotide encodes a second molecule that is a second fusion protein. In certain embodiments, the first heterologous polynucleotide further comprises a first promoter capable of driving expression of the first fusion protein. In certain embodiments, the second heterologous polynucleotide further comprises a second promoter capable of driving expression of the second fusion protein. In certain embodiments, the first heterologous polynucleotide and the second heterologous polynucleotide are arranged in opposite directions with respect to protein translation. In certain embodiments, the first promoter and the second promoter are in a head-to-head orientation.
The term "head-to-head orientation" as used in this patent refers to two promoters that are directly adjacent to each other on the viral genome and that drive protein expression in opposite directions. Illustrative examples are shown in fig. 1 and 4.
The term "promoter" as used in this patent refers to a polynucleotide sequence that controls transcription of a coding sequence. Promoter sequences include specific sequences sufficient for RNA polymerase recognition, binding, and transcription initiation. In addition, the promoter sequence may include sequences that regulate such recognition, binding, and transcription initiation activity of the RNA polymerase. Promoters can affect the transcription of genes located on the same nucleic acid molecule as themselves or on different nucleic acid molecules than themselves. Depending on the nature of the regulation, the function of the promoter sequence may be constitutive or stimulus-inducible. "constitutive" promoter as used in this patent refers to a promoter that functions to continuously activate expression of a gene in a host cell. An "inducible" promoter as used in this patent refers to a promoter that activates gene expression in a host cell in the presence of certain stimuli.
In certain embodiments, the promoters of the present disclosure are eukaryotic promoters, such as promoters from CMV (e.g., CMV immediate early promoter (CMV promoter)), epstein-Barr virus (EBV) promoters, human Immunodeficiency Virus (HIV) promoters (e.g., HIV Long Terminal Repeat (LTR) promoters), moloney virus promoters, mouse Mammary Tumor Virus (MMTV) promoters, rous Sarcoma Virus (RSV) promoters, SV40 early promoters, promoters from human genes, such as the human myoglobin promoter, the human hemoglobin promoter, the human muscle creatine promoter, the human metallothionein beta-actin promoter, the human ubiquitin C (UBC) promoter, the mouse phosphoglycerate kinase (PGK) 1 promoter, the human Thymidine Kinase (TK) promoter, the human elongation factor 1 alpha (EF 1A) promoter, the cauliflower mosaic virus (CaMV) 35S promoter, the E2F-1 promoter (E2F 1 transcription factor 1 promoter), the alpha-fetoprotein promoter, the cholecystokinin promoter, the carcinoembryonic antigen promoter, the C-erbB2/neu oncogene promoter, the cyclooxygenase gene promoter, the CXC chemokine receptor 4 (CXCR 4) promoter, the human epididymal protein 4 (HE 4) promoter, the hexokinase type II promoter, the L-wire bundle protein (L-stin) promoter, the mucin-like glycoprotein (MUC 1) promoter, the Prostate Specific Antigen (PSA) promoter, the survivin promoter, tyrosinase related protein (TRP 1) promoter and tyrosinase promoter.
In certain embodiments, the promoters of the present disclosure may be tumor-specific promoters. The term "tumor-specific promoter" as used in this patent refers to a promoter that is used to preferentially or exclusively activate gene expression in tumor cells, and that is inactive or has reduced activity in non-tumor cells. Illustrative examples of tumor specific promoters include, but are not limited to, E2F-1 promoter, alpha fetoprotein promoter, cholecystokinin promoter, carcinoembryonic antigen promoter, C-erbB2/neu oncogene promoter, cyclooxygenase promoter, CXCR4 promoter, HE4 promoter, type II hexokinase promoter, L-silk bundle protein promoter, MUC1 promoter, PSA promoter, survivin promoter, TRP1 promoter, and tyrosinase promoter.
In certain embodiments, the first promoter and the second promoter are the same or different. In certain embodiments, the first promoter and the second promoter are both early and late promoters. In certain embodiments, the early and late promoters are pSE/L. In certain embodiments, the pSE/L promoter has the nucleic acid sequence of SEQ ID NO. 15 (AAAAATTGAAATTTTATTTTTTTTTTTTGGAATATAAATAAG).
In certain embodiments, the first heterologous polynucleotide further comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: first promoter-polynucleotide encoding first fusion protein-first stop codon. The second heterologous polynucleotide further comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: second promoter-polynucleotide encoding a second fusion protein-second stop codon. In certain embodiments, the first heterologous polynucleotide is immediately upstream or immediately downstream of the second heterologous polynucleotide.
In certain embodiments, the first stop codon and the second stop codon are the same or different. In certain embodiments, the first stop codon and the second stop codon have the nucleic acid sequence of SEQ ID NO. 16 (TTTTTNT, where N is A, T, C or G).
In certain embodiments, the first fusion protein expressed from the first heterologous polynucleotide and the second fusion protein expressed from the second heterologous polynucleotide are expressed as separate proteins. In other words, they are not expressed as fusion proteins, nor are they linked to each other (whether covalently linked or via a linker).
In certain embodiments, the first heterologous polynucleotide and the second heterologous polynucleotide are configured such that they are expressed in the same or different phases of the replication cycle of the modified oncolytic virus. For example, both polynucleotides may be driven by an early promoter that is induced early in viral replication; or by a late promoter induced late in viral replication; or one driven by an early promoter and the other driven by a late promoter.
In certain embodiments, the modified oncolytic virus does not comprise any other heterologous polynucleotide encoding a protein in addition to the first heterologous polynucleotide and the second heterologous polynucleotide.
In certain embodiments, the first heterologous polynucleotide encodes a first molecule that is an anti-PD-1 antibody, and the second heterologous polynucleotide encodes a second molecule that is a second fusion protein. In certain embodiments, the first heterologous polynucleotide further comprises a third heterologous polynucleotide and a fourth heterologous polynucleotide. In certain embodiments, the third heterologous polynucleotide further comprises a third promoter capable of driving expression of the heavy chain of the anti-PD-1 antibody. In certain embodiments, the fourth heterologous polynucleotide further comprises a fourth promoter capable of driving expression of the anti-PD-1 antibody light chain.
In certain embodiments, the third heterologous polynucleotide further comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: third promoter-polynucleotide encoding heavy chain of anti-PD-1 antibody-third stop codon. In certain embodiments, the fourth heterologous polynucleotide further comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: fourth promoter-polynucleotide encoding anti-PD-1 antibody light chain-fourth stop codon. In certain embodiments, the third promoter and the fourth promoter are in a head-to-head orientation.
In certain embodiments, the second heterologous polynucleotide further comprises a second promoter capable of driving expression of the second fusion protein. In certain embodiments, the second heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: second promoter-polynucleotide encoding a second fusion protein-second stop codon.
In certain embodiments, the first heterologous polynucleotide comprising a third heterologous polynucleotide and a fourth heterologous polynucleotide therein is immediately upstream or immediately downstream of the second heterologous polynucleotide. In certain embodiments, the third heterologous polynucleotide is immediately upstream or immediately downstream of the fourth heterologous polynucleotide.
In certain embodiments, the second promoter, the third promoter, and the fourth promoter are the same or different. In certain embodiments, the second promoter, the third promoter, and the fourth promoter are all early and late promoters. In certain embodiments, the early and late promoters are pSE/L. In certain embodiments, the pSE/L promoter has the nucleic acid sequence of SEQ ID NO. 15.
In certain embodiments, the second stop codon, the third stop codon, and the fourth stop codon are the same or different. In certain embodiments, the second stop codon, the third stop codon, and the fourth stop codon have the nucleic acid sequence of SEQ ID NO. 16.
In certain embodiments, the anti-PD-1 antibody heavy chain expressed by the third heterologous polynucleotide and the anti-PD-1 antibody light chain expressed by the fourth heterologous polynucleotide are expressed as separate proteins. In other words, they are not expressed as fusion proteins, nor are they linked to each other (whether covalently linked or via a linker).
In certain embodiments, the anti-PD-1 antibody expressed by the first heterologous polynucleotide comprising the third heterologous polynucleotide and the fourth heterologous polynucleotide and the second fusion protein expressed by the second heterologous polynucleotide are expressed as separate proteins. In other words, they are not expressed as fusion proteins, nor are they linked to each other (whether covalently linked or via a linker).
When the third heterologous polynucleotide and the fourth heterologous polynucleotide encode the heavy and light chain, respectively, of an anti-PD-1 antibody, the third heterologous polynucleotide, the fourth heterologous polynucleotide, and the second heterologous polynucleotide are arranged in sequence for protein translation.
In certain embodiments, the third heterologous polynucleotide, the fourth heterologous polynucleotide, and the second heterologous polynucleotide are configured such that they are expressed in the same or different phases of the replication cycle of the modified oncolytic virus. For example, the polynucleotide may be driven by an early promoter that is induced early in viral replication; or by a late promoter induced late in viral replication; or one driven by an early promoter and the other two driven by a late promoter; or one driven by the late promoter and the other two driven by the early promoter.
In certain embodiments, the modified oncolytic virus does not comprise any other heterologous polynucleotide encoding a protein other than the third heterologous polynucleotide, the fourth heterologous polynucleotide, and the second heterologous polynucleotide.
Pharmaceutical composition
In another aspect, the present disclosure provides a pharmaceutical composition comprising a modified oncolytic virus described in the present disclosure and a pharmaceutically acceptable carrier.
The term "pharmaceutically acceptable" as used in this patent is directed to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. In certain embodiments, pharmaceutically acceptable compounds, materials, compositions, and/or dosage forms refer to those compounds, materials, compositions, and/or dosage forms for animals, more particularly for humans, approved by a regulatory agency, such as the united states food and drug administration, chinese food and drug administration, or european medicines administration, or listed in a recognized pharmacopeia, such as the united states pharmacopeia, chinese pharmacopeia, or european pharmacopeia.
Pharmaceutically acceptable carriers for pharmaceutical compositions of the invention can include, but are not limited to, for example, pharmaceutically acceptable liquids, gels or solid carriers, aqueous vehicles (e.g., sodium chloride injection, ringer's injection, isotonic dextrose injection, sterile water injection, or dextrose and lactate ringer's injection), non-aqueous vehicles (e.g., fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil, or peanut oil), antimicrobial agents, isotonic agents (such as sodium chloride or dextrose), buffers (such as phosphate or citrate buffers), antioxidants (such as sodium bisulfate), anesthetics (such as procaine hydrochloride), suspending/dispersing agents (such as sodium carboxymethylcellulose, hydroxypropyl methylcellulose or polyvinylpyrrolidone), chelating agents (such as EDTA (ethylenediamine tetraacetic acid) or EGTA (ethylene glycol tetraacetic acid)), emulsifying agents (such as polysorbate 80 (tween 80)), diluents, adjuvants, excipients or nontoxic auxiliary substances, other components known in the art, or various combinations thereof. Suitable components may include, for example, fillers, binders, disintegrants, buffers, preservatives, lubricants, flavouring agents, thickening agents, colouring agents or emulsifying agents.
In certain embodiments, the pharmaceutical composition is an oral formulation. Oral formulations include, but are not limited to, capsules, cachets, pills, tablets, lozenges (for taste bases, typically sucrose and acacia or tragacanth), powders, granules or aqueous or non-aqueous solutions or suspensions, or water-in-oil or oil-in-water emulsions, or elixirs or syrups, or candy lozenges (for inert bases such as gelatin and glycerin, or sucrose or acacia) and/or mouthwashes and the like.
In certain embodiments, the pharmaceutical composition may be an injectable formulation, including a sterile aqueous solution or dispersion, suspension or emulsion. In all cases, the injectable formulation should be sterile and should be liquid to facilitate injection. It should be stable under the conditions of manufacture and storage and should be resistant to infection by microorganisms such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycols, and the like), and suitable mixtures thereof and/or vegetable oils. The injectable formulation should maintain proper fluidity, which can be maintained in a variety of ways, for example, by the use of coatings such as lecithin, by the use of surfactants, and the like. Antimicrobial contamination can be achieved by the addition of various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like).
In certain embodiments, the unit dose parenteral formulations are packaged in ampules, vials or needled syringes. As known and practiced in the art, all formulations for parenteral administration should be sterile and not cause fever.
Application method
In another aspect, the present disclosure provides a use of a pharmaceutical composition for treating a tumor and a method of use comprising administering to a subject an effective amount of a modified oncolytic virus of the present disclosure or a pharmaceutical composition of the present disclosure.
The term "subject" as used in this patent refers to both human and non-human animals. Non-human animals include all vertebrates, such as mammals and non-mammals. The "subject" may also be a livestock animal (e.g., cow, pig, goat, chicken, rabbit, or horse), or a rodent (e.g., rat or mouse), or a primate (e.g., gorilla or monkey), or a domestic animal (e.g., dog or cat). The "subject" may be male or female, or may have different ages. In certain embodiments, the subject is a human. The human "subject" may be caucasian, african, asian, sumer or other race, or a mixed blood of different race. The human "subject" may be an elderly person, adult, adolescent, child or infant.
The term "tumor" as used in this patent refers to any medical condition mediated by neoplastic or malignant cell growth, proliferation or metastasis, including solid tumors and non-solid tumors such as leukemia. In the present disclosure, "tumor" is used interchangeably with the terms "cancer," malignant tumor, "" hyperproliferative, "and" neoplasm. The term "tumor cell" is interchangeable with the terms "cancer cell", "malignant cell", "hyperproliferative cell" and "neoplastic cell" unless explicitly stated otherwise. In certain embodiments, the tumor is selected from the group consisting of head and neck tumors, breast tumors, colorectal tumors, liver tumors, pancreatic adenocarcinoma, gall bladder and bile duct tumors, ovarian tumors, cervical tumors, small cell lung tumors, non-small cell lung tumors, renal cell carcinoma, bladder tumors, prostate tumors, bone tumors, mesothelioma, brain tumors, soft tissue sarcomas, uterine tumors, thyroid tumors, nasopharyngeal carcinoma, and melanoma. In certain embodiments, the tumor is a solid tumor. In certain embodiments, the tumor is melanoma, non-small cell lung cancer, renal cell carcinoma, hodgkin's lymphoma, head and neck squamous cell carcinoma, bladder cancer, colorectal cancer, triple negative breast cancer, or hepatocellular carcinoma. In certain embodiments, the tumor is pancreatic cancer, ovarian cancer, colon cancer, pharyngeal squamous cell carcinoma, ovarian teratoma. In certain embodiments, the tumor is refractory to prior treatment (e.g., administration of oncolytic viruses, immune checkpoint inhibitors, and/or immune activators alone).
