CN113684214B - Preparation and application of tumor vaccine based on attenuated listeria - Google Patents

Preparation and application of tumor vaccine based on attenuated listeria Download PDF

Info

Publication number
CN113684214B
CN113684214B CN202010420039.4A CN202010420039A CN113684214B CN 113684214 B CN113684214 B CN 113684214B CN 202010420039 A CN202010420039 A CN 202010420039A CN 113684214 B CN113684214 B CN 113684214B
Authority
CN
China
Prior art keywords
recombinant
cells
tumor
gly gly
nucleic acid
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.)
Active
Application number
CN202010420039.4A
Other languages
Chinese (zh)
Other versions
CN113684214A (en
Inventor
代楠
韩毅峰
赵勇刚
张可
薛亚东
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 Ruotai Pharmaceutical Technology Co ltd
Suzhou Royaltech Med Co ltd
Original Assignee
Shanghai Ruotai Pharmaceutical Technology Co ltd
Suzhou Royaltech Med 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 Ruotai Pharmaceutical Technology Co ltd, Suzhou Royaltech Med Co ltd filed Critical Shanghai Ruotai Pharmaceutical Technology Co ltd
Priority to CN202010420039.4A priority Critical patent/CN113684214B/en
Publication of CN113684214A publication Critical patent/CN113684214A/en
Application granted granted Critical
Publication of CN113684214B publication Critical patent/CN113684214B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present disclosure relates to the preparation and use of attenuated listeria-based tumor vaccines. In particular, the disclosure relates to a recombinant nucleic acid molecule, a recombinant plasmid or recombinant expression vector containing the recombinant nucleic acid molecule, a recombinant protein and a recombinant listeria. At the same time, the present disclosure also relates to pharmaceutical compositions, vaccines, and uses thereof comprising the above ingredients, and further provides methods of sustained killing of tumors and methods of inducing multi-target immune responses in a subject.

Description

Preparation and application of tumor vaccine based on attenuated listeria
Technical Field
The present disclosure relates generally to the field of biotechnology. In particular, the present disclosure provides for the preparation and use of a personalized tumor vaccine. More specifically, the present disclosure provides for the preparation and use of a multi-target personalized tumor vaccine based on attenuated listeria.
Background
In recent years, with the deep search for anti-tumor immune responses, tumor immune escape mechanisms, tumor microenvironment, etc., personalized tumor immunotherapy has become a very active field of cancer research [1]. The main research directions of the current personalized immunotherapy of tumors are tumor immune checkpoint inhibitors, adoptive cell immunotherapy, oncolytic viruses and tumor vaccines [2]. Tumor immune checkpoint inhibitors (Immune Checkpoint Inhibitors, ICIs) induce potent anti-tumor immune responses by eliminating immunosuppressive factors, and have shown exciting results in clinical therapies, but ICI has a low response rate to various cancer patients as a single drug [3-4]. Adoptive cellular immunotherapy (Adoptive Cell Transfer, ACT) involves isolation of immunocompetent cells from tumor patients, in vitro expansion and functional identification, and then delivery to patients, either directly killing the tumor or stimulating an immune response of the organism to kill the tumor, but ACT requires very high selection of appropriate targets, and the cost of reinfusion after cell in vitro culture expansion is quite significant [5-6]. Oncolytic viruses are a class of tumor killing viruses with replication capacity, which can selectively infect and replicate in tumor cells to kill the tumor cells and stimulate the body to generate specific anti-tumor immune response. At present, how to find safe doses capable of exerting the maximum anticancer effect and how to find suitable biomarkers to screen patients becomes a urgent need for oncolytic viruses [7]. Tumor vaccine overcomes the immunosuppression state caused by tumor by expressing specific tumor antigen (such as polypeptide, DNA, RNA, etc.) with immunogenicity, with the help of cytokine, chemotactic factor, etc., activates or strengthens the body's own anti-tumor immunity, induces body's cellular immunity and humoral immunity response, and further kills and eliminates tumor cells [8]. The main reasons for restricting the development of tumor vaccines are that the body has weak immunogenicity to self antigens and is easy to cause tumor immune tolerance and immune escape. The neoantigen is generated by the gene mutation of tumor cells, has tumor specificity and strong immunogenicity, and can induce the immune response of T cells, so that the development of the neoantigen is an important direction for the development of personalized tumor vaccines [9]. However, in personalized treatment, a plurality of neoantigens with higher scores are often selected as targets for treatment, so whether a personalized tumor vaccine carrier can efficiently load a plurality of targets becomes a key of technical implementation.
At the same time, the tumor treatment schemes existing in the prior art have the following defects:
for tumor immune checkpoint inhibitors, they cannot specifically activate tumor-specific T cell responses and ICI as a single drug has a low response rate to various cancer patients; for adoptive cell immunotherapy, the selection requirement on the suitable target point is very high, the tumor cells can not be effectively identified, and the cost of reinfusion after in-vitro culture and amplification of the cells is high; in the case of oncolytic viruses, the occurrence mechanism of the toxic and side effects of the oncolytic viruses is not clear, and the application of the oncolytic viruses to the immune system of the organism is greatly influenced.
Prior art literature:
[1]Topalian S L,Weiner G J,Pardoll D M.Cancer immunotherapy comes of age.[J].Nature Clinical Practice Oncology,2011,2(3):115.
[2]Yoshitaro Shindo,Shoichi Hazama,Ryouichi Tsunedomi.Novel technologies and emerging biomarkers for personalized cancer immunotherapy[J].Journal for ImmunoTherapy of Cancer,2016,4(1):3.
[3]Koji K.Advances in cancer immunotherapy for gastroenterological malignancy[J].Annals of Gastroenterological Surgery,2018,2(4):244-245.
[4]Hodi F S,O'Day S J,Mcdermott D F,et al.Improved Survival with Ipilimumab in Patients with Metastatic Melanoma[J].New England Journal of Medicine,2010,363(8):711-723.
[5]Rosenberg S A,Restifo N P,Yang J C,et al.Adoptive cell transfer:a clinical path to effective cancer immunotherapy[J].NATURE REVIEWS CANCER,2008,8(4):299-308.
[6]Rosenberg S A,Restifo N P.Adoptive cell transfer as personalized immunotherapy for human cancer[J].Science,2015,348(6230):62-68.
[7]Lun X,Yang W,Alain T,et al.Myxoma virus is a novel oncolytic virus with significant antitumor activity against experimental human gliomas.[J].Cancer Research,2005,65(21):9982-9990.
[8]Chakraborty N G,Chattopadhyay S,Mehrotra S,et al.Regulatory T-cell response and tumor vaccine-induced cytotoxic T lymphocytes in human melanoma[J].Human Immunology,2004,65(8):0-802.
[9]Ott P A,Hu Z,Keskin D B,et al.An immunogenic personal neoantigen vaccine for patients with melanoma[J].Nature,2017,547(7662):217.
disclosure of Invention
Problems to be solved by the invention
The invention constructs a non-integrated plasmid expression multi-target tumor vaccine based on listeria, which can improve the immunogenicity of tumor antigens, fully activate various tumor-specific T cells in vivo, simultaneously identify a plurality of targets of a certain tumor or different targets of various tumors, improve the curative effect of the tumor vaccine and lay a foundation for the application of the personalized tumor vaccine of the neoantigens.
Solution for solving the problem
The present disclosure provides the following technical solutions.
(1) A recombinant nucleic acid molecule comprising an open reading frame encoding a recombinant polypeptide comprising a heterologous antigen fused to a derivatized listeriolysin (LLO) polypeptide, wherein the amino acid sequence of the heterologous antigen is a polypeptide having the amino acid sequence depicted in SEQ ID No. 2 substituted, repeated, deleted, or added with one or more amino acids and having or partially having the activity of a heterologous antigen depicted in SEQ ID No. 2.
(2) The recombinant nucleic acid molecule according to (1), wherein the amino acid sequence of the heterologous antigen is a sequence having at least 80% identity to the amino acid sequence shown in SEQ ID NO. 2.
(3) The recombinant nucleic acid molecule according to any one of (1) - (2), wherein the nucleotide sequence encoding the heterologous antigen is as set forth in SEQ ID NO:1, and a sequence shown in 1.
(4) The recombinant nucleic acid molecule according to any one of (1) to (3), wherein the sequence of the recombinant nucleic acid molecule is as set forth in SEQ ID NO: 4.
(5) A recombinant plasmid or recombinant expression vector comprising the sequence of the recombinant nucleic acid molecule according to any one of (1) to (4).
(6) A recombinant protein encoded by the recombinant nucleic acid molecule of any one of (1) to (4), or formed by the recombinant plasmid or recombinant expression vector of (5).
(7) A recombinant Listeria comprising the recombinant nucleic acid molecule of any one of (1) - (4), or comprising the recombinant plasmid or recombinant expression vector of (5), or expressing the recombinant protein of (6).
(8) A pharmaceutical composition comprising a therapeutically effective amount of the recombinant Listeria according to (7).
(9) A prophylactic or therapeutic vaccine, characterized in that it comprises a prophylactically or therapeutically effective amount of the recombinant Listeria according to (7).
(10) The recombinant listeria of (7), (8) said pharmaceutical composition or (9) said vaccine for use in the manufacture of a medicament for killing cells.
(11) The use according to (10), wherein the cells are selected from the group consisting of proliferative, neoplastic, precancerous, or metastatic cells; preferably, the cell is selected from the group consisting of metastatic cells; more preferably, the metastatic cells are selected from metastatic tumor cells.
(12) The use of the recombinant listeria of (7), (8) said pharmaceutical composition or (9) said vaccine in the manufacture of a medicament for treating a patient with a tumor.
(13) A method of sustaining killing a cell comprising contacting said cell with (7) said recombinant listeria, (8) said pharmaceutical composition or (9) said vaccine; preferably, the cell is a tumor cell.