The term "treating" a disorder as used herein includes preventing or alleviating the disorder, slowing the onset or rate of progression of the disorder, reducing the risk of developing the disorder, preventing or delaying the progression of symptoms associated with the disorder, alleviating or terminating symptoms associated with the disorder, causing complete or partial regression of the disorder, curing the disorder, or some combination thereof. With respect to a tumor, "treating" may refer to inhibiting or slowing the growth, proliferation, or metastasis of a neoplastic or malignant cell, preventing or delaying the progression of the growth, proliferation, or metastasis of a neoplastic or malignant cell, or some combination thereof. With respect to tumors, "treatment" includes eradicating all or part of the tumor, inhibiting or slowing the growth and metastasis of the tumor, preventing or delaying the progression of the tumor, or some combination thereof.
The modified oncolytic viruses and pharmaceutical compositions may be administered via any suitable route known in the art including, but not limited to, parenteral, oral, enteral, buccal, nasal, topical, rectal, vaginal, transmucosal, epidermal, transdermal, dermal, ocular, pulmonary, and subcutaneous routes of administration. In certain embodiments, the route of administration is topical. In certain embodiments, the route of administration is intratumoral injection.
In certain embodiments, the modified oncolytic viruses and pharmaceutical compositions are administered in a therapeutically effective dose. The term "therapeutically effective dose" as used in this patent refers to an amount of a drug that is capable of ameliorating or eliminating a disease or condition in a subject, or prophylactically inhibiting or preventing the occurrence of a disease or condition. A therapeutically effective amount may be an amount of a drug that ameliorates one or more diseases or symptoms of a subject to some extent; an amount of drug capable of partially or completely restoring one or more physiological or biochemical parameters associated with the etiology of the disease or condition to normal; and/or an amount of a drug capable of reducing the likelihood of occurrence of a disease or symptom.
The therapeutically effective dose of the modified oncolytic viruses and pharmaceutical compositions depends on various factors known in the art, such as, for example, body weight, age, pre-existing medical conditions, currently accepted treatment, the health of the subject, the strength of drug interactions, allergies, hyper-allergies and side effects, as well as the route of administration and the extent of disease progression. Those skilled in the art (e.g., a physician or veterinarian) can reduce or increase the dosage according to these or other conditions or requirements.
In certain embodiments, the modified oncolytic viruses and pharmaceutical compositions may be administered at a dose of about 104PFU to about 1014PFU (e.g., about 104PFU, about 2 x 104PFU, about 5 x 104PFU, about 105PFU, about 2 x 105PFU, about 5 x 105PFU, about 106PFU, about 2 x 106PFU, about 5 x 106PFU, about 107PFU, about 2 x 107PFU, about 5 x 107PFU, about 108PFU, about 2 x 108PFU, about 5 x 108PFU, about 109PFU, about 2 x 109PFU, about 5 x 109PFU, about 1010PFU, about 2 x 1o10 u, about 5 x 1010PFU, about 1011PFU, about 2 x 1011 u, about 1012PFU, about 2 x 1012PFU, about 5 x 1012PFU, about 1013PFU, about 2 x 101u, about 1014PFU, or about 3 x PFU). In certain of these embodiments, the modified oncolytic viruses and pharmaceutical compositions are administered at a dose of about 1011PFU or less. In the above embodiments, the dosage is 5 x 1010PFU or less, 2 x 1010PFU or less, 5 x 109PFU or less, 4 x 109PFU or less, 3 x 109PFU or less, 2 x 109PFU or less, or 109PFU or less. The particular dose may be divided and administered multiple times at intervals, such as once a day, two or more times a day, twice a month or more, once a week, once every two weeks, once every three weeks, once a month, or once every two months or more. In certain embodiments, the dosage administered may vary over the course of treatment. For example, in certain embodiments, the initial administered dose may be higher than the subsequently administered dose. In certain embodiments, the dosage administered is adjusted during the course of treatment according to the response of the subject to whom it is administered.
The term "PFU" as used in this patent refers to plaque forming units, which is a measure of the number of particles capable of forming plaques.
The dosing regimen may be adjusted to provide the best desired response (e.g., therapeutic response). For example, a single dose may be administered, or several divided doses may be administered over time.
Combination of two or more kinds of materials
In certain embodiments, the pharmaceutical composition may be used in combination with one or more other drugs. In certain embodiments, the composition comprises at least one additional drug.
In certain embodiments, the other drug is an antineoplastic agent. Any agent known to have antitumor activity can be used as the antitumor agent. In certain embodiments, the anti-neoplastic agent is selected from a chemical agent, a polynucleotide, a peptide, a protein, or any combination thereof.
In certain embodiments, the antineoplastic agent is a chemical agent. Illustrative examples of anti-tumor chemicals include, but are not limited to, mitomycin C, daunorubicin, doxorubicin, etoposide, tamoxifen, paclitaxel, vincristine, and rapamycin.
In certain embodiments, the anti-neoplastic agent is a polynucleotide. Illustrative examples of anti-tumor polynucleotides include, but are not limited to, antisense oligonucleotides such as bcl-2 antisense oligonucleotides, clusterin antisense oligonucleotides, and c-myc antisense oligonucleotides; and RNAs capable of RNA interference, including small interfering RNAs (sirnas), short hairpin RNAs (shrnas), and micro-interfering RNAs (mirnas), such as anti-VEGF sirnas, shrnas or mirnas, anti-bcl-2 sirnas, shrnas or mirnas, and anti-claudin-3 sirnas, shrnas or mirnas.
In certain embodiments, the antineoplastic agent is a peptide or protein. Illustrative examples of anti-tumor peptides or proteins include, but are not limited to, antibodies such as trastuzumab, rituximab, alemtuzumab, darizumab, nituzumab, gemtuzumab, ibritumomab and ibritumomab, protein therapeutics such as endostatin, angiostatin K1-3, leuprorelin, sex hormone binding globulin, and Bikunin.
Medical application
In another aspect, the present disclosure provides the use of a modified oncolytic virus of the present disclosure or a pharmaceutical composition of the present disclosure in the manufacture of a medicament for treating a tumor.
In another aspect, the present disclosure provides a modified oncolytic virus of the present disclosure or a pharmaceutical composition of the present disclosure for use in treating a tumor.
Examples
The following examples are shown to aid in the understanding of the present disclosure and should not be construed to limit in any way the scope of the invention as defined in the claims following the examples.
Example 1: virus construction
The initial Western Reserve (WR) strain of vaccinia virus was obtained from ATCC (www.atcc.org: VR-1354). Due to the involvement of multiple genes, the viral WR-GO-001 (see FIG. 1) with the genomic sequence SEQ ID NO:17 has been established by stepwise engineering methods. Briefly, in a first step, WR DNA was recombined with the modified pJS1175 vector to insert a selection gene into the Thymidine Kinase (TK) locus. Then, recombinant plasmids having HR and J3R flanking sequences and encoding recombinant human PD-1Fc chimeric proteins and recombinant human TGF- βrii Fc chimeric proteins were transfected into WR-infected CV-1 cells to completely delete TK and insert chimeric protein sequences (see table 4 below).
TABLE 4 WR-GO-001 plaque production and purification methods
The amino acid sequences of the recombinant human PD-1Fc chimeric protein and the recombinant human TGF-beta RII Fc chimeric protein are shown in SEQ ID NO. 5 and 6 respectively. The nucleic acid sequence encoding the recombinant human PD-1Fc chimeric protein is shown as SEQ ID NO. 13 and the recombinant human TGF- βRII Fc chimeric protein is shown as SEQ ID NO. 14 (see Table 5).
WR-GO-002 (FIG. 2 shows the insertion of the gene encoding the recombinant human TGF- βRII Fc chimeric protein, wherein the genomic sequence SEQ ID NO:18 is shown in Table 5), WR-GO-003 (FIG. 3 shows the insertion of the gene encoding the recombinant human PD-1Fc chimeric protein, wherein the genomic sequence SEQ ID NO:19 is shown in Table 5) and WR-GO-004 (the insertion of the gene encoding the recombinant human TGF- βRII Fc chimeric protein and the anti-human PD-1 heavy and light chains, wherein the genomic sequence SEQ ID NO:22 is shown in Table 5). The amino acid sequences of the anti-human PD-1 light and heavy chains shown in SEQ ID Nos. 20 and 21, respectively, and the amino acid sequence of the recombinant human TGF- βRII Fc chimeric protein shown in SEQ ID No. 6 were prepared by the same protocol as WR-GO-001 (see Table 5).
TABLE 5 sequences in WR-GO-001, WR-GO-002, WR-GO-003, WR-GO-004
/>
/>
/>
/>
/>
/>
/>
Example 2: characterization of WR-GO-001, WR-GO-002, WR-GO-003 and WR-GO-004
During engineering of these new viruses, their genomic integrity and protein expression are closely monitored.
PCR and sequencing
The identity of the viruses (WR-GO-001, WR-GO-002, WR-GO-003 and WR-GO-004) was confirmed by qPCR (TaqMan). TK deletion was also verified by sanger sequencing. Mulberry sequencing of the WR-GO-001 viral genome was performed with an alignment of DNA sequences designed for expression of recombinant human PD-1Fc chimeric protein (rPD-1 Fc) and recombinant human TGF- βRII Fc chimeric protein (rTGF- βRII Fc) in WR-GO-001. Alignment shows that the viral genome of WR-GO-001 is identical to the designed DNA sequence.
Confirmation of expression of human PD-1Fc chimeric proteins, human TGF-beta RII Fc chimeric proteins and anti-human PD-1 antibodies using ELISA
Recombinant human PD-1Fc chimeric proteins (rPD-1 Fc) expressed by WR-GO-001 and WR-GO-003 and WR-WT infected supernatants and intracellular samples were tested by ELISA (R & D Systems). Recombinant human TGF-beta RII Fc chimeric proteins expressed by WR-GO-001, WR-GO-002 and WR-GO-004 (rTGF-beta RII Fc) and WR-WT infected supernatants and intracellular samples were tested by ELISA (R & D Systems). The WR-GO-004 infected supernatants and intracellular samples were tested for anti-human PD-1 antibodies by ELISA (R & D Systems). The capture antibodies were diluted in PBS without carrier protein. The 96-well microwell plates were immediately coated with 100 μl per well of diluted capture antibody. Seal plate and incubate overnight at room temperature. Each well was aspirated and washed with wash buffer, and the process was repeated twice for a total of three washes. Wash by filling each well with wash buffer (400 μl) using a spray bottle, manifold dispenser, or automatic washer. Complete removal of liquid in each step is critical for good performance. After the last wash, any residual wash buffer was removed by aspiration or by inverting the plate and blotting with a clean paper towel. Each well of the microplate is closed. Incubate at room temperature for at least 1 hour. To each well 100 μl of reagent diluent or appropriate diluent for the sample or standard is added, the plate is covered with adhesive tape and incubated for 2 hours at room temperature. The aspiration/washing steps were repeated as previously described. To each well, 100 μl of detection antibody was added, the plate was covered with fresh adhesive tape, and incubated for 2 hours at room temperature. The aspiration/washing steps were repeated as previously described. To each well was added 100. Mu.L of working dilution of streptavidin-HRP, covered with plate, and incubated for 20 min at room temperature. Avoiding placing the plate under direct light. The aspiration/washing steps were repeated as previously described. To each well 100 μl of substrate solution was added and incubated for 20 minutes at room temperature. Avoiding placing the plate under direct light. To each well 50. Mu.L of stop solution was added. The plate was gently tapped to ensure adequate mixing. The optical density of each well was measured immediately using a microplate reader set at 450 nm.
Expression of human PD-1Fc chimeric proteins, human TGF-beta receptor II Fc chimeric proteins, and anti-human PD-1 antibodies in both supernatant and intracellular samples of Hela cells infected with WR-GO-002, WR-GO-003, or WR-GO-004 was measured using ELISA methods.
The results of the expression levels are shown in fig. 5A, 5B and 5C. FIG. 5A shows that WR-GO-001 stably expresses the PD-l-Fc protein, which is detected mainly in WR-GO-003 supernatant. FIG. 5B shows that WR-GO-004 stably expressed anti-PD-1 antibodies were detected primarily in the WR-GO-004 supernatant. FIG. 5C shows that all WR-GO-001, WR-GO-002 and WR-GO-004 stably expressed the TGFBRII-Fc protein, which was detected mainly in the supernatant.
Example 3: in vitro and in vivo studies of WR-GO-001, WR-GO-002, WR-GO-003 and WR-GO-004 recombinant viruses
The following in vitro and in vivo studies were performed to test the selectivity and anti-tumor effects of the novel viral constructs.
Stage 1. Determination of in vitro killing effect on cancer cells and transgene expression.
1. A sufficient amount of each of the four recombinant viruses was prepared and purified with sucrose buffer for in vitro assays and in vivo experiments.
2. Viral identity was confirmed by transgene using qPCR (TaqMan) and ELISA (R & D Systems).
3. Mycoplasma testing was performed on genetically engineered vaccinia viruses and cell lines used above to confirm sterility.
4. Cell killing effects of Hepal-6, 4T1, faDu and a549 at four different time points (24 hours, 48 hours, 72 hours and 96 hours) and three different infectious titers (MOI) were measured using MTS (3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxyphenyl) -2- (4-sulfophenyl) -2H-tetrazol-e). Hepal-6 (ATCC, CRL-1830) and 4T1 (ATCC, CRL-2539) are mouse tumor cell lines, faDu (ATCC, HTB-43) and A549 (ATCC, CCL-185) are human tumor cell lines.
5. ELISA (R & D Systems) measurements of in vitro expression of recombinant PD-1Fc (rPD-1 Fc) and recombinant TGF- βRII Fc (rTGF- βRII Fc) in FaDu human tumor cell lines.
6. Four recombinant viruses were formulated at 108PFU/ml and 107PFU/ml, respectively, in buffers compatible with in vivo administration.
7. Viral replication assays in the 4 different tumor cell lines described above were tested by q-RT-PCR (Taqman).
Stage 2. Testing of viral vector safety and biodistribution.
BALB/C mice and BALB/C nude mice were used at 1.5 weeks of age. BALB/C mice were distributed among 5 treatment groups (i.e., PBS control group, WR-WT control group, low, medium and high dose WR-GO-001 group). BALB/C nude mice were treated in the same manner as BALB/C mice.
2. Oncolytic virus therapy is initiated when the tumor size of the tumor group reaches 100mm3-300mm 3. The recombinant virus is administered in a single intratumoral injection.
3. Body weight, tumor volume and health status of recombinant virus treated mice were monitored.
4. On day 9 post virus injection, mice were sacrificed and tissues from heart, liver, spleen, stomach, kidneys, lungs and tumors were collected. Vaccinia genome copies in different tissues were quantified by qPCR.
5. And re-exciting. After tumor regression, tumors were implanted into the other side of the mice. Tumor volume and health status of treated mice were monitored.
Stage 3 anti-tumor Activity in tumor model
1.5 week old BALB/C mice were used for implantation of the 4T1 tumor cell line (day 1). Mice were further distributed among 6 treatment groups (i.e., PBS control group, WR-WT control group, WR-GO-001 group, WR-GO-002 group, WR-GO-003 group, and WR-GO-004 group).
Treatment is started when the tumor size reaches 100mm3-300mm 3.
2. Viruses (1X 107 pfu) were injected into each tumor twice on day 7 and day 10 from the day of tumor implantation, respectively.
3. Mice were monitored for body weight and tumor size.
4. On day 28, mice were sacrificed and PBMC cells were collected.
5. Antigen specific spleen cell responses were determined by ELISPOT and CTL.
Stage 4 measurement of tumor-specific cytotoxicity
Cytotoxicity of WR-GO 001, WR-GO 002, WR-GO-003 and WR-GO 004 in two classes of mouse tumor cell lines (CT 26 and MC 38), three classes of human tumor cell lines (FaDu, PA1 and PANC-1) and one class of human normal cell lines (HUVEC) was measured using the CCK-8 method.
With WR-GO-001, WR-GO-002, WR-GO-003 and WR-GO-004 respectively at 10.0,
MOI of 1.0 and 0.1 infects specific cells. Cytotoxicity was tested 24 hours, 48 hours and 72 hours after virus infection.
2.1 cell culture:
200. Mu.L of medium containing 2.5X104 specific cells was inoculated in 96-well plates and incubated overnight. Cell count prior to viral infection.
2.2 viral infection:
cells in 96-well plates were infected with WR-GO-001, WR-GO-002, WR-GO-003 and WR-GO-004 at three different MOI (0.1, 1.0 and 10.0 PFU/cell), respectively.
2.3 cell viability experiments:
at the indicated time points (24, 48 and 72 hours after virus infection), 10 μl CCK-8 was added to the test wells and incubated in the incubator for 2 hours. Then, cell viability was measured by uv spectrophotometer at 450nm (n=3).
FIGS. 6A-6H show the apparent cytotoxicity of these recombinant viruses against MC38 and CT26 cells in vitro 72 hours after viral infection at MOI of 10.0 and 1.0.
FIGS. 7A to 7D show cytotoxicity assays of WR-GO-003 in human tumor cell lines (FaDu, PA1, PANC-1) and human normal cell lines (HUVEC). Shows that WR-GO-003 has obvious cytotoxicity to human tumor cells in vitro, and that the cytotoxicity of WR-GO-003 to FaDu and PANC-1 is more obvious than that to PA 1. Meanwhile, cytotoxicity to HUVEC was not obvious, which suggests that WR-GO-003 has tumor-specific cytotoxicity.
FIGS. 8A to 8D show cytotoxicity assays of WR-GO-004 in human tumor cell lines (FaDu, PA1, PANC-1) and human normal cell lines (HUVEC). The apparent cytotoxicity of WR-GO-004 against human tumor cells in vitro was shown, and the cytotoxicity of WR-GO-004 against FaDu, PANC-1 was more pronounced than that of PA1 24 hours after viral infection. Meanwhile, cytotoxicity to HUVEC was not obvious, which suggests that WR-GO-004 has tumor-specific cytotoxicity.
Viral replication (viral particles; VP) curves in FaDu, measured by q-RT-PCR.
5.0X105 tumor cells inoculated overnight in 6-well plates were infected with WR-GO-003 or WR-GO-004 at a MOI of 0.1 in serum-free medium. Cell lysates and supernatants were harvested at the indicated time points (24 hours, 48 hours, 72 hours after virus infection). Genomic DNA was extracted using tissue and blood DNA extraction kits. VP was finally evaluated by q-RT-PCR system.
The results are shown in FIGS. 9A and 9B, and demonstrate that WR-GO-003 and WR-GO-004 replicate efficiently in FaDu cells.
The above description is only a preferred embodiment of the present invention, and the protection scope of the present invention is not limited to the above examples, and all technical solutions belonging to the concept of the present invention belong to the protection scope of the present invention. It should be noted that modifications and adaptations to the present invention may occur to one skilled in the art without departing from the principles of the present invention and are intended to be within the scope of the present invention.
Sequence listing
<110> Shanghai brocade biotechnology Co., ltd
Shanghai Jinzhiyuan biotechnology Co., ltd
<120> modified oncolytic viruses, compositions and uses thereof
<130> 068615-8002WO01
<160> 40
<170> patent version 3.5
<210> 1
<211> 149
<212> PRT
<213> Homo sapiens (Homo sapiens)
<220>
<221> PD-1 ECD G22-170
<222> (1)..(149)
<400> 1
Gly Trp Phe Leu Asp Ser Pro Asp Arg Pro Trp Asn Pro Pro Thr Phe
1 5 10 15
Ser Pro Ala Leu Leu Val Val Thr Glu Gly Asp Asn Ala Thr Phe Thr
20 25 30
Cys Ser Phe Ser Asn Thr Ser Glu Ser Phe Val Leu Asn Trp Tyr Arg
35 40 45
Met Ser Pro Ser Asn Gln Thr Asp Lys Leu Ala Ala Phe Pro Glu Asp
50 55 60
Arg Ser Gln Pro Gly Gln Asp Cys Arg Phe Arg Val Thr Gln Leu Pro
65 70 75 80
Asn Gly Arg Asp Phe His Met Ser Val Val Arg Ala Arg Arg Asn Asp
85 90 95
Ser Gly Thr Tyr Leu Cys Gly Ala Ile Ser Leu Ala Pro Lys Ala Gln
100 105 110
Ile Lys Glu Ser Leu Arg Ala Glu Leu Arg Val Thr Glu Arg Arg Ala
115 120 125
Glu Val Pro Thr Ala His Pro Ser Pro Ser Pro Arg Pro Ala Gly Gln
130 135 140
Phe Gln Thr Leu Val
145
<210> 2
<211> 144
<212> PRT
<213> Chile person
<220>
<221> TGF-beta receptor II ECD23-166
<222> (1)..(144)
<400> 2
Thr Ile Pro Pro His Val Gln Lys Ser Val Asn Asn Asp Met Ile Val
1 5 10 15
Thr Asp Asn Asn Gly Ala Val Lys Phe Pro Gln Leu Cys Lys Phe Cys
20 25 30
Asp Val Arg Phe Ser Thr Cys Asp Asn Gln Lys Ser Cys Met Ser Asn
35 40 45
Cys Ser Ile Thr Ser Ile Cys Glu Lys Pro Gln Glu Val Cys Val Ala
50 55 60
Val Trp Arg Lys Asn Asp Glu Asn Ile Thr Leu Glu Thr Val Cys His
65 70 75 80
Asp Pro Lys Leu Pro Tyr His Asp Phe Ile Leu Glu Asp Ala Ala Ser
85 90 95
Pro Lys Cys Ile Met Lys Glu Lys Lys Lys Pro Gly Glu Thr Phe Phe
100 105 110
Met Cys Ser Cys Ser Ser Asp Glu Cys Asn Asp Asn Ile Ile Phe Ser
115 120 125
Glu Glu Tyr Asn Thr Ser Asn Pro Asp Leu Leu Leu Val Ile Phe Gln
130 135 140
<210> 3
<211> 232
<212> PRT
<213> Chile person
<220>
<221> human IgG1-Fc region
<222> (1)..(232)
<400> 3
Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala
1 5 10 15
Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro
20 25 30
Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val
35 40 45
Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val
50 55 60
Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln
65 70 75 80
Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln
85 90 95
Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala
100 105 110
Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro
115 120 125
Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr
130 135 140
Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser
145 150 155 160
Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr
165 170 175
Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr
180 185 190
Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe
195 200 205
Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys
210 215 220
Ser Leu Ser Leu Ser Pro Gly Lys
225 230
<210> 4
<211> 17
<212> PRT
<213> Chile person
<220>
<221> CD33 Signal peptide
<222> (1)..(17)
<400> 4
Met Pro Leu Leu Leu Leu Leu Pro Leu Leu Trp Ala Gly Ala Leu Ala
1 5 10 15
Met
<210> 5
<211> 398
<212> PRT
<213> artificial sequence
<220>
<223> CD33 sp-PD-1-Fc
<400> 5
Met Pro Leu Leu Leu Leu Leu Pro Leu Leu Trp Ala Gly Ala Leu Ala
1 5 10 15
Met Gly Trp Phe Leu Asp Ser Pro Asp Arg Pro Trp Asn Pro Pro Thr
20 25 30
Phe Ser Pro Ala Leu Leu Val Val Thr Glu Gly Asp Asn Ala Thr Phe
35 40 45
Thr Cys Ser Phe Ser Asn Thr Ser Glu Ser Phe Val Leu Asn Trp Tyr
50 55 60
Arg Met Ser Pro Ser Asn Gln Thr Asp Lys Leu Ala Ala Phe Pro Glu
65 70 75 80
Asp Arg Ser Gln Pro Gly Gln Asp Cys Arg Phe Arg Val Thr Gln Leu
85 90 95
Pro Asn Gly Arg Asp Phe His Met Ser Val Val Arg Ala Arg Arg Asn
100 105 110
Asp Ser Gly Thr Tyr Leu Cys Gly Ala Ile Ser Leu Ala Pro Lys Ala
115 120 125
Gln Ile Lys Glu Ser Leu Arg Ala Glu Leu Arg Val Thr Glu Arg Arg
130 135 140
Ala Glu Val Pro Thr Ala His Pro Ser Pro Ser Pro Arg Pro Ala Gly