(14) A method of inducing an immune response in a subject, the method comprising administering to the subject: (7) The recombinant listeria, (8) the pharmaceutical composition or (9) the vaccine.
(15) An isolated peptide consisting of an amino acid sequence selected from the group consisting of those that are conservatively mutated and which comprise a sequence identical to SEQ ID NO:2 or SEQ ID NO:4, a sequence having at least 80% identity to the amino acid sequence of seq id no; or the amino acid sequence is selected from the group consisting of SEQ ID NOs: 2 or SEQ ID NO: 4.
(16) A nucleotide sequence for encoding the isolated peptide of (15).
ADVANTAGEOUS EFFECTS OF INVENTION
The patent provides a listeria personalized multi-target tumor vaccine method for simultaneously expressing a plurality of antigen peptides by non-integrated plasmids.
In one embodiment, the method is advantageous in that listeria harboring multiple tumor antigen targets can enhance an anti-tumor immune response.
In another embodiment, the method is further capable of simultaneously activating multiple tumor-specific T cells in the body, recognizing multiple targets for a tumor or recognizing different targets for multiple tumors, thereby inducing a multi-target immune response in the subject.
In another embodiment, the method can increase the efficacy of a tumor vaccine and drive the development of a personalized tumor vaccine against a neoantigen.
Drawings
FIG. 1 shows a plasmid map of a Listeria-expressed antigen gene;
FIG. 2 shows colony PCR detection of LM-LLO 267 -MA2 vaccine sequence of interestDetection results, wherein M shows a 500bp DNA band reference; line 1-3 shows Lm 10403 S.DELTA.acta (pAM 401-LLO) respectively 267 -MA 2-His) PCR product;
FIG. 3 shows Western blot verification of LM-LLO 267 -protein expression of MA2 vaccine;
FIG. 4 shows Elispot validated Normal mice versus LM-LLO 267 -activation of MA2 vaccine specific immune response;
FIG. 5 shows a tumor growth curve;
figure 6 shows the results of the Elispot response 7 days after cell therapy.
Detailed Description
Definition of the definition
The terms "a" or "an" when used in conjunction with the term "comprising" in the claims and/or specification may refer to "one" but may also refer to "one or more", "at least one" and "one or more".
As used in the claims and specification, the words "comprise," "have," "include" or "contain" mean including or open-ended, and do not exclude additional, unrecited elements or method steps. In the meantime, "comprising," "having," "including," or "containing" may also mean enclosed, excluding additional, unrecited elements or method steps.
Throughout this application, the term "about" means: one value includes the standard deviation of the error of the device or method used to determine the value.
Although the disclosure supports the definition of the term "or" as being inclusive of alternatives and "and/or", the term "or" in the claims means "and/or" unless expressly indicated otherwise as being exclusive of each other, as defined by the alternatives or alternatives.
When used in the claims or specification, the term "numerical range" is intended to include both the numerical endpoints of the range and all natural numbers covered in the middle of the numerical endpoints relative to the numerical endpoints.
The terms "inhibit," "reduce," or "prevent," or any variation of these terms, when used in the claims and/or specification, include any measurable reduction or complete inhibition to achieve a desired result (e.g., cancer treatment). Desirable outcomes include, but are not limited to, alleviation, diminishment, slowing or eradication of cancer or a proliferative disorder or cancer-associated symptoms, and improved quality of life or life prolongation.
The vaccination methods in the present disclosure are useful for treating cancer in a mammal. The term "cancer" as used in this disclosure includes any cancer, including but not limited to melanoma, sarcoma, lymphoma, carcinoma (e.g., brain cancer, breast cancer, liver cancer, stomach cancer, lung cancer, and colon cancer), and leukemia.
The term "mammal" in this disclosure refers to both human and non-human mammals.
The methods of the present disclosure comprise administering to a mammal a vaccine that expresses a tumor antigen to which the mammal has a pre-existing immunity. The term "pre-existing immunity" as used in the present disclosure is meant to include immunity induced by vaccination with an antigen as well as naturally occurring immunity in mammals.
The term "OVA" in the present disclosure refers to chicken Ovalbumin (ovabumin), also known as chicken Ovalbumin, consisting of 386 amino acids with a molecular weight of about 45kD.
The term "Phyy" in the present disclosure is a promoter of LLO (lysin) gene.
The term "vaccine" in the present disclosure is an immune preparation for preventing diseases, which is prepared by artificially attenuating, inactivating or using transgenes and the like, of pathogenic microorganisms (such as bacteria and the like) and metabolites thereof.
The term "radiotherapeutic agent" in the present disclosure includes the use of drugs that cause DNA damage. Radiation therapy has been widely used in cancer and disease treatment and includes those commonly referred to as gamma rays, X-rays and/or targeted delivery of radioisotopes to tumor cells.
The term "chemotherapeutic agent" in this disclosure is a chemical compound useful in the treatment of cancer. Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, photosensitizers, anti-estrogens and selective estrogen receptor modulators, anti-progesterone, estrogen receptor down-regulators, estrogen receptor antagonists, luteinizing hormone releasing hormone agonists, anti-androgens, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, antisense oligonucleotides inhibiting expression of genes involved in abnormal cell proliferation or tumor growth. Chemotherapeutic agents useful in the methods of treatment of the present disclosure include cytostatic and/or cytotoxic agents.
The term "immunotherapeutic" in this disclosure includes "immunomodulators" and agents that promote or mediate antigen presentation that promote a cell-mediated immune response. Wherein an "immune modulator" comprises an immune checkpoint modulator, such as an immune checkpoint protein receptor and its ligand-mediated inhibition of T cell-mediated cytotoxicity, and is typically expressed on a tumor or non-responsive T cells in the tumor microenvironment, and allows the tumor to evade immune attack. Inhibitors of the activity of immunosuppressive checkpoint protein receptors and their ligands can overcome the immunosuppressive tumor environment to allow cytotoxic T cell attack of the tumor. Examples of immune checkpoint proteins include, but are not limited to, PD-1, PD-L1, PDL2, CTLA4, LAG3, TIM3, TIGIT and CD103. Modulation (including inhibition) of the activity of such proteins may be accomplished by immune checkpoint modulators, which may include, for example, antibodies, aptamers, small molecules, and soluble forms of checkpoint receptor proteins that target checkpoint proteins, and the like. PD-1 targeted inhibitors include approved pharmaceutical agents pembrolizumab (pembrolizumab) and nivolumab (nivolumab), while iplimumab (ipilimumab) is an approved CTLA-4 inhibitor. Antibodies specific for PD-L1, PD-L2, LAG3, TIM3, TIGIT and CD103 are known and/or commercially available and may also be generated by one of skill in the art.
The term "substitution, repetition, deletion, or addition of one or more amino acids" in the present disclosure includes "conservative mutations". The term "conservative mutation" in the present disclosure refers to a conservative mutation that can normally maintain the function of a protein. Representative examples of conservative mutations are conservative substitutions. Conservative substitutions are, for example, mutations that, when the substitution site is an aromatic amino acid, result in a mutual substitution between Phe, trp, tyr; a mutation which, when the substitution site is a hydrophobic amino acid, causes mutual substitution between Leu, ile, val; in the case of polar amino acids, a mutation by which Gln and Asn are substituted for each other; in the case of basic amino acids, a mutation by which the amino acids are substituted for each other between Lys, arg, his; in the case of acidic amino acids, a mutation by which Asp and Glu are substituted for each other; in the case of an amino acid having a hydroxyl group, a mutation is substituted between Ser and Thr. Specific examples of the substitution to be regarded as a conservative substitution include substitution of Ala to Ser or Thr, substitution of Arg to Gln, his or Lys, substitution of Asn to Glu, gln, lys, his or Asp, substitution of Asp to Asn, glu or Gln, substitution of Cys to Ser or Ala, substitution of Gln to Asn, glu, lys, his, asp or Arg, substitution of Glu to Gly, asn, gln, lys or Asp, substitution of Gly to Pro, substitution of His to Asn, lys, gln, arg or Tyr, substitution of Ile to Leu, met, val or Phe, substitution of Leu to Ile, met, val or Phe, substitution of Lys to Asn, glu, gln, his or Arg, substitution of Met to Ile, leu, val or Phe, substitution of Phe to Trp, tyr, met, ile or Leu, substitution of Ser to Thr or Ala, substitution of Thr to Ser or Ala, substitution of Trp to Phe or Tyr, substitution of Tyr to His, phe or Trp, and substitution of Val to Met, ile or Leu. In addition, conservative mutations include naturally occurring mutations resulting from individual differences, strains, species differences, and the like from which the gene is derived.