145 150 155 160
Gln Phe Gln Thr Leu Val Glu Pro Lys Ser Cys Asp Lys Thr His Thr
165 170 175
Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe
180 185 190
Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro
195 200 205
Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val
210 215 220
Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr
225 230 235 240
Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val
245 250 255
Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys
260 265 270
Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser
275 280 285
Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro
290 295 300
Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val
305 310 315 320
Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly
325 330 335
Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp
340 345 350
Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp
355 360 365
Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His
370 375 380
Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys
385 390 395
<210> 6
<211> 393
<212> PRT
<213> artificial sequence
<220>
<223> CD33 sp-TGF-beta receptor II-Fc
<400> 6
Met Pro Leu Leu Leu Leu Leu Pro Leu Leu Trp Ala Gly Ala Leu Ala
1 5 10 15
Met Thr Ile Pro Pro His Val Gln Lys Ser Val Asn Asn Asp Met Ile
20 25 30
Val Thr Asp Asn Asn Gly Ala Val Lys Phe Pro Gln Leu Cys Lys Phe
35 40 45
Cys Asp Val Arg Phe Ser Thr Cys Asp Asn Gln Lys Ser Cys Met Ser
50 55 60
Asn Cys Ser Ile Thr Ser Ile Cys Glu Lys Pro Gln Glu Val Cys Val
65 70 75 80
Ala Val Trp Arg Lys Asn Asp Glu Asn Ile Thr Leu Glu Thr Val Cys
85 90 95
His Asp Pro Lys Leu Pro Tyr His Asp Phe Ile Leu Glu Asp Ala Ala
100 105 110
Ser Pro Lys Cys Ile Met Lys Glu Lys Lys Lys Pro Gly Glu Thr Phe
115 120 125
Phe Met Cys Ser Cys Ser Ser Asp Glu Cys Asn Asp Asn Ile Ile Phe
130 135 140
Ser Glu Glu Tyr Asn Thr Ser Asn Pro Asp Leu Leu Leu Val Ile Phe
145 150 155 160
Gln Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro
165 170 175
Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys
180 185 190
Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val
195 200 205
Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr
210 215 220
Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu
225 230 235 240
Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His
245 250 255
Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys
260 265 270
Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln
275 280 285
Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu
290 295 300
Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro
305 310 315 320
Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn
325 330 335
Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu
340 345 350
Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val
355 360 365
Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln
370 375 380
Lys Ser Leu Ser Leu Ser Pro Gly Lys
385 390
<210> 7
<211> 447
<212> DNA
<213> Chile person
<220>
<221> PD-1 ECD G22-170
<222> (1)..(447)
<400> 7
ggctggttcc tcgactctcc cgacagacct tggaacccac ctacattcag ccccgccctc 60
ctcgtggtga ccgagggcga caacgccacc ttcacatgca gcttcagcaa cacatccgag 120
tctttcgtgc tgaactggta tcggatgtct ccatctaacc agaccgacaa gctcgccgcc 180
ttccccgagg accgctcgca gcctggccag gattgcagat tcagagtgac ccagctccca 240
aacggcaggg acttccacat gagcgtggtg cgcgcccgga gaaacgactc tggcacatac 300
ctgtgcggcg ccattagcct cgcccctaag gcccagatta aggagtccct gcgggccgag 360
ctgagagtga cagagcggag agccgaggtg cctaccgccc acccatctcc atctcctaga 420
cccgccggcc agttccagac actcgtg 447
<210> 8
<211> 432
<212> DNA
<213> Chile person
<220>
<221> TGF-beta receptor II ECD23-166
<222> (1)..(432)
<400> 8
acaattcctc ctcacgtgca gaagagcgtg aataatgata tgattgtgac cgataataac 60
ggcgccgtga aatttcccca gctgtgtaag ttttgtgatg tgagattcag cacctgtgat 120
aatcagaagt cttgcatgtc taactgttcc atcacatcca tctgtgagaa gccacaggag 180
gtgtgcgtgg ccgtgtggag aaagaatgat gagaatatca cacttgagac cgtgtgccac 240
gacccaaaac tgccatacca cgattttatt ctggaggatg ccgcctctcc taagtgcatt 300
atgaaggaga agaagaagcc aggcgagacc ttctttatgt gtagctgttc ctctgatgag 360
tgtaatgata atattatctt cagcgaagag tataacacca gtaatcctga cctgctgctt 420
gtgattttcc ag 432
<210> 9
<211> 696
<212> DNA
<213> Chile person
<220>
<221> human IgG1-Fc region of PD-1
<222> (1)..(696)
<400> 9
gagcctaagt cttgcgacaa gacccacaca tgcccacctt gccccgcccc tgagctgctg 60
ggcggcccat ccgtgttcct gttcccacca aagccaaagg acacactcat gattagcagg 120
acacctgagg tgacatgcgt ggtggtggac gtgtctcacg aggaccccga ggtgaagttc 180
aactggtacg tggacggcgt ggaggtgcac aacgccaaga caaagcctag agaggagcag 240
tacaactcta cataccgcgt ggtgtccgtg ctcacagtgc tgcaccagga ctggctgaac 300
ggcaaggagt acaagtgcaa ggtgtccaac aaggccctcc ccgcccctat tgaaaaaacc 360
attagcaagg ccaagggcca gccacgcgag cctcaggtgt acacactgcc tccttctagg 420
gacgagctga ccaagaacca ggtgtccctc acatgcctcg tgaagggctt ctaccctagc 480
gacattgccg tggagtggga gtctaacggc cagcctgaga acaactacaa gacaacacca 540
cctgtgctcg actctgacgg ctctttcttc ctgtactcta agctcacagt ggacaagagt 600
agatggcagc agggcaacgt gttctcttgc tctgtgatgc acgaggccct gcacaaccac 660
tacacccaga agtctctgtc tctgtctcca ggcaag 696
<210> 10
<211> 696
<212> DNA
<213> Chile person
<220>
<221> human IgG1-Fc region of TGF-beta receptor II
<222> (1)..(696)
<400> 10
gagcctaagt cttgtgacaa gacacacaca tgccctccct gtccagcccc tgagctgctg 60
ggcggcccta gcgtgtttct gttcccccct aagcctaaag atacactgat gattagcaga 120
acaccagagg tgacctgtgt ggtggtggat gtgagccacg aggaccctga ggtgaagttt 180
aactggtatg tggatggagt ggaagtgcac aatgctaaga ccaagcctag agaggagcag 240
tataatagca catatagagt ggtgtctgtg ctgaccgtgc tgcaccagga ctggctgaat 300
ggcaaagagt ataaatgtaa ggtgagcaac aaggccctgc ccgcccccat tgagaaaacc 360
atctctaagg ctaagggaca gccaagagag cctcaggtgt atacactgcc tccttctaga 420
gatgagctga ccaaaaacca ggttagcctg acatgcctgg tgaaaggctt ctatccaagc 480
gatatcgctg tggaatggga gagcaatggc cagcctgaga ataattataa aaccacccca 540
ccagtgctgg atagcgatgg cagctttttc ctgtattcta agctgacagt ggataaatct 600
aggtggcagc agggcaatgt gtttagctgc tctgtgatgc acgaggccct gcacaatcac 660
tatacacaga agagcctgag cctgagccct ggcaag 696
<210> 11
<211> 51
<212> DNA
<213> Chile person
<220>
<221> CD33 Signal peptide of PD-1
<222> (1)..(51)
<400> 11
atgccactgc tcctcctgct gccactgctc tgggccggcg ccctcgctat g 51
<210> 12
<211> 51
<212> DNA
<213> Chile person
<220>
<221> CD33 Signal peptide of TGF-beta receptor II
<222> (1)..(51)
<400> 12
atgcctctgc tgctgctgct gcctctgctg tgggccggcg ccctggccat g 51
<210> 13
<211> 1197
<212> DNA
<213> artificial sequence
<220>
<223> CD33 sp-PD-1-Fc
<400> 13
atgccactgc tcctcctgct gccactgctc tgggccggcg ccctcgctat gggctggttc 60
ctcgactctc ccgacagacc ttggaaccca cctacattca gccccgccct cctcgtggtg 120
accgagggcg acaacgccac cttcacatgc agcttcagca acacatccga gtctttcgtg 180
ctgaactggt atcggatgtc tccatctaac cagaccgaca agctcgccgc cttccccgag 240
gaccgctcgc agcctggcca ggattgcaga ttcagagtga cccagctccc aaacggcagg 300
gacttccaca tgagcgtggt gcgcgcccgg agaaacgact ctggcacata cctgtgcggc 360
gccattagcc tcgcccctaa ggcccagatt aaggagtccc tgcgggccga gctgagagtg 420
acagagcgga gagccgaggt gcctaccgcc cacccatctc catctcctag acccgccggc 480
cagttccaga cactcgtgga gcctaagtct tgcgacaaga cccacacatg cccaccttgc 540
cccgcccctg agctgctggg cggcccatcc gtgttcctgt tcccaccaaa gccaaaggac 600
acactcatga ttagcaggac acctgaggtg acatgcgtgg tggtggacgt gtctcacgag 660
gaccccgagg tgaagttcaa ctggtacgtg gacggcgtgg aggtgcacaa cgccaagaca 720
aagcctagag aggagcagta caactctaca taccgcgtgg tgtccgtgct cacagtgctg 780
caccaggact ggctgaacgg caaggagtac aagtgcaagg tgtccaacaa ggccctcccc 840
gcccctattg aaaaaaccat tagcaaggcc aagggccagc cacgcgagcc tcaggtgtac 900
acactgcctc cttctaggga cgagctgacc aagaaccagg tgtccctcac atgcctcgtg 960
aagggcttct accctagcga cattgccgtg gagtgggagt ctaacggcca gcctgagaac 1020
aactacaaga caacaccacc tgtgctcgac tctgacggct ctttcttcct gtactctaag 1080
ctcacagtgg acaagagtag atggcagcag ggcaacgtgt tctcttgctc tgtgatgcac 1140
gaggccctgc acaaccacta cacccagaag tctctgtctc tgtctccagg caagtga 1197
<210> 14
<211> 1182
<212> DNA
<213> artificial sequence
<220>
<223> CD33 sp-TGF-beta receptor II-Fc
<400> 14
atgcctctgc tgctgctgct gcctctgctg tgggccggcg ccctggccat gacaattcct 60
cctcacgtgc agaagagcgt gaataatgat atgattgtga ccgataataa cggcgccgtg 120
aaatttcccc agctgtgtaa gttttgtgat gtgagattca gcacctgtga taatcagaag 180
tcttgcatgt ctaactgttc catcacatcc atctgtgaga agccacagga ggtgtgcgtg 240
gccgtgtgga gaaagaatga tgagaatatc acacttgaga ccgtgtgcca cgacccaaaa 300
ctgccatacc acgattttat tctggaggat gccgcctctc ctaagtgcat tatgaaggag 360
aagaagaagc caggcgagac cttctttatg tgtagctgtt cctctgatga gtgtaatgat 420
aatattatct tcagcgaaga gtataacacc agtaatcctg acctgctgct tgtgattttc 480
caggagccta agtcttgtga caagacacac acatgccctc cctgtccagc ccctgagctg 540
ctgggcggcc ctagcgtgtt tctgttcccc cctaagccta aagatacact gatgattagc 600
agaacaccag aggtgacctg tgtggtggtg gatgtgagcc acgaggaccc tgaggtgaag 660
tttaactggt atgtggatgg agtggaagtg cacaatgcta agaccaagcc tagagaggag 720
cagtataata gcacatatag agtggtgtct gtgctgaccg tgctgcacca ggactggctg 780
aatggcaaag agtataaatg taaggtgagc aacaaggccc tgcccgcccc cattgagaaa 840
accatctcta aggctaaggg acagccaaga gagcctcagg tgtatacact gcctccttct 900
agagatgagc tgaccaaaaa ccaggttagc ctgacatgcc tggtgaaagg cttctatcca 960
agcgatatcg ctgtggaatg ggagagcaat ggccagcctg agaataatta taaaaccacc 1020
ccaccagtgc tggatagcga tggcagcttt ttcctgtatt ctaagctgac agtggataaa 1080
tctaggtggc agcagggcaa tgtgtttagc tgctctgtga tgcacgaggc cctgcacaat 1140
cactatacac agaagagcct gagcctgagc cctggcaagt ga 1182
<210> 15
<211> 42
<212> DNA
<213> vaccinia Virus
<220>
<221> pSE/L promoter
<222> (1)..(42)
<400> 15
aaaaattgaa attttatttt ttttttttgg aatataaata ag 42
<210> 16
<211> 10
<212> DNA
<213> vaccinia Virus
<220>
<221> T5NT stop codon
<222> (1)..(7)
<220>
<221> misc_feature
<222> (6)..(6)
<223> n may be a, t, g or c.
<220>
<221> misc_feature
<222> (8)..(10)
<223> n does not exist.