"sequence identity" and "percent identity" in the present disclosure refer to the percentage of nucleotides or amino acids that are identical (i.e., identical) between two or more polynucleotides or polypeptides. Sequence identity between two or more polynucleotides or polypeptides may be determined by: the nucleotide or amino acid sequences of the polynucleotides or polypeptides are aligned and the number of positions in the aligned polynucleotides or polypeptides that contain the same nucleotide or amino acid residue is scored and compared to the number of positions in the aligned polynucleotides or polypeptides that contain a different nucleotide or amino acid residue. Polynucleotides may differ at one position, for example, by containing different nucleotides (i.e., substitutions or mutations) or by deleting nucleotides (i.e., nucleotide insertions or nucleotide deletions in one or both polynucleotides). The polypeptides may differ at one position, for example, by containing different amino acids (i.e., substitutions or mutations) or by deleting amino acids (i.e., amino acid insertions or amino acid deletions in one or both polypeptides). Sequence identity can be calculated by dividing the number of positions containing the same nucleotide or amino acid residue by the total number of amino acid residues in the polynucleotide or polypeptide. For example, percent identity can be calculated by dividing the number of positions containing the same nucleotide or amino acid residue by the total number of nucleotide or amino acid residues in the polynucleotide or polypeptide and multiplying by 100.
In one aspect of the disclosure, the MA2 antigen peptide sequence and the sequence as set forth in SEQ ID NO:2, having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% sequence identity (including all ranges and percentages between these values). In the present disclosure, a MA2 antigenic peptide sequence having a specific percentage of identity refers to a sequence encoding the MA2 antigenic peptide when the MA2 antigenic peptide is still present for its antigenic activity.
The "routine biological methods in the art" in the present disclosure can be referred to the corresponding methods described in the publications such as "the latest molecular biology laboratory method Assembly (Current Protocols in Molecular Biology, wiley publication)", "the molecular cloning laboratory Manual (Molecular Cloning: A Laboratory Manual, cold spring harbor laboratory publication)", and the like.
In the present disclosure, the numbers of nucleotides or amino acids indicated by different sequence numbers have the following meanings:
SEQ ID NO:1 is the nucleotide sequence of the MA2 antigen peptide;
SEQ ID NO:2 is the amino acid sequence of the MA2 antigen peptide;
SEQ ID NO:3 is the nucleotide sequence of LLO267-MA 2;
SEQ ID NO:4 is the amino acid sequence of LLO267-MA 2.
Examples
Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.
Unless specifically indicated, all reagents and starting materials employed in the disclosure are commercially available.
Example 1: attenuated listeria carrying non-integrated G4S sequence as connecting sequence for connecting multiple antigen peptide plasmids Construction of bacterial strains and vectors
The strain used as vaccine in this study was Lm 10403S Δacta. The preparation methods of the aforementioned strains can be exemplified by the following documents: shen H et al, PNAS,92 (9): 3987-91, 1995. The bacterium lacks the actA gene, so that the bacterium infecting host cells cannot spread to adjacent cells through the special actin tail, thereby greatly weakening the toxicity and pathogenicity. Compared with the wild strain Lm 10403S (LD) 50 1x10 4 ) LD of Lm- Δacta 50 0.5-1x10 8 Proved to be highly attenuated.
The plasmids used for expression of the antigen genes in this study were essentially composed of: (as shown in FIG. 1)
(1) Basic sequence to maintain plasmid replication stability: pAM401;
(2) Promoters for transcription of antigen genes: phly, the promoter of the virulence island LLO on Lm chromosomes;
(3) Expression of signal peptide sequences secreting antigenic proteins outside listeria: LLO signal peptide (LLO 1-28 aa) and LLO half-cut gene for increasing the expression level of exogenous proteins;
(4) Listeria belongs to prokaryotic cells, while antigenic peptides which are usually required to be used as tumor vaccines belong to eukaryotic cells, and eukaryotic cell proteins can be expressed in the prokaryotic cells by performing corresponding codon optimization;
(5) Detecting the tag sequence of the secreted protein: flag tag or His tag;
cleavage site for antigen peptide insertion: pstI.
Example 2: construction of antigenic peptide plasmids
The construction of listeria vaccine plasmid requires that antigen genes are inserted into a plasmid vector, enzyme cutting sites are designed on the vector, and the gene sequence of target antigen is synthesized after gene codon optimization by a company. The composition for connecting 2 antigen peptides of mice by using G4S as linker is specifically shown below, and comprises Ovalbumin 257-264 and Tyrosinase related protein-2 180-188 antigen peptides, which are abbreviated as MA2.
We cloned the product into pAM401-phly-LLO using homologous recombination techniques based on certain homologous sequences 1-28 -LLO 22-267 -PstI-LLO 524-529 The PstI site on the His vector (abbreviated as PstI vector plasmid) to obtain pAM401-phly-LLO 1-28 -LLO 22-267 -PstI-MA2-PstI-LLO 524-529 His, abbreviated pAM401-LLO 267 -MA2 vector plasmid and preparing LM-LLO267-MA2 vaccine.
The homologous sequences are: 5' terminal homologous sequence (CCGAAATATAGTAATAAACTGCAG); 3' homologous sequence (CTGCAGGTAGATAATCCAATCGAA)
The method mainly comprises the following steps:
PstI vector plasmid 20. Mu.L PstI Single cleavage System:
PstI plasmid 2ug
PstI restriction endonuclease (NEB) 2μL
10x NEBuffer 3.1 2μL
Deionized water Supplement to 20 mu L
And (3) a water bath kettle at 37 ℃ for reaction for 30min.
DNA recovery and purification of 20. Mu.L of the digested product, namely, digestion linearization PstI vector
Homologous recombination 20 μl system:
Figure BDA0002496521630000101
ddH 2 o: supplement to 20 mu L
The mixture was immediately placed on ice for 5 minutes in a water bath at 37℃and 100. Mu.L of E.coli competent was all transformed, and the resistance plate was coated and the monoclonal was selected for sequencing verification.
Example 3: preparation of attenuated listeria harboring non-integral antigenic peptide plasmids
The correct plasmid is converted into attenuated listeria strain by electrotransformation technology, and monoclonal is selected for subsequent plasmid and expression verification.
The specific steps of the electric conversion are as follows:
(1) Preparation of electrotransformation competence
a) Listeria cultured overnight was transferred to BHI medium and shake cultured at 37℃to OD 600 A value of 0.2 to 0.25;
b) PNG was added and culture continued for about two hours to OD 600 To 0.3-0.9;
c) After 10 minutes of ice bath, 5000g of the bacteria are collected centrifugally;
d) Re-suspending the cells with 200ml glycerol and washing twice;
e) The cells were resuspended in 45ml glycerol and added with sterile lysozyme solution, water bath at 37℃for 20 min;
f) Centrifuging 5000g to collect thalli at 4 ℃, and washing the thalli once with 20ml of glycerol;
g) The cells were resuspended in 1ml glycerol and stored in 50. Mu.L/tube aliquots.
(2) Determining the optimal electrotransformation conditions:
a) Taking a tube of competent cells, thawing with the heart, and placing on ice;