<400> 16
tttttntnnn 10
<210> 17
<211> 4392
<212> DNA
<213> artificial sequence
<220>
<223> WR-GO-001
<400> 17
atggagaatg ttcctaatgt atactttaat cctgtgttta tagagcccac gtttaaacat 60
tctttattaa gtgtttataa acacagatta atagttttat ttgaagtatt cgttgtattc 120
attctaatat atgtattttt tagatctgaa ttaaatatgt tcttcatgcc taaacgaaaa 180
atacccgatc ctattgatag attacgacgt gctaatctag cgtgtgaaga cgataaatta 240
atgatctatg gattaccatg gatgacaact caaacatctg cgttatcaat aaatagtaaa 300
ccgatagtgt ataaagattg tgcaaagctt ttgcgatcaa taaatggatc acaaccagta 360
tctcttaacg atgttcttcg cagatgatga ttcatttttt aagtatttgg ctagtcaaga 420
tgatgaatct tcattatctg atatattgca aatcactcaa tatctagact ttctgttatt 480
attattgatc caatcaaaaa ataaattaga agccgtgggt cattgttatg aatctctttc 540
agaggaatac agacaattga caaaattcac agactttcaa gattttaaaa aactgtttaa 600
caaggtccct attgttacag atggaagggt caaacttaat aaaggatatt tgttcgactt 660
tgtgattagt ttgatgcgat tcaaaaaaga atcctctcta gctaccaccg caatagatcc 720
tgttagatac atagatcctc gtcgcaatat cgcattttct aacgtgatgg atatattaaa 780
gtcgaataaa gtgaacaata attaattctt tattgtcaag gcctagaaaa acctcacttg 840
cctggagaca gagacagaga cttctgggtg tagtggttgt gcagggcctc gtgcatcaca 900
gagcaagaga acacgttgcc ctgctgccat ctactcttgt ccactgtgag cttagagtac 960
aggaagaaag agccgtcaga gtcgagcaca ggtggtgttg tcttgtagtt gttctcaggc 1020
tggccgttag actcccactc cacggcaatg tcgctagggt agaagccctt cacgaggcat 1080
gtgagggaca cctggttctt ggtcagctcg tccctagaag gaggcagtgt gtacacctga 1140
ggctcgcgtg gctggccctt ggccttgcta atggtttttt caataggggc ggggagggcc 1200
ttgttggaca ccttgcactt gtactccttg ccgttcagcc agtcctggtg cagcactgtg 1260
agcacggaca ccacgcggta tgtagagttg tactgctcct ctctaggctt tgtcttggcg 1320
ttgtgcacct ccacgccgtc cacgtaccag ttgaacttca cctcggggtc ctcgtgagac 1380
acgtccacca ccacgcatgt cacctcaggt gtcctgctaa tcatgagtgt gtcctttggc 1440
tttggtggga acaggaacac ggatgggccg cccagcagct caggggcggg gcaaggtggg 1500
catgtgtggg tcttgtcgca agacttaggc tccacgagtg tctggaactg gccggcgggt 1560
ctaggagatg gagatgggtg ggcggtaggc acctcggctc tccgctctgt cactctcagc 1620
tcggcccgca gggactcctt aatctgggcc ttaggggcga ggctaatggc gccgcacagg 1680
tatgtgccag agtcgtttct ccgggcgcgc accacgctca tgtggaagtc cctgccgttt 1740
gggagctggg tcactctgaa tctgcaatcc tggccaggct gcgagcggtc ctcggggaag 1800
gcggcgagct tgtcggtctg gttagatgga gacatccgat accagttcag cacgaaagac 1860
tcggatgtgt tgctgaagct gcatgtgaag gtggcgttgt cgccctcggt caccacgagg 1920
agggcggggc tgaatgtagg tgggttccaa ggtctgtcgg gagagtcgag gaaccagccc 1980
atagcgaggg cgccggccca gagcagtggc agcaggagga gcagtggcat ggcttattta 2040
tattccaaaa aaaaaaaata aaatttcaat ttttactagt aaaaattgaa attttatttt 2100
ttttttttgg aatataaata aggatgcctc tgctgctgct gctgcctctg ctgtgggccg 2160
gcgccctggc catgacaatt cctcctcacg tgcagaagag cgtgaataat gatatgattg 2220
tgaccgataa taacggcgcc gtgaaatttc cccagctgtg taagttttgt gatgtgagat 2280
tcagcacctg tgataatcag aagtcttgca tgtctaactg ttccatcaca tccatctgtg 2340
agaagccaca ggaggtgtgc gtggccgtgt ggagaaagaa tgatgagaat atcacacttg 2400
agaccgtgtg ccacgaccca aaactgccat accacgattt tattctggag gatgccgcct 2460
ctcctaagtg cattatgaag gagaagaaga agccaggcga gaccttcttt atgtgtagct 2520
gttcctctga tgagtgtaat gataatatta tcttcagcga agagtataac accagtaatc 2580
ctgacctgct gcttgtgatt ttccaggagc ctaagtcttg tgacaagaca cacacatgcc 2640
ctccctgtcc agcccctgag ctgctgggcg gccctagcgt gtttctgttc ccccctaagc 2700
ctaaagatac actgatgatt agcagaacac cagaggtgac ctgtgtggtg gtggatgtga 2760
gccacgagga ccctgaggtg aagtttaact ggtatgtgga tggagtggaa gtgcacaatg 2820
ctaagaccaa gcctagagag gagcagtata atagcacata tagagtggtg tctgtgctga 2880
ccgtgctgca ccaggactgg ctgaatggca aagagtataa atgtaaggtg agcaacaagg 2940
ccctgcccgc ccccattgag aaaaccatct ctaaggctaa gggacagcca agagagcctc 3000
aggtgtatac actgcctcct tctagagatg agctgaccaa aaaccaggtt agcctgacat 3060
gcctggtgaa aggcttctat ccaagcgata tcgctgtgga atgggagagc aatggccagc 3120
ctgagaataa ttataaaacc accccaccag tgctggatag cgatggcagc tttttcctgt 3180
attctaagct gacagtggat aaatctaggt ggcagcaggg caatgtgttt agctgctctg 3240
tgatgcacga ggccctgcac aatcactata cacagaagag cctgagcctg agccctggca 3300
agtgactttt tatgagctcg aattctatta tattttttat ctaaaaaact aaaaataaac 3360
attgattaaa ttttaatata atacttaaaa atggatgttg tgtcgttaga taaaccgttt 3420
atgtattttg aggaaattga taatgagtta gattacgaac cagaaagtgc aaatgaggtc 3480
gcaaaaaaac tgccgtatca aggacagtta aaactattac taggagaatt attttttctt 3540
agtaagttac agcgacacgg tatattagat ggtgccaccg tagtgtatat aggatctgct 3600
cccggtacac atatacgtta tttgagagat catttctata atttaggagt gatcatcaaa 3660
tggatgctaa ttgacggccg ccatcatgat cctattttaa atggattgcg tgatgtgact 3720
ctagtgactc ggttcgttga tgaggaatat ctacgatcca tcaaaaaaca actgcatcct 3780
tctaagatta ttttaatttc tgatgtgaga tccaaacgag gaggaaatga acctagtacg 3840
gcggatttac taagtaatta cgctctacaa aatgtcatga ttagtatttt aaaccccgtg 3900
gcgtctagtc ttaaatggag atgcccgttt ccagatcaat ggatcaagga cttttatatc 3960
ccacacggta ataaaatgtt acaacctttt gctccttcat attcagctga aatgagatta 4020
ttaagtattt ataccggtga gaacatgaga ctgactcgag ttaccaaatc agacgctgta 4080
aattatgaaa aaaagatgta ctaccttaat aagatcgtcc gtaacaaagt agttgttaac 4140
tttgattatc ctaatcagga atatgactat tttcacatgt actttatgct gaggaccgtg 4200
tactgcaata aaacatttcc tactactaaa gcaaaggtac tatttctaca acaatctata 4260
tttcgtttct taaatattcc aacaacatca actgaaaaag ttagtcatga accaatacaa 4320
cgtaaaatat ctagcaaaaa ttctatgtct aaaaacagaa atagcaagag atccgtacgc 4380
agtaataaat ag 4392
<210> 18
<211> 3199
<212> DNA
<213> artificial sequence
<220>
<223> WR-GO-002
<400> 18
atggagaatg ttcctaatgt atactttaat cctgtgttta tagagcccac gtttaaacat 60
tctttattaa gtgtttataa acacagatta atagttttat ttgaagtatt cgttgtattc 120
attctaatat atgtattttt tagatctgaa ttaaatatgt tcttcatgcc taaacgaaaa 180
atacccgatc ctattgatag attacgacgt gctaatctag cgtgtgaaga cgataaatta 240
atgatctatg gattaccatg gatgacaact caaacatctg cgttatcaat aaatagtaaa 300
ccgatagtgt ataaagattg tgcaaagctt ttgcgatcaa taaatggatc acaaccagta 360
tctcttaacg atgttcttcg cagatgatga ttcatttttt aagtatttgg ctagtcaaga 420
tgatgaatct tcattatctg atatattgca aatcactcaa tatctagact ttctgttatt 480
attattgatc caatcaaaaa ataaattaga agccgtgggt cattgttatg aatctctttc 540
agaggaatac agacaattga caaaattcac agactttcaa gattttaaaa aactgtttaa 600
caaggtccct attgttacag atggaagggt caaacttaat aaaggatatt tgttcgactt 660
tgtgattagt ttgatgcgat tcaaaaaaga atcctctcta gctaccaccg caatagatcc 720
tgttagatac atagatcctc gtcgcaatat cgcattttct aacgtgatgg atatattaaa 780
gtcgaataaa gtgaacaata attaattctt tattgtcaag gcctagaaaa acctcgaggc 840
ttatttatat tccaaaaaaa aaaaataaaa tttcaatttt tactagtaaa aattgaaatt 900
ttattttttt tttttggaat ataaataagg atgcctctgc tgctgctgct gcctctgctg 960
tgggccggcg ccctggccat gacaattcct cctcacgtgc agaagagcgt gaataatgat 1020
atgattgtga ccgataataa cggcgccgtg aaatttcccc agctgtgtaa gttttgtgat 1080
gtgagattca gcacctgtga taatcagaag tcttgcatgt ctaactgttc catcacatcc 1140
atctgtgaga agccacagga ggtgtgcgtg gccgtgtgga gaaagaatga tgagaatatc 1200
acacttgaga ccgtgtgcca cgacccaaaa ctgccatacc acgattttat tctggaggat 1260
gccgcctctc ctaagtgcat tatgaaggag aagaagaagc caggcgagac cttctttatg 1320
tgtagctgtt cctctgatga gtgtaatgat aatattatct tcagcgaaga gtataacacc 1380
agtaatcctg acctgctgct tgtgattttc caggagccta agtcttgtga caagacacac 1440
acatgccctc cctgtccagc ccctgagctg ctgggcggcc ctagcgtgtt tctgttcccc 1500
cctaagccta aagatacact gatgattagc agaacaccag aggtgacctg tgtggtggtg 1560
gatgtgagcc acgaggaccc tgaggtgaag tttaactggt atgtggatgg agtggaagtg 1620
cacaatgcta agaccaagcc tagagaggag cagtataata gcacatatag agtggtgtct 1680
gtgctgaccg tgctgcacca ggactggctg aatggcaaag agtataaatg taaggtgagc 1740
aacaaggccc tgcccgcccc cattgagaaa accatctcta aggctaaggg acagccaaga 1800
gagcctcagg tgtatacact gcctccttct agagatgagc tgaccaaaaa ccaggttagc 1860
ctgacatgcc tggtgaaagg cttctatcca agcgatatcg ctgtggaatg ggagagcaat 1920
ggccagcctg agaataatta taaaaccacc ccaccagtgc tggatagcga tggcagcttt 1980
ttcctgtatt ctaagctgac agtggataaa tctaggtggc agcagggcaa tgtgtttagc 2040
tgctctgtga tgcacgaggc cctgcacaat cactatacac agaagagcct gagcctgagc 2100
cctggcaagt gactttttat gagctcgaat tctattatat tttttatcta aaaaactaaa 2160
aataaacatt gattaaattt taatataata cttaaaaatg gatgttgtgt cgttagataa 2220
accgtttatg tattttgagg aaattgataa tgagttagat tacgaaccag aaagtgcaaa 2280
tgaggtcgca aaaaaactgc cgtatcaagg acagttaaaa ctattactag gagaattatt 2340
ttttcttagt aagttacagc gacacggtat attagatggt gccaccgtag tgtatatagg 2400
atctgctccc ggtacacata tacgttattt gagagatcat ttctataatt taggagtgat 2460
catcaaatgg atgctaattg acggccgcca tcatgatcct attttaaatg gattgcgtga 2520
tgtgactcta gtgactcggt tcgttgatga ggaatatcta cgatccatca aaaaacaact 2580
gcatccttct aagattattt taatttctga tgtgagatcc aaacgaggag gaaatgaacc 2640
tagtacggcg gatttactaa gtaattacgc tctacaaaat gtcatgatta gtattttaaa 2700
ccccgtggcg tctagtctta aatggagatg cccgtttcca gatcaatgga tcaaggactt 2760
ttatatccca cacggtaata aaatgttaca accttttgct ccttcatatt cagctgaaat 2820
gagattatta agtatttata ccggtgagaa catgagactg actcgagtta ccaaatcaga 2880
cgctgtaaat tatgaaaaaa agatgtacta ccttaataag atcgtccgta acaaagtagt 2940
tgttaacttt gattatccta atcaggaata tgactatttt cacatgtact ttatgctgag 3000
gaccgtgtac tgcaataaaa catttcctac tactaaagca aaggtactat ttctacaaca 3060
atctatattt cgtttcttaa atattccaac aacatcaact gaaaaagtta gtcatgaacc 3120
aatacaacgt aaaatatcta gcaaaaattc tatgtctaaa aacagaaata gcaagagatc 3180
cgtacgcagt aataaatag 3199
<210> 19
<211> 3214
<212> DNA
<213> artificial sequence
<220>
<223> WR-GO-003
<400> 19
atggagaatg ttcctaatgt atactttaat cctgtgttta tagagcccac gtttaaacat 60
tctttattaa gtgtttataa acacagatta atagttttat ttgaagtatt cgttgtattc 120
attctaatat atgtattttt tagatctgaa ttaaatatgt tcttcatgcc taaacgaaaa 180
atacccgatc ctattgatag attacgacgt gctaatctag cgtgtgaaga cgataaatta 240
atgatctatg gattaccatg gatgacaact caaacatctg cgttatcaat aaatagtaaa 300
ccgatagtgt ataaagattg tgcaaagctt ttgcgatcaa taaatggatc acaaccagta 360
tctcttaacg atgttcttcg cagatgatga ttcatttttt aagtatttgg ctagtcaaga 420
tgatgaatct tcattatctg atatattgca aatcactcaa tatctagact ttctgttatt 480
attattgatc caatcaaaaa ataaattaga agccgtgggt cattgttatg aatctctttc 540
agaggaatac agacaattga caaaattcac agactttcaa gattttaaaa aactgtttaa 600
caaggtccct attgttacag atggaagggt caaacttaat aaaggatatt tgttcgactt 660
tgtgattagt ttgatgcgat tcaaaaaaga atcctctcta gctaccaccg caatagatcc 720
tgttagatac atagatcctc gtcgcaatat cgcattttct aacgtgatgg atatattaaa 780
gtcgaataaa gtgaacaata attaattctt tattgtcaag gcctagaaaa acctcacttg 840
cctggagaca gagacagaga cttctgggtg tagtggttgt gcagggcctc gtgcatcaca 900
gagcaagaga acacgttgcc ctgctgccat ctactcttgt ccactgtgag cttagagtac 960
aggaagaaag agccgtcaga gtcgagcaca ggtggtgttg tcttgtagtt gttctcaggc 1020
tggccgttag actcccactc cacggcaatg tcgctagggt agaagccctt cacgaggcat 1080
gtgagggaca cctggttctt ggtcagctcg tccctagaag gaggcagtgt gtacacctga 1140
ggctcgcgtg gctggccctt ggccttgcta atggtttttt caataggggc ggggagggcc 1200
ttgttggaca ccttgcactt gtactccttg ccgttcagcc agtcctggtg cagcactgtg 1260
agcacggaca ccacgcggta tgtagagttg tactgctcct ctctaggctt tgtcttggcg 1320
ttgtgcacct ccacgccgtc cacgtaccag ttgaacttca cctcggggtc ctcgtgagac 1380
acgtccacca ccacgcatgt cacctcaggt gtcctgctaa tcatgagtgt gtcctttggc 1440
tttggtggga acaggaacac ggatgggccg cccagcagct caggggcggg gcaaggtggg 1500
catgtgtggg tcttgtcgca agacttaggc tccacgagtg tctggaactg gccggcgggt 1560
ctaggagatg gagatgggtg ggcggtaggc acctcggctc tccgctctgt cactctcagc 1620
tcggcccgca gggactcctt aatctgggcc ttaggggcga ggctaatggc gccgcacagg 1680
tatgtgccag agtcgtttct ccgggcgcgc accacgctca tgtggaagtc cctgccgttt 1740
gggagctggg tcactctgaa tctgcaatcc tggccaggct gcgagcggtc ctcggggaag 1800
gcggcgagct tgtcggtctg gttagatgga gacatccgat accagttcag cacgaaagac 1860
tcggatgtgt tgctgaagct gcatgtgaag gtggcgttgt cgccctcggt caccacgagg 1920
agggcggggc tgaatgtagg tgggttccaa ggtctgtcgg gagagtcgag gaaccagccc 1980
atagcgaggg cgccggccca gagcagtggc agcaggagga gcagtggcat ggcttattta 2040
tattccaaaa aaaaaaaata aaatttcaat ttttactagt aaaaattgaa attttatttt 2100
ttttttttgg aatataaata aggtcgactt tttatgagct cgaattctat tatatttttt 2160
atctaaaaaa ctaaaaataa acattgatta aattttaata taatacttaa aaatggatgt 2220
tgtgtcgtta gataaaccgt ttatgtattt tgaggaaatt gataatgagt tagattacga 2280
accagaaagt gcaaatgagg tcgcaaaaaa actgccgtat caaggacagt taaaactatt 2340
actaggagaa ttattttttc ttagtaagtt acagcgacac ggtatattag atggtgccac 2400
cgtagtgtat ataggatctg ctcccggtac acatatacgt tatttgagag atcatttcta 2460
taatttagga gtgatcatca aatggatgct aattgacggc cgccatcatg atcctatttt 2520
aaatggattg cgtgatgtga ctctagtgac tcggttcgtt gatgaggaat atctacgatc 2580
catcaaaaaa caactgcatc cttctaagat tattttaatt tctgatgtga gatccaaacg 2640
aggaggaaat gaacctagta cggcggattt actaagtaat tacgctctac aaaatgtcat 2700
gattagtatt ttaaaccccg tggcgtctag tcttaaatgg agatgcccgt ttccagatca 2760
atggatcaag gacttttata tcccacacgg taataaaatg ttacaacctt ttgctccttc 2820
atattcagct gaaatgagat tattaagtat ttataccggt gagaacatga gactgactcg 2880
agttaccaaa tcagacgctg taaattatga aaaaaagatg tactacctta ataagatcgt 2940
ccgtaacaaa gtagttgtta actttgatta tcctaatcag gaatatgact attttcacat 3000
gtactttatg ctgaggaccg tgtactgcaa taaaacattt cctactacta aagcaaaggt 3060
actatttcta caacaatcta tatttcgttt cttaaatatt ccaacaacat caactgaaaa 3120
agttagtcat gaaccaatac aacgtaaaat atctagcaaa aattctatgt ctaaaaacag 3180
aaatagcaag agatccgtac gcagtaataa atag 3214
<210> 20
<211> 233
<212> PRT
<213> artificial sequence
<220>
<223> anti-PD-1 antibody-LC
<400> 20
Met Ala Trp Ser Pro Leu Phe Leu Thr Leu Ile Thr His Cys Ala Gly
1 5 10 15
Ser Trp Ala Glu Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser Leu
20 25 30
Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val
35 40 45
Ser Ser Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg
50 55 60
Leu Leu Ile Tyr Asp Ala Ser Asn Arg Ala Thr Gly Ile Pro Ala Arg
65 70 75 80
Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser
85 90 95
Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Ser Ser Asn
100 105 110
Trp Pro Arg Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 21
<211> 459
<212> PRT
<213> artificial sequence
<220>
<223> anti-PD-1 antibody-HC
<400> 21
Met Asp Trp Thr Trp Arg Ile Leu Phe Leu Val Ala Ala Ala Thr Gly
1 5 10 15
Ala His Ser Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln
20 25 30
Pro Gly Arg Ser Leu Arg Leu Asp Cys Lys Ala Ser Gly Ile Thr Phe
35 40 45
Ser Asn Ser Gly Met His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu
50 55 60
Glu Trp Val Ala Val Ile Trp Tyr Asp Gly Ser Lys Arg Tyr Tyr Ala
65 70 75 80
Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn
85 90 95
Thr Leu Phe Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val
100 105 110
Tyr Tyr Cys Ala Thr Asn Asp Asp Tyr Trp Gly Gln Gly Thr Leu Val
115 120 125
Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala
130 135 140
Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu
145 150 155 160
Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly
165 170 175
Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser
180 185 190
Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu
195 200 205
Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr
210 215 220
Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro
225 230 235 240
Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu Phe Pro
245 250 255
Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr
260 265 270
Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln Phe Asn
275 280 285
Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg
290 295 300
Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val
305 310 315 320
Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser
325 330 335
Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys
340 345 350
Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Gln Glu
355 360 365
Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe
370 375 380
Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu
385 390 395 400
Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe
405 410 415
Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln Glu Gly
420 425 430
Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr
435 440 445
Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455
<210> 22
<211> 5338
<212> DNA
<213> artificial sequence
<220>
<223> WR-GO-004
<400> 22
atggagaatg ttcctaatgt atactttaat cctgtgttta tagagcccac gtttaaacat 60
tctttattaa gtgtttataa acacagatta atagttttat ttgaagtatt cgttgtattc 120
attctaatat atgtattttt tagatctgaa ttaaatatgt tcttcatgcc taaacgaaaa 180
atacccgatc ctattgatag attacgacgt gctaatctag cgtgtgaaga cgataaatta 240
atgatctatg gattaccatg gatgacaact caaacatctg cgttatcaat aaatagtaaa 300
ccgatagtgt ataaagattg tgcaaagctt ttgcgatcaa taaatggatc acaaccagta 360
tctcttaacg atgttcttcg cagatgatga ttcatttttt aagtatttgg ctagtcaaga 420
tgatgaatct tcattatctg atatattgca aatcactcaa tatctagact ttctgttatt 480
attattgatc caatcaaaaa ataaattaga agccgtgggt cattgttatg aatctctttc 540
agaggaatac agacaattga caaaattcac agactttcaa gattttaaaa aactgtttaa 600
caaggtccct attgttacag atggaagggt caaacttaat aaaggatatt tgttcgactt 660
tgtgattagt ttgatgcgat tcaaaaaaga atcctctcta gctaccaccg caatagatcc 720
tgttagatac atagatcctc gtcgcaatat cgcattttct aacgtgatgg atatattaaa 780
gtcgaataaa gtgaacaata attaattctt tattgtcaag gcctagaaaa accttacttc 840
cctaggctga gactcagtga tttttgggtg taatggttgt gcaacgcttc gtgcatcacg 900
gagcaggaaa agacattacc ttcctgccaa cggcttttat caacggtcag gcgcgagtac 960
agaaagaagc tgccgtcgct gtcaagcacg ggtggagtag tcttatagtt attctctggt 1020
tggccattac tctcccactc cacggctata tcgctagggt aaaaaccctt aaccagacag 1080
gtcagactta cctggttttt agtcatttct tcctgtgagg gtggtagggt gtagacctga 1140
ggttcccgag gctgtccctt ggctttgcta attgtctttt cgatggaaga gggcaatcct 1200
ttgtttgaca ccttgcactt atattccttg ccattcagcc aatcctggtg caacactgtc 1260
agcacgctga cgacccgata ggttgaattg aattgttcct cgcggggttt cgtctttgcg 1320
ttatgcactt caacgccgtc tacgtaccag ttgaattgca cttcgggatc ctcttgagag 1380
acatcaacca cgacacaagt cacttccggg gtccggctaa tcattagggt gtccttaggt 1440
tttggtggga acagaaacac ggacggtcca cccaagaact cgggggcagg acacggaggg 1500
caggggggtc catacttgga ttcgacccgc ttatctactt tagtattaga gggcttgtgg 1560
tcaacgttac aggtataagt tttcgtgcct agactagatg aaggaacagt gaccacggaa 1620
gatagtgaat agaggcccga gctctgcagc acggcaggga atgtgtgcac tccagaggtc 1680
agggcgccgc tgttccagga cacagtcacc ggttctggaa agtagtcctt cacgagacat 1740
cctagcgcag ctgtgctttc gcttgtgctc ctactgcagg gggcgagcgg gaacactgag 1800
ggtcccttgg tgcttgccga acttacggtc accaatgtgc cctgtcccca gtaatcgtcg 1860
tttgtggcgc agtagtatac ggctgtgtcc tcagcccgca gtgaattcat ctgtaggaac 1920
aatgtatttt tagagttgtc cctactaatg gtgaaccgtc ccttcacaga gtcggcgtaa 1980
taacgcttgg agccgtcata ccagataact gccacccact ccaagccctt gcctggcgct 2040
tgccgtaccc agtgcatccc gctgttagaa aaggtgatgc cggaagcctt acagtccaat 2100
cttagtgatc tgcctggctg gacaactccg ccgcctgact caaccagctg tacttggctg 2160
tgggcgcctg ttgccgcggc aaccaagaac aggattctcc aggtccaatc catctcgagc 2220
ttatttatat tccaaaaaaa aaaaataaaa tttcaatttt taaaaaattg aaattttatt 2280
tttttttttt ggaatataaa taaggtcgac atggcatggt ccccattatt tctgaccttg 2340
attactcact gcgccggctc ttgggctgaa atcgtactca cgcagtcccc tgctactctg 2400
agtctctcac caggagaacg cgctaccctt tcttgccgtg cgtcacagtc agtatcgtcc 2460
tatctggctt ggtatcagca aaaaccaggt caggcccccc gattattgat ttatgatgca 2520
tctaaccggg ctacagggat tcctgccaga tttagcggta gcgggagtgg aactgacttc 2580
actctaacca ttagctccct tgagccagag gatttcgccg tctactactg tcagcagtct 2640
tccaactggc ctcgtacttt cggacaggga acaaaggtgg aaatcaaacg taccgtggct 2700
gcacccagcg tgttcatttt tccaccaagc gacgagcagc tcaagagcgg aaccgcatcc 2760
gtagtatgtc tcctcaataa cttctaccca cgagaagcca aagtgcagtg gaaggtggat 2820
aatgccttgc aatccggaaa cagccaagaa agcgtgaccg aacaggattc aaaagacagc 2880
acctattctc tgtccagcac attgacactg agtaaagctg attatgagaa gcacaaggtc 2940
tacgcgtgtg aggttacaca tcaaggattg tcttcaccag tcaccaagag tttcaataga 3000
ggagagtgct gagaattctt tttattaaaa attgaaattt tatttttttt ttttggaata 3060
taaataagga tgcctctgct gctgctgctg cctctgctgt gggccggcgc cctggccatg 3120
acaattcctc ctcacgtgca gaagagcgtg aataatgata tgattgtgac cgataataac 3180
ggcgccgtga aatttcccca gctgtgtaag ttttgtgatg tgagattcag cacctgtgat 3240
aatcagaagt cttgcatgtc taactgttcc atcacatcca tctgtgagaa gccacaggag 3300
gtgtgcgtgg ccgtgtggag aaagaatgat gagaatatca cacttgagac cgtgtgccac 3360
gacccaaaac tgccatacca cgattttatt ctggaggatg ccgcctctcc taagtgcatt 3420
atgaaggaga agaagaagcc aggcgagacc ttctttatgt gtagctgttc ctctgatgag 3480
tgtaatgata atattatctt cagcgaagag tataacacca gtaatcctga cctgctgctt 3540
gtgattttcc aggagcctaa gtcttgtgac aagacacaca catgccctcc ctgtccagcc 3600
cctgagctgc tgggcggccc tagcgtgttt ctgttccccc ctaagcctaa agatacactg 3660
atgattagca gaacaccaga ggtgacctgt gtggtggtgg atgtgagcca cgaggaccct 3720
gaggtgaagt ttaactggta tgtggatgga gtggaagtgc acaatgctaa gaccaagcct 3780
agagaggagc agtataatag cacatataga gtggtgtctg tgctgaccgt gctgcaccag 3840
gactggctga atggcaaaga gtataaatgt aaggtgagca acaaggccct gcccgccccc 3900
attgagaaaa ccatctctaa ggctaaggga cagccaagag agcctcaggt gtatacactg 3960
cctccttcta gagatgagct gaccaaaaac caggttagcc tgacatgcct ggtgaaaggc 4020
ttctatccaa gcgatatcgc tgtggaatgg gagagcaatg gccagcctga gaataattat 4080
aaaaccaccc caccagtgct ggatagcgat ggcagctttt tcctgtattc taagctgaca 4140
gtggataaat ctaggtggca gcagggcaat gtgtttagct gctctgtgat gcacgaggcc 4200
ctgcacaatc actatacaca gaagagcctg agcctgagcc ctggcaagtg actttttatg 4260
agctcgaatt ctattatatt ttttatctaa aaaactaaaa ataaacattg attaaatttt 4320
aatataatac ttaaaaatgg atgttgtgtc gttagataaa ccgtttatgt attttgagga 4380
aattgataat gagttagatt acgaaccaga aagtgcaaat gaggtcgcaa aaaaactgcc 4440
gtatcaagga cagttaaaac tattactagg agaattattt tttcttagta agttacagcg 4500
acacggtata ttagatggtg ccaccgtagt gtatatagga tctgctcccg gtacacatat 4560
acgttatttg agagatcatt tctataattt aggagtgatc atcaaatgga tgctaattga 4620
cggccgccat catgatccta ttttaaatgg attgcgtgat gtgactctag tgactcggtt 4680
cgttgatgag gaatatctac gatccatcaa aaaacaactg catccttcta agattatttt 4740
aatttctgat gtgagatcca aacgaggagg aaatgaacct agtacggcgg atttactaag 4800
taattacgct ctacaaaatg tcatgattag tattttaaac cccgtggcgt ctagtcttaa 4860
atggagatgc ccgtttccag atcaatggat caaggacttt tatatcccac acggtaataa 4920
aatgttacaa ccttttgctc cttcatattc agctgaaatg agattattaa gtatttatac 4980
cggtgagaac atgagactga ctcgagttac caaatcagac gctgtaaatt atgaaaaaaa 5040
gatgtactac cttaataaga tcgtccgtaa caaagtagtt gttaactttg attatcctaa 5100
tcaggaatat gactattttc acatgtactt tatgctgagg accgtgtact gcaataaaac 5160
atttcctact actaaagcaa aggtactatt tctacaacaa tctatatttc gtttcttaaa 5220
tattccaaca acatcaactg aaaaagttag tcatgaacca atacaacgta aaatatctag 5280
caaaaattct atgtctaaaa acagaaatag caagagatcc gtacgcagta ataaatag 5338
<210> 23
<211> 5
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-HCDR 1
<222> (1)..(5)
<400> 23
Asn Ser Gly Met His
1 5
<210> 24
<211> 17
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-HCDR 2
<222> (1)..(17)
<400> 24
Val Ile Trp Tyr Asp Gly Ser Lys Arg Tyr Tyr Ala Asp Ser Val Lys
1 5 10 15
Gly
<210> 25
<211> 4
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-HCDR 3
<222> (1)..(4)
<400> 25
Asn Asp Asp Tyr
1
<210> 26
<211> 11
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-LCDR 1
<222> (1)..(11)
<400> 26
Arg Ala Ser Gln Ser Val Ser Ser Tyr Leu Ala
1 5 10
<210> 27
<211> 7