b) The plasmid to be transferred was added to competent cells, mixed well and ice-bathed for 5 min.
c) Adding the mixed system into a precooled 1mm electric rotating cup, and performing electric shock treatment;
d) Immediately adding the BHI culture medium, uniformly mixing, and taking out to an EP tube;
e) The bacterial cells were coated with BHI+ resistant plates, cultured upside down at 37℃and single colonies were picked for verification.
Example 4: improvement and detection of expression of exogenous protein of attenuated listeria harboring non-integral antigenic peptide plasmid
Listeria overnight was cultured in BHI broth at 37℃and centrifuged to remove the cells, and the supernatant was mixed with 3 volumes of 10% TCA (trichloroacetic acid)/acetone solution and precipitated overnight at-20 ℃. The precipitated protein was collected by centrifugation at 15000rpm for 30 minutes, washed twice with ice-chilled acetone, and the residual TCA was removed. Excess acetone was evaporated in the fume hood and the pellet was dissolved with protein loading buffer containing 0.01N NaOH. And (3) boiling the denatured protein, loading the denatured protein, and carrying out Western blot to detect the protein expression quantity through the flag tag or his tag received by the protein.
Collecting the strain with high expression, adding into DMSO frozen stock solution, preparing into LM-LLO267-MA2 vaccine, and preserving at-80deg.C.
Example 5: elispot to verify activation of specific immune response in normal mice to MA2 vaccine
Three normal C57 mice were inoculated with LM-LLO267-MA2 vaccine by tail vein, the mice were taken from all peripheral blood, red blood cells were lysed, PBS was washed twice to obtain peripheral blood mononuclear cells, and 1ml 1640 was used to complete the immunizationThe culture medium is resuspended for use. Taking lymph nodes and spleens of the cells, grinding a filter screen, staining the collected cells with CD8-PE dye, washing with PBS twice, incubating the collected cells and anti-PE magnetic beads on ice together, centrifuging, collecting, adsorbing the cells combined with the anti-PE magnetic beads on the magnetic column, removing the magnetic column from the magnetic frame, and collecting target cells, namely CD8+T cells. Lymph node T cells, spleen T cells were resuspended in 1640 complete medium, and 100 μl per well was added to Elispot pretreatment well plates, followed by the addition of 100 μl per well of mouse peripheral blood mononuclear cells. Then, the Tyrosinase related protein-2-180-188 polypeptide and the Ovalbumin 257-264 polypeptide are respectively added into an Elispot pore plate to stimulate T cells to generate INF-gamma, and finally, the spot number is quantified through an enzyme-linked reaction to indicate the specific immunoreaction condition of each group of response Tyrosinase related protein-2-180-188 polypeptide and OVA polypeptide (the specific Elispot experimental flow refers to BD TM ELISPOT Mouse IFN-gamma ELISPOT Set Specification, product No. 551083).
Example 6: molecular characterization of MA2 vaccine
LLO to be constructed 267 The vector plasmid connected with MA2 is electrically transformed into LM competence, bacterial cells are coated on BHI chloramphenicol plates, single colonies are grown after the incubator is inverted and cultured at 37 ℃, and colony PCR verification is carried out on each group of single colonies.
The experimental results are shown in fig. 2, and the experimental group appears to be the target product which obviously accords with the actual size. Indicating that the plasmid has been electrotransformed into LM competence and can be used for vaccine preparation.
267 Example 7: LM-LLO-MA2 vaccine expression verification
For accurate detection of LM-LLO 267 Protein expression of MA2 vaccine, we verified by Western blot experiments by culturing different single colonies to precipitate the supernatant protein. The specific process comprises the following steps: colonies No. 1 and No. 2 of MA2 were picked up and added to a chloramphenicol-resistant BHI broth, followed by shaking culture for 14-16 hours, and centrifugation at 4500rpm to pellet the cells. 10ml of the supernatant was mixed with TCA/acetone solution and precipitated overnight. Centrifuging at 15000rpm to collect precipitate protein, pre-cooling with acetone, washing twice, and removing residueLeaving TCA. Volatilizing excessive acetone in a fume hood, dissolving precipitate with protein loading buffer solution, boiling, denaturing and preserving.
Preparing 10% of separating gel and 4% of concentrating gel respectively, loading 20 mu L of electrophoresis sample at each hole, and changing the joint of the electrophoresis sample and the separating gel of the concentrating gel into 120v. After electrophoresis was completed, the separation gel was taken, filter paper and 0.22um PVDF membrane (activated in methanol in advance) were cut to the same size as the gel, and a transfer ice bath was performed. 5% skim milk TBST was blocked and TBST was washed 3 times. Incubation with HRP-labeled Anti-His antibody at room temperature, TBST washing 3 times. ECL developer is dripped on the PVDF film, and the film is developed by a Bio-Rad gel imager.
The experimental results are shown in FIG. 3, LM-LLO 267 The MA2 vaccine has significant protein expression.
267 Example 8: verifying that a plurality of antigen peptides connected by using G4S sequence as connecting sequence in LM-LLO-MA2 are connected in vivo Specific antigen presentation, activation of specific T cells
We took three normal mice for intravenous LM-LLO at the tail 267 After 7 days of immunization, taking mouse peripheral blood, lymph node T cells and spleen T cells for co-culture in an Elispot pore plate, taking the Ovalbumin 257-264 and Tyrosinase related protein-2-180-188 polypeptides as stimulators, and if a plurality of antigen peptides connected by taking a G4S sequence as a connecting sequence can be specifically presented, specific CD8+ T cells can be activated to secrete INF-gamma interferon so as to display spots in the Elispot, and taking co-cultured cells which are not stimulated by any polypeptides as negative controls. The specific process comprises the following steps: taking three normal C57 mice with LM-LLO inoculated at tail vein 267 -MA2 vaccine, day 7 of immunization, taking all peripheral blood of the mice, lysing erythrocytes, washing twice with PBS to obtain peripheral blood mononuclear cells, and re-suspending with 1640 complete medium for later use. Taking lymph nodes and spleens of the cells, grinding a filter screen, staining the collected cells with CD8-PE dye, washing with PBS twice, incubating the collected cells and anti-PE magnetic beads on ice together, centrifuging, collecting, adsorbing the cells combined with the anti-PE magnetic beads on the magnetic column, removing the magnetic column from the magnetic frame, and collecting target cells, namely CD8+T cells. Will be sprayed withThe barely T cells, splenic T cells were resuspended in 1640 complete medium and 100. Mu.l per well was inoculated into Elispot pretreatment well plates, followed by the addition of 100. Mu.l per well of mouse peripheral blood mononuclear cells. Then, the Tyrosinase related protein-2-180-188 polypeptide and the Ovalbumin 257-264 polypeptide are respectively added into an Elispot pore plate to stimulate T cells to generate INF-gamma, and finally, the spot number is quantified through an enzyme-linked reaction to indicate the specific immunoreaction condition of each group responding to OVA polypeptide (the specific Elispot experimental flow refers to BD TM ELISPOT Mouse IFN-gamma ELISPOT Set Specification, product No. 551083).
As shown in FIG. 4, the results of experiments show that the Elispot with the Ovalbumin 257-264 and Tyrosinase related protein-2-180-188 polypeptides as the stimulus has obvious speckle reaction, which proves that the LM-LLO 267 A plurality of antigen peptides connected by taking a G4S sequence as a connecting sequence in MA2 are presented by specific antigens in vivo, so that specific CD8+ T cells are activated to secrete INF-gamma interferon, and direct evidence is provided for in vivo anti-tumor multi-target personalized immune response.
267 Example 9: demonstration that multiple antigen peptides connected by using G4S sequence as connecting sequence in LM-LLO-MA2 activate multiple antigen peptides Effectiveness of specific CD8+ T cells in killing tumor cells in vivo
Taking 15C 57 mice, subcutaneously inoculating B16-OVA to generate tumor, injecting LM-LLO 267 MA2 vaccine verifies the anti-tumor effect. B16-OVA cell inoculum size was 2X 10 6 Tumor measurements were started on day 4 post inoculation. The 15 mice were uniformly divided into three groups according to tumor size as follows:
Figure BDA0002496521630000131
/>
Figure BDA0002496521630000141
PBS control and LLO on day 9 267 The control group and MA2 experimental group are injected with vaccine, and the tumor size is continuously observed. Tumor persistence trackingThe measurement was carried out for 23 days.
Tumor curves are shown in FIG. 5, PBS control group and LLO 267 The tumor growth of the mice in the control group is continuously increased along with the time, and the MA2 experimental group has relatively slow tumor growth from 12 days, and has obvious tumor inhibition effect.
267 Example 10: detection of LM-LLO-MA2 vs. B16-OVA tumor model mice in vivo from functionality Using Elispot Activation of tumor-specific immune response
We obtained peripheral blood mononuclear cells by 3-7 drops from tail vein, lysing erythrocytes, and PBS washing twice on day 7 after cell therapy injection, and finally resuspended cells using 100 μl of 1640 complete medium, respectively, into Elispot pretreatment well plates. Peripheral blood mononuclear cells were then stimulated to produce INF-gamma by addition of OVA257-264, TRP-2 180-188 polypeptides in Elispot plates, and the spot numbers were finally quantified by ELISA to indicate the specific immune response of each group in response to OVA polypeptides (see BD for specific Elispot protocols TM ELISPOT Mouse IFN-gamma ELISPOT Set Specification, product No. 551083).
The experimental results are shown in FIG. 6, PBS control group and LLO 267 The control group has no or a small amount of Elispot spot reaction, and the MA2 experimental group stimulated by OVA257-264 polypeptide and TRP-2 180-188 polypeptide has obvious Elispot reaction, which indicates that the LM-LLO 267 The MA2 takes the G4S sequence as the connecting sequence to connect a plurality of antigen peptides which can be presented by specific antigens in vivo, thereby recognizing a plurality of targets of tumor and activating a plurality of specific CD8+T cells of mice to kill tumor cells, and improving the curative effect of tumor vaccine.
The above examples of the present disclosure are merely examples for clearly illustrating the present disclosure and are not limiting of the embodiments of the present disclosure. Other variations or modifications of the above teachings will be apparent to those of ordinary skill in the art. It is not necessary here nor is it exhaustive of all embodiments. Any modifications, equivalent substitutions, improvements, etc. that fall within the spirit and principles of the present disclosure are intended to be included within the scope of the claims of the present disclosure.
Sequence listing
<110> Suzhou Ruotai pharmaceutical technology Co., ltd
Shanghai Ruotai Pharmaceutical Technology Co.,Ltd.
<120> preparation and use of an attenuated listeria-based tumor vaccine
<130> 6709-191365I-SH
<160> 4
<170> SIPOSequenceListing 1.0
<210> 1
<211> 531
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 1
ggtggtggcg gttcaggtgg tggtggcagc agcattatca actttgaaaa actgggcggc 60
ggcggttcag gtggtggtgg ctcaaaagtt gtgcgctttg ataaactggg cggtggtggt 120
agcggtggtg gtggtagcag cgtgtatgat ttttttgtgt ggctgggtgg tggtggttca 180
ggtggtggtg gcagccatta tttaagcacc cagagcgcgt taggcggtgg tggtagcggt 240
ggtggtggtt caactcctca tcctgcgcgt attggtttag gtggtggtgg tagcggtggt 300
ggtggtagcc ctagcaaacc gagctttcag gaatttgtgg attgggaaaa cgtgagcccg 360
gaactgaact caaccgatca gccgttttta ggcggtggtg gctcaggtgg tggtggtagc 420
ttacatagcg gccagaacca tctgaaagaa atggcgatta gcgtgttaga agcgcgtgca 480
tgtgcagcag caggtcaatc aggtggtggt ggttcaggtg gtggtggtag c 531
<210> 2
<211> 177
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 2
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Ser Ile Ile Asn Phe Glu
1 5 10 15
Lys Leu Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Lys Val Val Arg
20 25 30
Phe Asp Lys Leu Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Ser Val
35 40 45
Tyr Asp Phe Phe Val Trp Leu Gly Gly Gly Gly Ser Gly Gly Gly Gly
50 55 60
Ser His Tyr Leu Ser Thr Gln Ser Ala Leu Gly Gly Gly Gly Ser Gly
65 70 75 80
Gly Gly Gly Ser Thr Pro His Pro Ala Arg Ile Gly Leu Gly Gly Gly
85 90 95
Gly Ser Gly Gly Gly Gly Ser Pro Ser Lys Pro Ser Phe Gln Glu Phe
100 105 110
Val Asp Trp Glu Asn Val Ser Pro Glu Leu Asn Ser Thr Asp Gln Pro
115 120 125
Phe Leu Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Leu His Ser Gly
130 135 140
Gln Asn His Leu Lys Glu Met Ala Ile Ser Val Leu Glu Ala Arg Ala
145 150 155 160
Cys Ala Ala Ala Gly Gln Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly
165 170 175
Ser
<210> 3
<211> 1425
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 3
atgaaaaaaa taatgctagt ttttattaca cttatattag ttagtctacc aattgcgcaa 60
caaactgaag caaaggatgc atcggatcct actgaagcaa aggatgcatc tgcattcaat 120
aaagaaaatt caatttcatc catggcacca ccagcatctc cgcctgcaag tcctaagacg 180
ccaatcgaaa agaaacacgc ggatgaaatc gataagtata tacaaggatt ggattacaat 240
aaaaacaatg tattagtata ccacggagat gcagtgacaa atgtgccgcc aagaaaaggt 300
tacaaagatg gaaatgaata tattgttgtg gagaaaaaga agaaatccat caatcaaaat 360
aatgcagaca ttcaagttgt gaatgcaatt tcgagcctaa cctatccagg tgctctcgta 420
aaagcgaatt cggaattagt agaaaatcaa ccagatgttc tccctgtaaa acgtgattca 480
ttaacactca gcattgattt gccaggtatg actaatcaag acaataaaat cgttgtaaaa 540
aatgccacta aatcaaacgt taacaacgca gtaaatacat tagtggaaag atggaatgaa 600
aaatatgctc aagcttatcc aaatgtaagt gcaaaaattg attatgatga cgaaatggct 660
tacagtgaat cacaattaat tgcgaaattt ggtacagcat ttaaagctgt aaataatagc 720
ttgaatgtaa acttcggcgc aatcagtgaa gggaaaatgc aagaagaagt cattagtttt 780
aaacaaattt actataacgt gaatgttaat gaacctacaa gaccttcccc gaaatatagt 840
aataaactgc agggtggtgg cggttcaggt ggtggtggca gcagcattat caactttgaa 900
aaactgggcg gcggcggttc aggtggtggt ggctcaaaag ttgtgcgctt tgataaactg 960
ggcggtggtg gtagcggtgg tggtggtagc agcgtgtatg atttttttgt gtggctgggt 1020
ggtggtggtt caggtggtgg tggcagccat tatttaagca cccagagcgc gttaggcggt 1080
ggtggtagcg gtggtggtgg ttcaactcct catcctgcgc gtattggttt aggtggtggt 1140
ggtagcggtg gtggtggtag ccctagcaaa ccgagctttc aggaatttgt ggattgggaa 1200
aacgtgagcc cggaactgaa ctcaaccgat cagccgtttt taggcggtgg tggctcaggt 1260
ggtggtggta gcttacatag cggccagaac catctgaaag aaatggcgat tagcgtgtta 1320
gaagcgcgtg catgtgcagc agcaggtcaa tcaggtggtg gtggttcagg tggtggtggt 1380
agcctgcagg tagataatcc aatcgaacat catcatcatc atcat 1425
<210> 4
<211> 475
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 4
Met Lys Lys Ile Met Leu Val Phe Ile Thr Leu Ile Leu Val Ser Leu
1 5 10 15
Pro Ile Ala Gln Gln Thr Glu Ala Lys Asp Ala Ser Asp Pro Thr Glu
20 25 30
Ala Lys Asp Ala Ser Ala Phe Asn Lys Glu Asn Ser Ile Ser Ser Met
35 40 45
Ala Pro Pro Ala Ser Pro Pro Ala Ser Pro Lys Thr Pro Ile Glu Lys
50 55 60
Lys His Ala Asp Glu Ile Asp Lys Tyr Ile Gln Gly Leu Asp Tyr Asn
65 70 75 80
Lys Asn Asn Val Leu Val Tyr His Gly Asp Ala Val Thr Asn Val Pro
85 90 95
Pro Arg Lys Gly Tyr Lys Asp Gly Asn Glu Tyr Ile Val Val Glu Lys
100 105 110
Lys Lys Lys Ser Ile Asn Gln Asn Asn Ala Asp Ile Gln Val Val Asn
115 120 125
Ala Ile Ser Ser Leu Thr Tyr Pro Gly Ala Leu Val Lys Ala Asn Ser
130 135 140
Glu Leu Val Glu Asn Gln Pro Asp Val Leu Pro Val Lys Arg Asp Ser
145 150 155 160
Leu Thr Leu Ser Ile Asp Leu Pro Gly Met Thr Asn Gln Asp Asn Lys
165 170 175
Ile Val Val Lys Asn Ala Thr Lys Ser Asn Val Asn Asn Ala Val Asn
180 185 190
Thr Leu Val Glu Arg Trp Asn Glu Lys Tyr Ala Gln Ala Tyr Pro Asn
195 200 205
Val Ser Ala Lys Ile Asp Tyr Asp Asp Glu Met Ala Tyr Ser Glu Ser
210 215 220
Gln Leu Ile Ala Lys Phe Gly Thr Ala Phe Lys Ala Val Asn Asn Ser
225 230 235 240
Leu Asn Val Asn Phe Gly Ala Ile Ser Glu Gly Lys Met Gln Glu Glu
245 250 255
Val Ile Ser Phe Lys Gln Ile Tyr Tyr Asn Val Asn Val Asn Glu Pro
260 265 270
Thr Arg Pro Ser Pro Lys Tyr Ser Asn Lys Leu Gln Gly Gly Gly Gly
275 280 285
Ser Gly Gly Gly Gly Ser Ser Ile Ile Asn Phe Glu Lys Leu Gly Gly
290 295 300
Gly Gly Ser Gly Gly Gly Gly Ser Lys Val Val Arg Phe Asp Lys Leu
305 310 315 320
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Ser Val Tyr Asp Phe Phe
325 330 335
Val Trp Leu Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser His Tyr Leu
340 345 350
Ser Thr Gln Ser Ala Leu Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
355 360 365
Thr Pro His Pro Ala Arg Ile Gly Leu Gly Gly Gly Gly Ser Gly Gly
370 375 380
Gly Gly Ser Pro Ser Lys Pro Ser Phe Gln Glu Phe Val Asp Trp Glu
385 390 395 400
Asn Val Ser Pro Glu Leu Asn Ser Thr Asp Gln Pro Phe Leu Gly Gly
405 410 415
Gly Gly Ser Gly Gly Gly Gly Ser Leu His Ser Gly Gln Asn His Leu
420 425 430
Lys Glu Met Ala Ile Ser Val Leu Glu Ala Arg Ala Cys Ala Ala Ala
435 440 445
Gly Gln Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Leu Gln Val
450 455 460
Asp Asn Pro Ile Glu His His His His His His
465 470 475