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-LCDR 2
<222> (1)..(7)
<400> 27
Asp Ala Ser Asn Arg Ala Thr
1 5
<210> 28
<211> 9
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-LCDR 3
<222> (1)..(9)
<400> 28
Gln Gln Ser Ser Asn Trp Pro Arg Thr
1 5
<210> 29
<211> 113
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-VH
<222> (1)..(113)
<400> 29
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Asp Cys Lys Ala Ser Gly Ile Thr Phe Ser Asn Ser
20 25 30
Gly Met His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Val Ile Trp Tyr Asp Gly Ser Lys Arg Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Phe
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Thr Asn Asp Asp Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser
100 105 110
Ser
<210> 30
<211> 110
<212> PRT
<213> Chile person
<220>
<221> anti-PD-1 antibody-VL
<222> (1)..(110)
<400> 30
Glu Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Ser Ser Tyr
20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45
Tyr Asp Ala Ser Asn Arg Ala Thr Gly Ile Pro Ala Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Glu Pro
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Ser Ser Asn Trp Pro Arg
85 90 95
Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Arg Thr Val
100 105 110
<210> 31
<211> 15
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-HCDR 1
<222> (1)..(15)
<400> 31
aacagcggga tgcac 15
<210> 32
<211> 51
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-HCDR 2
<222> (1)..(51)
<400> 32
gttatctggt atgacggctc caagcgttat tacgccgact ctgtgaaggg a 51
<210> 33
<211> 12
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-HCDR 3
<222> (1)..(12)
<400> 33
aacgacgatt ac 12
<210> 34
<211> 33
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-LCDR 1
<222> (1)..(33)
<400> 34
cgtgcgtcac agtcagtatc gtcctatctg gct 33
<210> 35
<211> 21
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-LCDR 2
<222> (1)..(21)
<400> 35
gatgcatcta accgggctac a 21
<210> 36
<211> 27
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-LCDR 3
<222> (1)..(27)
<400> 36
cagcagtctt ccaactggcc tcgtact 27
<210> 37
<211> 339
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-VH
<222> (1)..(339)
<400> 37
caagtacagc tggttgagtc aggcggcgga gttgtccagc caggcagatc actaagattg 60
gactgtaagg cttccggcat caccttttct aacagcggga tgcactgggt acggcaagcg 120
ccaggcaagg gcttggagtg ggtggcagtt atctggtatg acggctccaa gcgttattac 180
gccgactctg tgaagggacg gttcaccatt agtagggaca actctaaaaa tacattgttc 240
ctacagatga attcactgcg ggctgaggac acagccgtat actactgcgc cacaaacgac 300
gattactggg gacagggcac attggtgacc gtaagttcg 339
<210> 38
<211> 330
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-VL
<222> (1)..(330)
<400> 38
gaaatcgtac tcacgcagtc ccctgctact ctgagtctct caccaggaga acgcgctacc 60
ctttcttgcc gtgcgtcaca gtcagtatcg tcctatctgg cttggtatca gcaaaaacca 120
ggtcaggccc cccgattatt gatttatgat gcatctaacc gggctacagg gattcctgcc 180
agatttagcg gtagcgggag tggaactgac ttcactctaa ccattagctc ccttgagcca 240
gaggatttcg ccgtctacta ctgtcagcag tcttccaact ggcctcgtac tttcggacag 300
ggaacaaagg tggaaatcaa acgtaccgtg 330
<210> 39
<211> 1379
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-HC
<222> (1)..(983)
<400> 39
atggattgga cctggagaat cctgttcttg gttgccgcgg caacaggcgc ccacagccaa 60
gtacagctgg ttgagtcagg cggcggagtt gtccagccag gcagatcact aagattggac 120
tgtaaggctt ccggcatcac cttttctaac agcgggatgc actgggtacg gcaagcgcca 180
ggcaagggct tggagtgggt ggcagttatc tggtatgacg gctccaagcg ttattacgcc 240
gactctgtga agggacggtt caccattagt agggacaact ctaaaaatac attgttccta 300
cagatgaatt cactgcgggc tgaggacaca gccgtatact actgcgccac aaacgacgat 360
tactggggac agggcacatt ggtgaccgta agttcggcaa gcaccaaggg accctcagtg 420
ttcccgctcg ccccctgcag taggagcaca agcgaaagca cagctgcgct aggatgtctc 480
gtgaaggact actttccaga accggtgact gtgtcctgga acagcggcgc cctgacctct 540
ggagtgcaca cattccctgc cgtgctgcag agctcgggcc tctattcact atcttccgtg 600
gtcactgttc cttcatctag tctaggcacg aaaacttata cctgtaacgt tgaccacaag 660
ccctctaata ctaaagtaga taagcgggtc gaatccaagt atggaccccc ctgccctccg 720
tgtcctgccc ccgagttctt gggtggaccg tccgtgtttc tgttcccacc aaaacctaag 780
gacaccctaa tgattagccg gaccccggaa gtgacttgtg tcgtggttga tgtctctcaa 840
gaggatcccg aagtgcaatt caactggtac gtagacggcg ttgaagtgca taacgcaaag 900
acgaaacccc gcgaggaaca attcaattca acctatcggg tcgtcagcgt gctgacagtg 960
ttgcaccagg attggctgaa tggcaaggaa tataagtgca aggtgtcaaa caaaggattg 1020
ccctcttcca tcgaaaagac aattagcaaa gccaagggac agcctcggga acctcaggtc 1080
tacaccctac caccctcaca ggaagaaatg actaaaaacc aggtaagtct gacctgtctg 1140
gttaagggtt tttaccctag cgatatagcc gtggagtggg agagtaatgg ccaaccagag 1200
aataactata agactactcc acccgtgctt gacagcgacg gcagcttctt tctgtactcg 1260
cgcctgaccg ttgataaaag ccgttggcag gaaggtaatg tcttttcctg ctccgtgatg 1320
cacgaagcgt tgcacaacca ttacacccaa aaatcactga gtctcagcct agggaagta 1379
<210> 40
<211> 702
<212> DNA
<213> Chile person
<220>
<221> anti-PD-1 antibody-LC
<222> (1)..(315)
<400> 40
atggcatggt ccccattatt tctgaccttg attactcact gcgccggctc ttgggctgaa 60
atcgtactca cgcagtcccc tgctactctg agtctctcac caggagaacg cgctaccctt 120
tcttgccgtg cgtcacagtc agtatcgtcc tatctggctt ggtatcagca aaaaccaggt 180
caggcccccc gattattgat ttatgatgca tctaaccggg ctacagggat tcctgccaga 240
tttagcggta gcgggagtgg aactgacttc actctaacca ttagctccct tgagccagag 300
gatttcgccg tctactactg tcagcagtct tccaactggc ctcgtacttt cggacaggga 360
acaaaggtgg aaatcaaacg taccgtggct gcacccagcg tgttcatttt tccaccaagc 420
gacgagcagc tcaagagcgg aaccgcatcc gtagtatgtc tcctcaataa cttctaccca 480
cgagaagcca aagtgcagtg gaaggtggat aatgccttgc aatccggaaa cagccaagaa 540
agcgtgaccg aacaggattc aaaagacagc acctattctc tgtccagcac attgacactg 600
agtaaagctg attatgagaa gcacaaggtc tacgcgtgtg aggttacaca tcaaggattg 660
tcttcaccag tcaccaagag tttcaataga ggagagtgct ga 702

Claims (76)

1. A modified oncolytic virus comprising a viral genome having a first heterologous polynucleotide encoding a first molecule capable of inhibiting an interaction between PD-1 and PD-L1 and a second heterologous polynucleotide encoding a second molecule capable of inhibiting TGF- β signaling.
2. The modified oncolytic virus of claim 1, wherein the oncolytic virus is selected from the group consisting of vaccinia virus, adenovirus, reovirus, herpes simplex virus, semliki forest virus, venezuelan equine encephalitis virus, parvovirus, chicken anemia virus, measles virus, coxsackie virus, vesicular stomatitis virus, saint valley virus, maraba virus, newcastle disease virus, and myxoma virus.
3. The modified oncolytic virus of claim 2, wherein the oncolytic virus is a vaccinia virus.
4. The modified oncolytic virus of claim 1, wherein the modified oncolytic virus is attenuated and capable of replication in tumor cells.
5. The modified oncolytic virus of claim 1, wherein the viral genome comprises at least one deletion or disruption such that the virus is capable of selective replication in tumor cells.
6. The modified oncolytic virus of claim 5, wherein the deletion or disruption is in an Open Reading Frame (ORF) encoding at least a portion of an enzyme that is both critical for replication of the virus and preferentially expressed in tumor cells over non-tumor cells.
7. The modified oncolytic virus of claim 6, wherein the enzyme is a kinase.
8. The modified oncolytic virus of claim 7, wherein the enzyme is thymidine kinase.
9. The modified oncolytic virus of claim 2, wherein the oncolytic virus is derived from a Western Reserve strain.
10. The modified oncolytic virus of claim 4, wherein the first heterologous polynucleotide and the second heterologous polynucleotide are inserted in place of the deletion.
11. The modified oncolytic virus of claim 1, wherein the first heterologous polynucleotide and the second heterologous polynucleotide are configured such that they are expressed in the same or different phases of the replication cycle of the modified oncolytic virus.
12. The modified oncolytic virus of claim 1, wherein the first molecule is a first fusion protein and the second molecule is a second fusion protein.
13. The modified oncolytic virus of claim 12, wherein the first heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: a first promoter-a polynucleotide encoding the first fusion protein-a first stop codon, and the second heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: second promoter-polynucleotide encoding the second fusion protein-second stop codon.
14. The modified oncolytic virus of claim 13, wherein the first fusion protein expressed from the first heterologous polynucleotide and the second fusion protein expressed from the second heterologous polynucleotide are expressed as separate proteins.
15. The modified oncolytic virus of claim 13, wherein the first heterologous polynucleotide is immediately upstream or immediately downstream of the second heterologous polynucleotide.
16. The modified oncolytic virus of claim 13, wherein the first promoter is capable of driving expression of the first fusion protein and the second promoter is capable of driving expression of the second fusion protein, wherein the first and second promoters are in a head-to-head orientation.
17. The modified oncolytic virus of claim 13, wherein the first promoter and the second promoter are the same or different.
18. The modified oncolytic virus of claim 17, wherein the first promoter and the second promoter are both early and late promoters.
19. The modified oncolytic virus of claim 18, wherein the early and late promoters are pses/ls.
20. The modified oncolytic virus of claim 13, wherein the first stop codon and the second stop codon are the same or different.
21. The modified oncolytic virus of claim 12, wherein the first fusion protein comprises a PD-1 extracellular domain (PD-1 ECD).
22. The modified oncolytic virus of claim 21, wherein the first fusion protein further comprises a first immunoglobulin Fc region.
23. The modified oncolytic virus of claim 22, wherein the first immunoglobulin Fc region is a first human IgG1 Fc region.
24. The modified oncolytic virus of claim 23, wherein the PD-1ECD is operably linked to the first immunoglobulin Fc region at the C-terminus of the PD-1 ECD.
25. The modified oncolytic virus of claim 21, wherein the first fusion protein further comprises a signal peptide.
26. The modified oncolytic virus of claim 25, wherein the signal peptide is operably linked to the PD-1ECD at the C-terminus of the signal peptide.
27. The modified oncolytic virus of claim 25, wherein the signal peptide is a CD33 signal peptide.
28. The modified oncolytic virus of claim 21, wherein the PD-1ECD comprises the amino acid sequence of SEQ ID NO:1 or a homologous sequence thereof having at least 80% sequence identity.
29. The modified oncolytic virus of claim 28, wherein the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID No. 7 or a homologous sequence thereof having at least 80% sequence identity.
30. The modified oncolytic virus of claim 23, wherein the first human IgG1 Fc region comprises the amino acid sequence of SEQ ID No. 3 or a homologous sequence thereof having at least 80% sequence identity.
31. The modified oncolytic virus of claim 30, wherein the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO 9 or a homologous sequence thereof having at least 80% sequence identity.
32. The modified oncolytic virus of claim 27, wherein the CD33 signal peptide comprises the amino acid sequence of SEQ ID No. 4 or a homologous sequence thereof having at least 80% sequence identity.