Claims (17)

1. A recombinant nucleic acid molecule comprising an open reading frame encoding a recombinant polypeptide comprising a heterologous antigen fused to a derivatized listeriolysin (LLO) polypeptide, wherein the amino acid sequence of the heterologous antigen is a polypeptide having the activity of a heterologous antigen of the amino acid sequence shown in SEQ ID No. 2.
2. The recombinant nucleic acid molecule of claim 1, wherein the nucleotide sequence encoding the heterologous antigen is as set forth in SEQ ID NO:1, and a sequence shown in 1.
3. The recombinant nucleic acid molecule of claim 1 or 2, wherein the sequence of the recombinant nucleic acid molecule is as set forth in SEQ ID NO: 3.
4. A recombinant plasmid or recombinant expression vector comprising the sequence of the recombinant nucleic acid molecule of any one of claims 1-3.
5. A recombinant protein encoded by the recombinant nucleic acid molecule of any one of claims 1-3, or formed from the recombinant plasmid or recombinant expression vector of claim 4.
6. A recombinant listeria comprising the recombinant nucleic acid molecule of any one of claims 1-3, or comprising the recombinant plasmid or recombinant expression vector of claim 4, or expressing the recombinant protein of claim 5.
7. A pharmaceutical composition comprising a therapeutically effective amount of the recombinant listeria of claim 6.
8. A prophylactic or therapeutic vaccine, characterized in that it comprises a prophylactically or therapeutically effective amount of the recombinant listeria of claim 6.
9. Use of the recombinant listeria of claim 6, the pharmaceutical composition of claim 7 or the vaccine of claim 8 in the manufacture of a medicament for killing cells.
10. The use according to claim 9, wherein the cells are selected from proliferative, neoplastic, precancerous or metastatic cells.
11. The use according to claim 10, wherein the cells are selected from metastatic cells.
12. The use according to claim 11, wherein the metastatic cells are selected from metastatic tumor cells.
13. Use of the recombinant listeria of claim 6, the pharmaceutical composition of claim 7 or the vaccine of claim 8 in the manufacture of a medicament for treating a patient with a tumor.
14. A method of sustaining killing a cell comprising contacting the cell with the recombinant listeria of claim 6, the pharmaceutical composition of claim 7, or the vaccine of claim 8; the method is not aimed at diagnosis and treatment of diseases.
15. The method of claim 14, wherein the cell is a tumor cell.
16. An isolated peptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 2 or SEQ ID NO: 4.
17. A nucleic acid molecule encoding the isolated peptide of claim 16.
CN202010420039.4A 2020-05-18 2020-05-18 Preparation and application of tumor vaccine based on attenuated listeria Active CN113684214B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010420039.4A CN113684214B (en) 2020-05-18 2020-05-18 Preparation and application of tumor vaccine based on attenuated listeria

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010420039.4A CN113684214B (en) 2020-05-18 2020-05-18 Preparation and application of tumor vaccine based on attenuated listeria

Publications (2)

Publication Number Publication Date
CN113684214A CN113684214A (en) 2021-11-23
CN113684214B true CN113684214B (en) 2023-05-05

Family

ID=78575471

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010420039.4A Active CN113684214B (en) 2020-05-18 2020-05-18 Preparation and application of tumor vaccine based on attenuated listeria

Country Status (1)

Country Link
CN (1) CN113684214B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110408634A (en) * 2018-04-27 2019-11-05 苏州若泰医药科技有限公司 A kind of nonconformity Listeria vaccine and anti-tumor immune response method
CN110579608A (en) * 2018-06-11 2019-12-17 苏州若泰医药科技有限公司 Method for screening non-integrated attenuated Listeria strain with high-expression foreign protein

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110408634A (en) * 2018-04-27 2019-11-05 苏州若泰医药科技有限公司 A kind of nonconformity Listeria vaccine and anti-tumor immune response method
CN110579608A (en) * 2018-06-11 2019-12-17 苏州若泰医药科技有限公司 Method for screening non-integrated attenuated Listeria strain with high-expression foreign protein

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Zhisong Chen 等.Episomal expression of truncated listeriolysin O in LmddA-LLO-E7 vaccine enhances antitumor efficacy by preferentially inducing expansions of CD4+FoxP3−and CD8+ T cells.《Cancer Immunol Res》.2014,第2卷(第9期), *
重组单增李斯特菌溶血素O在HEK293T细胞中的分泌表达及活性分析;常玉梅;《河北农业大学学报 》;20151022(第5期);全文 *

Also Published As

Publication number Publication date
CN113684214A (en) 2021-11-23

Similar Documents

Publication Publication Date Title
CN107574138B (en) Escherichia coli anti-tumor targeted engineering strain and construction method and application thereof
CN111440244B (en) Metastatic cancer vaccine targeting VEGFR2
US11648306B2 (en) Non-integrative listeria-based vaccine and method for inducing antitumor immune response
CN113528547B (en) Novel coronavirus B.1.617.1 mutant strain DNA vaccine
CN112533939B (en) Tumor immunotherapy composition based on attenuated listeria activated antigen presenting cells, preparation method and application
CN116396974B (en) African swine fever virus antigen protein recombinant expression vector, recombinant plant lactobacillus, and preparation method and application thereof
CN113528545B (en) Nucleic acid sequence for coding novel coronavirus B.1.1.7 mutant strain antigen and application thereof
CN113528549B (en) DNA molecule for encoding novel coronavirus B.1.351 mutant strain antigen, DNA vaccine and application
US9925259B2 (en) Immunogenic polypeptide surface layer-expressing bifidobacterium
CN108484776A (en) A kind of fusion protein, preparation method and applications
CN113249408B (en) Construction and application of nucleic acid vaccine vector for targeting activation of humoral immunity and cellular immunity
CN112538452A (en) Cervical cancer therapeutic vaccine based on recombinant attenuated Listeria monocytogenes and preparation method thereof
CN114478715B (en) Novel coronavirus vaccine presentation system for attenuated salmonella secretion expression of S2 structural domain protein and application thereof
CN113684214B (en) Preparation and application of tumor vaccine based on attenuated listeria
CN116732078A (en) Method for preparing tumor-associated antigen NY-ESO-1 by taking pET28a as carrier and application
CN105420174A (en) Establishment of genetically engineered bacterium expressing recombined VEGF fusion protein
CN111349645B (en) Method for improving safety of non-integrated attenuated listeria vaccine
CN111334521B (en) Method for improving expression of non-integrated attenuated Listeria exogenous antigen protein
CN106085932A (en) A kind of structure of the genetic engineering bacterium of VEGF Yu GnRH fusion protein of recombinating
CN110579608B (en) Method for screening non-integrated attenuated Listeria strain with high-expression foreign protein
CN116948038A (en) Recombinant antigenic peptides and recombinant listeria for inducing tumor-specific immune responses
CN111118063A (en) FAP α and survivin-based DNA and application thereof in preparation of tumor vaccine
CN115605595A (en) Fusion body of survivin and GM-CSF, coding DNA, recombinant expression vector, anti-tumor vaccine and application thereof
CN114790248B (en) MUC1-PDL1-IgG1Fc tumor vaccine and preparation method and application thereof
CN116751266B (en) Henipav protective antigen and application thereof

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
GR01 Patent grant
GR01 Patent grant
PE01 Entry into force of the registration of the contract for pledge of patent right
PE01 Entry into force of the registration of the contract for pledge of patent right

Denomination of invention: Preparation and application of a tumor vaccine based on attenuated Listeria monocytogenes

Granted publication date: 20230505

Pledgee: China Construction Bank Suzhou Industrial Park sub branch

Pledgor: SUZHOU ROYALTECH MED Co.,Ltd.

Registration number: Y2024980022288