33. The modified oncolytic virus of claim 32, wherein the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID No. 11 or a homologous sequence thereof having at least 80% sequence identity.
34. The modified oncolytic virus of claim 12, wherein the first fusion protein comprises the amino acid sequence of SEQ ID No. 5 or a homologous sequence thereof having at least 80% sequence identity.
35. The modified oncolytic virus of claim 34, wherein the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID No. 13 or a homologous sequence thereof having at least 80% sequence identity.
36. The modified oncolytic virus of claim 1, wherein the first molecule is an anti-PD-1 antibody and the second molecule is a second fusion protein.
37. The modified oncolytic virus of claim 36, wherein the anti-PD-1 antibody comprises:
HCDR1 having the amino acid sequence of SEQ ID NO. 23 or a homologous sequence having at least 80% sequence identity,
HCDR2 having the amino acid sequence of SEQ ID NO. 24 or a homologous sequence having at least 80% sequence identity,
HCDR3 having the amino acid sequence of SEQ ID NO. 25 or a homologous sequence having at least 80% sequence identity,
LCDR1 having the amino acid sequence of SEQ ID NO. 26 or a homologous sequence having at least 80% sequence identity,
LCDR2 having the amino acid sequence of SEQ ID NO 27 or a homologous sequence thereof having at least 80% sequence identity, and
LCDR3 having the amino acid sequence of SEQ ID NO. 28 or a homologous sequence thereof having at least 80% sequence identity.
38. The modified oncolytic virus of claim 37, wherein
The HCDR1 is encoded by the nucleic acid sequence of SEQ ID NO. 31 or a homologous sequence thereof having at least 80% sequence identity,
the HCDR2 is encoded by the nucleic acid sequence of SEQ ID NO. 32 or a homologous sequence thereof having at least 80% sequence identity,
the HCDR3 is encoded by the nucleic acid sequence of SEQ ID NO. 33 or a homologous sequence thereof having at least 80% sequence identity,
the LCDR1 is encoded by the nucleic acid sequence of SEQ ID NO. 34 or a homologous sequence thereof having at least 80% sequence identity,
the LCDR2 is encoded by the nucleic acid sequence of SEQ ID NO. 35 or a homologous sequence thereof having at least 80% sequence identity, an
The LCDR3 is encoded by the nucleic acid sequence of SEQ ID NO. 36 or a homologous sequence thereof having at least 80% sequence identity.
39. The modified oncolytic virus of claim 36, wherein the anti-PD-1 antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID No. 29 or a homologous sequence thereof having at least 80% sequence identity and a light chain variable region having the amino acid sequence of SEQ ID No. 30 or a homologous sequence thereof having at least 80% sequence identity.
40. The modified oncolytic virus of claim 39, wherein the anti-PD-1 antibody heavy chain variable region is encoded by the nucleic acid sequence of SEQ ID NO:37 or a homologous sequence thereof having at least 80% sequence identity and the anti-PD-1 antibody light chain variable region is encoded by the nucleic acid sequence of SEQ ID NO:38 or a homologous sequence thereof having at least 80% sequence identity.
41. The modified oncolytic virus of claim 36, wherein the anti-PD-1 antibody comprises a full length heavy chain having the amino acid sequence of SEQ ID No. 21 or a homologous sequence thereof having at least 80% sequence identity and a full length light chain having the amino acid sequence of SEQ ID No. 20 or a homologous sequence thereof having at least 80% sequence identity.
42. The modified oncolytic virus of claim 41, wherein the anti-PD-1 antibody full length heavy chain is encoded by the nucleic acid sequence of SEQ ID No. 39 or a homologous sequence thereof having at least 80% sequence identity and the anti-PD-1 antibody full length light chain is encoded by the nucleic acid sequence of SEQ ID No. 40 or a homologous sequence thereof having at least 80% sequence identity.
43. The modified oncolytic virus of claim 36, wherein the first heterologous polynucleotide further comprises a third heterologous polynucleotide and a fourth heterologous polynucleotide, wherein the third heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: third promoter-polynucleotide encoding the heavy chain of the anti-PD-1 antibody-third stop codon, and wherein the fourth heterologous polynucleotide comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: fourth promoter-polynucleotide encoding the anti-PD-1 antibody light chain-fourth stop codon; and the second heterologous polynucleotide further comprises the following in-frame elements in the 5 'to 3' orientation of the sense strand: second promoter-polynucleotide encoding the second fusion protein-second stop codon.
44. The modified oncolytic virus of claim 43, wherein the third heterologous polynucleotide is immediately upstream or immediately downstream of the fourth heterologous polynucleotide.
45. The modified oncolytic virus of claim 44, wherein the third promoter is capable of driving expression of the anti-PD-1 antibody heavy chain and the fourth promoter is capable of driving expression of the anti-PD-1 antibody light chain, wherein the third promoter and the fourth promoter are in a head-to-head orientation.
46. The modified oncolytic virus of claim 36, wherein the first heterologous polynucleotide is immediately upstream or immediately downstream of the second heterologous polynucleotide.
47. The modified oncolytic virus of claim 46, wherein the anti-PD-1 antibody expressed from the first heterologous polynucleotide and the second fusion protein expressed from the second heterologous polynucleotide are expressed as separate proteins.
48. The modified oncolytic virus of claim 42, wherein the second promoter, the third promoter, and the fourth promoter are the same or different.
49. The modified oncolytic virus of claim 47, wherein the second promoter, the third promoter, and the fourth promoter are all early and late promoters.
50. The modified oncolytic virus of claim 48, wherein the early and late promoters are pSE/L.
51. The modified oncolytic virus of claim 42, wherein the second stop codon, the third stop codon, and the fourth stop codon are the same or different.
52. The modified oncolytic virus of claim 12 or 36, wherein the second fusion protein comprises a TGF- β receptor II extracellular domain (TGFBRII ECD).
53. The modified oncolytic virus of claim 52, wherein the second fusion protein further comprises a second immunoglobulin Fc region.
54. The modified oncolytic virus of claim 53, wherein the immunoglobulin Fc region is a second human IgG1Fc region.
55. The modified oncolytic virus of claim 54, wherein the TGFBRII ECD is operably linked to the second immunoglobulin Fc region at the C-terminus of the TGFBRIIECD.
56. The modified oncolytic virus of claim 52, wherein the second fusion protein further comprises a signal peptide.
57. The modified oncolytic virus of claim 56, wherein said signal peptide is operably linked to said TGFBRII ECD at the C-terminus of said signal peptide.
58. The modified oncolytic virus of claim 56, wherein the signal peptide is a CD33 signal peptide.
59. A modified oncolytic virus of claim 52, wherein said TGFBRII ECD comprises the amino acid sequence of SEQ ID NO. 2 or a homologous sequence thereof having at least 80% sequence identity.
60. The modified oncolytic virus of claim 59, wherein the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO. 8 or a homologous sequence thereof having at least 80% sequence identity.
61. The modified oncolytic virus of claim 54, wherein the second human IgG1 Fc region comprises the amino acid sequence of SEQ ID No. 3 or a homologous sequence thereof having at least 80% sequence identity.
62. The modified oncolytic virus of claim 61, wherein the amino acid sequence is encoded by the nucleic acid sequence of SEQ ID NO. 10 or a homologous sequence thereof having at least 80% sequence identity.
63. The modified oncolytic virus of claim 58, wherein the CD33 signal peptide comprises the amino acid sequence of SEQ ID No. 4 or a homologous sequence thereof having at least 80% sequence identity.
64. The modified oncolytic virus of claim 63, wherein the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID No. 12 or a homologous sequence thereof having at least 80% sequence identity.
65. The modified oncolytic virus of claim 12 or 36, wherein said second fusion protein comprises the amino acid sequence of SEQ ID No. 6 or a homologous sequence thereof having at least 80% sequence identity.
66. The modified oncolytic virus of claim 65, wherein the amino acid sequence is encoded by a nucleic acid sequence comprising SEQ ID No. 14 or a homologous sequence thereof having at least 80% sequence identity.
67. The modified oncolytic virus of claim 12, wherein the first fusion protein and the second fusion protein are capable of forming dimers.
68. The modified oncolytic virus of claim 67, wherein the dimer is formed by association of the first immunoglobulin Fc region with the second immunoglobulin Fc region.
69. The modified oncolytic virus of claim 1, having the nucleic acid sequence of SEQ ID No. 17 or SEQ ID No. 22.
70. A pharmaceutical composition comprising the modified oncolytic virus of any one of claims 1-69 and a pharmaceutically acceptable carrier.
71. A method of treating a tumor, the method comprising administering to a subject an effective amount of the modified oncolytic virus of any one of claims 1-69 or the pharmaceutical composition of claim 70.
72. The method of claim 71, wherein the subject is a human.
73. The method of claim 71, wherein the tumor is a solid tumor.
74. The method of claim 71, wherein the tumor is melanoma, non-small cell lung cancer, renal cell carcinoma, hodgkin's lymphoma, head and neck squamous cell carcinoma, bladder cancer, colorectal cancer, triple negative breast cancer, hepatocellular carcinoma, pancreatic cancer, ovarian cancer, colon cancer, pharyngeal squamous cell carcinoma, or ovarian teratoma.
75. The method of claim 71, wherein the route of administration is topical.
76. The method of claim 75, wherein the route of administration is intratumoral injection.
CN202180083706.7A 2020-12-11 2021-12-10 Modified oncolytic virus, compositions comprising modified oncolytic virus and uses thereof Pending CN117222436A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CNPCT/CN2020/135585 2020-12-11
CN2020135585 2020-12-11
PCT/CN2021/137194 WO2022122026A1 (en) 2020-12-11 2021-12-10 A modified oncolytic virus, composition and use thereof

Publications (1)

Publication Number Publication Date
CN117222436A true CN117222436A (en) 2023-12-12

Family

ID=81973099

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180083706.7A Pending CN117222436A (en) 2020-12-11 2021-12-10 Modified oncolytic virus, compositions comprising modified oncolytic virus and uses thereof

Country Status (2)

Country Link
CN (1) CN117222436A (en)
WO (1) WO2022122026A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106397592A (en) * 2015-07-31 2017-02-15 苏州康宁杰瑞生物科技有限公司 Single-domain antibody directed at programmed death ligand (PD-L1) and derived protein thereof
CN108165536A (en) * 2017-12-11 2018-06-15 浙江大学 A kind of recombination oncolytic vaccinia virus and preparation method and application
CN111770936A (en) * 2018-01-12 2020-10-13 百时美施贵宝公司 Combination therapy of anti-IL-8 and anti-PD-1 antibodies for the treatment of cancer
CN111826395A (en) * 2019-04-18 2020-10-27 艾生命序公司 Recombinant oncolytic virus expression anti-immune checkpoint fusion antibody and immunostimulatory molecule
CN111690070A (en) * 2020-05-13 2020-09-22 深圳市众循精准医学研究院 sPD-1-Fc-sTGF beta RII fusion protein and application thereof
CN113501879B (en) * 2021-06-30 2022-09-06 拜盖特生物科技(上海)有限公司 Bifunctional antibody for relieving immunosuppression in tumor immune microenvironment, and application and preparation method thereof

Also Published As

Publication number Publication date
WO2022122026A1 (en) 2022-06-16

Similar Documents

Publication Publication Date Title
KR102338453B1 (en) Methods and compositions comprising a combination of a vegf antagonist and an anti-ctla-4 antibody
CN109053895B (en) Bifunctional antibody for resisting PD-1-VEGFA, pharmaceutical composition and application thereof
AU2023200891A1 (en) Novel anti-PD-1 antibodies
KR101837053B1 (en) Cross-species-specific pscaxcd3, cd19xcd3, c-metxcd3, endosialinxcd3, epcamxcd3, igf-1rxcd3 or fapalphaxcd3 bispecific single chain antibody
TWI688572B (en) Multivalent molecules comprising dr5-binding domains
JP7282401B2 (en) Use of Anti-FAM19A5 Antibodies for Cancer Therapy
JP2018523493A (en) Multivalent and multispecific GITR binding fusion proteins
KR20200128385A (en) Recombinant human sialidase, sialidase fusion protein, and methods of using the same
AU2020390967A1 (en) Anti-PD-1-anti-VEGFA bispecific antibody, pharmaceutical composition and use thereof
CN108424461B (en) CD47-CAR-T cells
KR20230084507A (en) Multi-specific immune targeting molecules and uses thereof
KR20220032576A (en) Recombinant human sialidase, sialidase fusion proteins, and methods of using the same
CN113164538A (en) Modified oncolytic viruses, compositions and uses thereof
KR20210143096A (en) Antibody specific for CD22 and uses thereof
EP4357365A1 (en) Antibody targeting axl protein and antigen binding fragment thereof, and preparation method therefor and application thereof
KR20230093437A (en) Vectorized anti-TNF-α antibodies for ocular indications
CN112912398A (en) Novel antibodies and nucleotide sequences and uses thereof
CN117222436A (en) Modified oncolytic virus, compositions comprising modified oncolytic virus and uses thereof
CN115073588A (en) Method for improving safety of medicine containing immunoglobulin Fc fragment
TW202241943A (en) Tau-specific antibody gene therapy compositions, methods and uses thereof
KR20230059789A (en) Anti-Variable MUC1* Antibodies and Uses Thereof
KR20230034320A (en) Recombinant sialidase with reduced protease sensitivity, sialidase fusion proteins, and methods of using the same
CA3157427A1 (en) Chimeric antigen receptor spacers
CN115772544B (en) AAV vectors against VEGF-A and ANG-2
CN114369171B (en) Chimeric antigen receptor fusing CXCR3 and alpha FR antibodies, effector cells thereof and application thereof in treatment of ovarian cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination