CN114409782B - anti-IL13Ra2 nano antibody and application thereof - Google Patents

anti-IL13Ra2 nano antibody and application thereof Download PDF

Info

Publication number
CN114409782B
CN114409782B CN202111681616.6A CN202111681616A CN114409782B CN 114409782 B CN114409782 B CN 114409782B CN 202111681616 A CN202111681616 A CN 202111681616A CN 114409782 B CN114409782 B CN 114409782B
Authority
CN
China
Prior art keywords
chimeric antigen
antigen receptor
seq
cells
il13ra2
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
CN202111681616.6A
Other languages
Chinese (zh)
Other versions
CN114409782A (en
Inventor
狄升蒙
冯冬歌
陈鑫
李嘉涛
范艳秋
田琳
余学军
刘芳
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.)
Huadao Shanghai Biopharma Co ltd
Original Assignee
Huadao Shanghai Biopharma 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 Huadao Shanghai Biopharma Co ltd filed Critical Huadao Shanghai Biopharma Co ltd
Priority to CN202310611114.9A priority Critical patent/CN116589585B/en
Priority to CN202111681616.6A priority patent/CN114409782B/en
Priority to CN202310610963.2A priority patent/CN116514982B/en
Priority to CN202310610970.2A priority patent/CN116813773A/en
Priority to CN202310611121.9A priority patent/CN116444671B/en
Publication of CN114409782A publication Critical patent/CN114409782A/en
Application granted granted Critical
Publication of CN114409782B publication Critical patent/CN114409782B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • 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
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • 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
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • 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
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention provides an anti-IL13Ra2 nano antibody and application thereof. The amino acid sequence of CDR3 of the nano antibody comprises a sequence shown as SEQ ID NO.8, SEQ ID NO.9 or SEQ ID NO. 10. The nano antibody of the invention can specifically bind to IL13Ra2 antigen, has better affinity, and can be used as an antigen binding domain to construct chimeric antigen receptor and CAR-T cell, thereby having obvious killing activity on IL13Ra2 positive tumor cells.

Description

anti-IL13Ra2 nano antibody and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to an anti-IL13Ra2 nano antibody and application thereof.
Background
Gliomas are the most common primary malignant tumors in the cranium, and are classified according to WHO, and can be classified into classes I-IV from low to high in malignancy, wherein I and II are low-grade gliomas, III and IV are high-grade gliomas, which areGlioblastoma multiforme (Glioblastoma multiforme, GBM) is the highest in malignancy, accounting for more than 80% of glioblastoma. Although diagnostic and therapeutic methods have been rapidly developed over the last decade, the prognosis is still not ideal due to the characteristics of high grade gliomas (grade iii-iv), strong aggressiveness, high difficulty in treatment, poor prognosis, high recurrence rate, etc., with a median survival of only about 14 months [1] Survival rate of 2 years is less than 25 percent [2,3] . Although low grade gliomas (grade i-ii) have a higher prognosis, they can still give patients neurological symptoms such as epilepsy, cognitive disorders, etc.
Traditional treatment schemes of gliomas are that surgical excision is combined with auxiliary temozolomide chemotherapy and synchronous radiotherapy, but treatment measures for malignant gliomas which are difficult to carry out surgical treatment at multiple and growing positions are limited. Such non-specific treatment regimens do not provide complete relief of glioblastoma and can cause damage to healthy brain tissue, which is not beneficial to improving patient quality of life. In addition, individual glioma cells can metastasize through white matter tracts, callus or cerebrospinal fluid, leading to recurrence, with more than half of patients finding metastases in the contralateral hemispheres [4] . In view of the poor effect of traditional therapies, the development of chimeric antigen receptor T-cells (CAR-T) has been fast in recent years, which brings new eosin to the treatment of GBM.
A number of in vitro pre-clinical results have demonstrated that the 5 tumor-associated antigens IL13Rα2 (Interleukin-13 receptor αchain variable 2, interleukin 13receptor α2), EGFR VIII (epidermal growth factor receptor-VIII, EGFR variant VIII), erbB2 (epidermal growth factor receptor, EGFR 2), ephA2 (ephrinA 2 receptor, tyrosine protein kinase receptor A2) and B7-H3 are likely targets for the treatment of GBM, have achieved good results in preclinical animal models and in part of the clinical trials, and have been developed in clinical trials.
Interleukin 13receptor alpha 2 (IL 13Rα2), a membrane-bound protein, is closely related to IL13Rα1. It has been demonstrated that adult, pediatric brain tumors and meningiomas IL13Rα2 are abundantly expressed, in contrast, they are barely detectable in normal human brain tissueThe expression quantity on the surface of GBM cells is 3 ten thousand times that of normal tissue cells, and 58% of the gene expression of patients is up-regulated [5] Flow cytometry analysis of the average expression levels in U373 cell lines and 3 GBM patient cells was as high as 73% [6] Is an effective target for immunotherapy GBM. It plays an important role in immune response and tumor microenvironment, and can block apoptosis pathways, resulting in immune escape. The first generation and second generation CAR-T targeting IL13 Ralpha 2 show obvious anti-tumor effect [7-9] . First generation CAR-T cells expressing IL13E13Y muteins have higher affinity for IL13Rα2 and compete for binding to IL-13a1/IL4Ra expressed in the central nervous system, as reported [10] 3 cases of recurrent GBM were injected directly into CD8 (+) CTL cells of IL13 (E13Y) -zetakine multiple times through the cranial cavity after surgery, 2 cases had a survival period exceeding 14 months, and adverse reactions were not obvious. However, the survival time of first generation CAR-T cells in glioma environments is limited to within 15 days. Recent research results indicate that [8] The survival time of the CAR-T cells fused with the co-stimulatory structure is prolonged, and stronger anti-glioma activity can be exerted. In vitro experiments [9] Second generation CAR-T expressing E13K and/or E13Y can kill IL13rα2 + Without causing damage to normal tissue cells. In-situ glioma experiment of mice proves [11] The second generation E13KCAR-T cells fused with CD28 can kill tumor cells, and prolong the survival time of experimental mice. Brown et al [12] IL13-zetakine modified targeted T cells were found to kill IL13Rα2 simultaneously + Glioma cells and IL13Rα2 + Stem cell-like tumor cell subpopulations of (2), perhaps for complete eradication of IL13Rα2 + Is helpful for glioma.
In summary, the anti-IL13Rα2 antibody with high specificity and affinity can be effectively applied to preparing the CAR-T taking IL13Rα2 as a target, and has important significance in the field of glioma treatment.
Reference to the literature
1. Xu Pengfei, yang Jian, yang Xue, yuan Fanen Chen Qianxue. Research progress on childhood gliomas and adult gliomas. Journal of medical research (2019) 48 (2): 5-11.
2.Wen P YKesari S.Malignant gliomas in adults.N Engl J Med(2008)359(5):492-507.doi:10.1056/NEJMra0708126
3.Woehrer A,Bauchet LBarnholtz-Sloan J S.Glioblastoma survival:has it improvedEvidence from population-based studies.Curr Opin Neurol(2014)27(6):666-74.doi:10.1097/WCO.0000000000000144
4. Baiyue, zhong Xiaosong Li Wen recent advances in chimeric antigen receptor T cell therapy for glioblastoma multiforme Chinese tumor clinic (2017) 44 (6): 794-799.
5.Bart Thaci,Christine E.Brown,Emanuela Binello,Katherine Werbaneth,Prakash SampathSengupta S.Significance of interleukin-13receptor alpha 2-targeted glioblastoma therapy.Neuro Oncol.(2014)16(10):1304–1312.
6.Hegde M,Corder A,Chow K K,Mukherjee M,Ashoori A,Kew Y,Zhang J,Baskin D S,Merchant F A,Brawley V S,et al.Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in Glioblastoma.Mol Ther(2013).doi:10.1038/mt.2013.185mt2013185[pii].
7.Hegde M,Corder A,Chow K K,Mukherjee M,Ashoori A,Kew Y,Zhang Y J,Baskin D S,Merchant F A,Brawley V S,et al.Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma.Mol Ther(2013)21(11):2087-101.doi:10.1038/mt.2013.185.
8.Krenciute G,Krebs S,Torres D,Wu M F,Liu H,Dotti G,Li X N,Lesniak M S,Balyasnikova I VGottschalk S.Characterization and Functional Analysis of scFv-based Chimeric Antigen Receptors to Redirect T Cells to IL13Ralpha2-positive Glioma.Mol Ther(2016)24(2):354-363.doi:10.1038/mt.2015.199.
9.Debinski W,Dickinson P,Rossmeisl J H,Robertson JGibo D M.New agents for targeting of IL-13RA2expressed in primary human and canine brain tumors.PLoS One(2013)8(10):e77719.doi:10.1371/journal.pone.0077719.
10.Brown Ce,Badie B,Barish Me,Weng L,Ostberg Jr,Chang Wc,Naranjo A,Starr R4,Wagner J,Wright C4,et al.Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+T Cells in Patients with Recurrent Glioblastoma.Clin Cancer Res(2015)21(18):4062-72.
11.Kong S,Sengupta S,Tyler B,Bais A J,Ma Q,Doucette S,Zhou J,Sahin A,Carter B S,Brem H,et al.Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells.Clin Cancer Res(2012)18(21):5949-60.doi:10.1158/1078-0432.CCR-12-0319.
12.Brown C E,Starr R,Aguilar B,Shami A F,Martinez C,D'apuzzo M,Barish M E,Forman S JJensen M C.Stem-like tumor-initiating cells isolated from IL13Ralpha2expressing gliomas are targeted and killed by IL13-zetakine-redirected T Cells.Clin Cancer Res(2012)18(8):2199-209.doi:10.1158/1078-0432.CCR-11-1669.
Disclosure of Invention
Aiming at the defects and actual demands of the prior art, the invention provides an anti-IL13Ra2 nano antibody and application thereof. The nanometer antibody of the anti-IL13Ra2 has high affinity, can be used as an antigen binding domain of a chimeric antigen receptor molecule to prepare the CAR-T cell, and has good application prospect in the aspect of tumor treatment.
In order to achieve the above purpose, the invention adopts the following technical scheme:
in a first aspect, the invention provides an anti-IL13Ra2 nanobody, the amino acid sequence of CDR1 of which comprises the sequence shown in SEQ ID No.1, SEQ ID No.2 or SEQ ID No.3, the amino acid sequence of CDR2 of which comprises the sequence shown in SEQ ID No.4, SEQ ID No.5, SEQ ID No.6 or SEQ ID No.7, and the amino acid sequence of CDR3 of which comprises the sequence shown in SEQ ID No.8, SEQ ID No.9 or SEQ ID No. 10.
According to the invention, an unimmunized alpaca is immunized by IL13Ra2 recombinant protein, a phage display nanobody library is constructed, and screening is carried out on anti-IL13Ra2 antibodies according to the phage display nanobody library, so that monoclonal antibodies capable of specifically binding IL13Ra2 antigens are obtained.
SEQ ID NO.1:GFTLDYYA。
SEQ ID NO.2:GANLKNYA。
SEQ ID NO.3:RFTLDYYA。
SEQ ID NO.4:ISSSEGST。
SEQ ID NO.5:LRVRYANT。
SEQ ID NO.6:ISSSGGST。
SEQ ID NO.7:LRVRYANS。
SEQ ID NO.8:AADSTGRCWARPLYEYDY。
SEQ ID NO.9:AARPQQPSADCSLLANDYDN。
SEQ ID NO.10:AARPQQPSADCSLSANDYDN。
Preferably, the amino acid sequence of CDR1 of the nanobody comprises the sequence shown in SEQ ID NO.1, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID NO.4, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID NO. 8.
Preferably, the amino acid sequence of CDR1 of the nanobody comprises the sequence shown in SEQ ID NO.2, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID NO.5, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID NO. 9.
Preferably, the amino acid sequence of CDR1 of the nanobody comprises the sequence shown in SEQ ID NO.6, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID NO.8, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID NO. 9.
Preferably, the amino acid sequence of CDR1 of the nanobody comprises the sequence shown in SEQ ID NO.3, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID NO.4, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID NO. 8.
Preferably, the amino acid sequence of CDR1 of the nanobody comprises the sequence shown in SEQ ID NO.2, the amino acid sequence of CDR2 comprises the sequence shown in SEQ ID NO.7, and the amino acid sequence of CDR3 comprises the sequence shown in SEQ ID NO. 10.
Preferably, the heavy chain variable region of the nanobody further comprises: a framework region 1 (FR 1) shown by SEQ ID No.11, SEQ ID No.12 or SEQ ID No.13, a framework region 2 (FR 2) shown by SEQ ID No.14, SEQ ID No.15 or SEQ ID No.16, a framework region 3 (FR 3) shown by SEQ ID No.17, SEQ ID No.18, SEQ ID No.19 or SEQ ID No.20, a framework region 4 (FR 4) shown by SEQ ID No.21 or SEQ ID No. 22.
SEQ ID NO.11:QVQLVESGGGLVQPGGSLRLSCAAS。
SEQ ID NO.12:QVQLVESGGGLAHPGGRLRVTCTAS。
SEQ ID NO.13:QVQLVESGGGSVQAGGSLRLSCTSS。
SEQ ID NO.14:IAWFRQAPGKRREGVSC。
SEQ ID NO.15:VGWFRQRPGKQREEVAC。
SEQ ID NO.16:VGWFRRRPGKQREEVAC。
SEQ ID NO.17:
NYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYC。
SEQ ID NO.18:
NKAPSVRERVHVFREENNNLVYMLMSDLTPEDTGIYY。
SEQ ID NO.19:
NYANSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYC。
SEQ ID NO.20:
NKAPSVRERVHVFREEKNNLVYMLMSDLTPEDTGIYYC。
SEQ ID NO.21:RGQGTQVTVSS。
SEQ ID NO.22:WGQGIQVTVSE。
Preferably, the heavy chain variable region of the nanobody comprises the amino acid sequence shown as SEQ ID NO.23, SEQ ID NO.24, SEQ ID NO.26 or SEQ ID NO. 27.
SEQ ID NO.23:
QVQLVESGGGLVQPGGSLRLSCAASGFTLDYYAIAWFRQAPGKRREGVSCISSSEGSTNYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAADSTGRCWARPLYEYDYRGQGTQVTVSS。
SEQ ID NO.24:
QVQLVESGGGLAHPGGRLRVTCTASGANLKNYAVGWFRQRPGKQREEVACLRVRYANTNKAPSVRERVHVFREENNNLVYMLMSDLTPEDTGIYYCAARPQQPSADCSLLANDYDNWGQGIQVTVSE。
SEQ ID NO.25:
QVQLVESGGGLVQPGGSLRLSCAASGFTLDYYAIAWFRQAPGKRREGVSCISSSGGSTNYANSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAADSTGRCWARPLYEYDYRGQGTQVTVSS。
SEQ ID NO.26:
QVQLVESGGGSVQAGGSLRLSCTSSRFTLDYYAIAWFRQAPGKRREGVSCISSSEGSTNYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAADSTGRCWARPLYEYDYRGQGTQVTVSS。
SEQ ID NO.27:
QVQLVESGGGLAHPGGRLRVTCTASGANLKNYAVGWFRRRPGKQREEVACLRVRYANSNKAPSVRERVHVFREEKNNLVYMLMSDLTPEDTGIYYCAARPQQPSADCSLSANDYDNWGQGIQVTVSE。
In the invention, the phage display technology is utilized to screen an IL13Ra2 immune camel VHH library, and the obtained nano antibody has high affinity and has important application prospect in the aspect of constructing a chimeric antigen receptor of targeted IL13Ra 2.
In a second aspect, the invention provides a nucleic acid molecule comprising a gene encoding the nanobody of anti-IL13Ra2 of the first aspect.
In a third aspect, the invention provides a chimeric antigen receptor comprising a signal peptide, an antigen binding domain comprising a nanobody of anti-IL13Ra2 of the first aspect, a hinge region, a transmembrane region, and a signal transduction domain.
Preferably, the signal peptide comprises a CD8 a signal peptide.
Preferably, the hinge region comprises a CD8 a hinge region.
Preferably, the transmembrane region comprises any one or a combination of at least two of a CD8 a transmembrane region, a CD28 transmembrane region or a DAP10 transmembrane region.
Preferably, the signal transduction domain comprises an immunoreceptor tyrosine activation motif (cd3ζ).
Preferably, the signal transduction domain further comprises a co-stimulatory molecule comprising any one or a combination of at least two of 4-1BB, the intracellular domain of CD28, OX40, ICOS or the intracellular domain of DAP 10.
Preferably, the chimeric antigen receptor comprises a CD8 a signal peptide, a nanobody of anti-IL13Ra2 of the first aspect, a CD8 a hinge region, a CD8 a transmembrane region, and an immune receptor tyrosine activation motif.
In a fourth aspect, the present invention provides an expression vector comprising a gene encoding the chimeric antigen receptor of the third aspect.
Preferably, the expression vector is any one of a lentiviral vector, a retroviral vector or an adeno-associated viral vector comprising a gene encoding the chimeric antigen receptor according to the third aspect, preferably a lentiviral vector.
In a fifth aspect, the present invention provides a recombinant lentivirus comprising the expression vector of the fourth aspect.
Preferably, the recombinant lentivirus is prepared from mammalian cells transfected with the expression vector and helper plasmid of the fourth aspect.
In a sixth aspect, the present invention provides a chimeric antigen receptor immune cell expressing the chimeric antigen receptor of the third aspect.
Preferably, the chimeric antigen receptor immune cell comprises the recombinant lentivirus of the expression vector and/or the substrate aspect of the fourth aspect.
Preferably, the chimeric antigen receptor immune cells comprise any one or a combination of at least two of T cells, B cells, NK cells, mast cells or macrophages.
In a seventh aspect, the invention provides a pharmaceutical composition comprising the chimeric antigen receptor immune cell of the sixth aspect.
Preferably, the pharmaceutical composition further comprises pharmaceutically acceptable excipients.
Preferably, the auxiliary materials comprise any one or a combination of at least two of carriers, diluents, excipients, fillers, binders, wetting agents, disintegrants, emulsifiers, cosolvents, solubilizers, osmotic pressure regulators, surfactants, coating materials, colorants, pH regulators, antioxidants, bacteriostats or buffers.
In an eighth aspect, the invention provides the use of the nanobody against IL13Ra2 of the first aspect, the nucleic acid molecule of the second aspect, the chimeric antigen receptor of the third aspect, the expression vector of the fourth aspect, the recombinant lentivirus of the fifth aspect, the chimeric antigen receptor immune cell of the sixth aspect or the pharmaceutical composition of the seventh aspect in the preparation of a medicament for treating a tumor.
Preferably, the tumor comprises a tumor expressing IL13Ra 2.
Compared with the prior art, the invention has at least the following beneficial effects:
(1) The invention uses IL13Ra2 recombinant protein to immunize the non-immunized alpaca to construct phage display nanometer antibody library, and screens the anti-IL13Ra2 antibody according to the phage display nanometer antibody library, the obtained nanometer antibody can specifically bind IL13Ra2 antigen with better affinity, and the KD (M) is 1.61 multiplied by 10 respectively through the determination of antibody affinity -8 、3.24×10 -8 、1.07×10 -9 、1.66×10 -8 And 2.57×10 -9
(2) The nanometer antibody of the anti-IL13Ra2 has better affinity, is used as an antigen binding domain to construct a chimeric antigen receptor, and is used for preparing T cells, wherein the CAR-T cells have killing activity on IL13Ra2 positive tumor cells, and secrete cytokines IFN-gamma and TNF-alpha with high efficiency after being co-cultured with IL13Ra2 positive cells.
Drawings
FIG. 1A is a graph showing the affinity of the anti-IL13Ra2 nanobody VHH-1 detected by Biacore in example 2;
FIG. 1B is a graph showing the affinity of the anti-IL13Ra2 nanobody VHH-7 detected by Biacore in example 2;
FIG. 1C is a graph showing the affinity of the anti-IL13Ra2 nanobody VHH-8 detected by Biacore in example 2;
FIG. 1D is a graph showing the affinity of the anti-IL13Ra2 nanobody VHH-13 detected by Biacore in example 2;
FIG. 1E is a graph showing the affinity of the anti-IL13Ra2 nanobody VHH-16 detected by Biacore in example 2;
FIG. 2 is a graph showing the results of FACS detection of IL-13 Ra2 antigen recognized by anti-IL-13 Ra2 nanobody in example 3;
FIG. 3 is a plasmid map of a chimeric antigen receptor lentiviral vector targeting IL13Ra2 of example 4;
FIG. 4 is a schematic representation of the chimeric antigen receptor structure expressing IL13Ra2 in example 4;
FIG. 5 is a graph showing the results of flow assay of the expression rate of chimeric antigen receptor of T lymphocytes in example 6;
FIG. 6A is a FACS detection of T cells and CAR-T cell phenotype (CD 3) in example 6 + CD4 + ) The obtained result graph;
FIG. 6B is a FACS detection of T cells and CAR-T cell phenotype (CD 3) in example 6 + CD8 + ) The obtained result graph;
FIG. 7 is a graph of the killing effect of CAR-T cells on 293T cells as described in example 7;
FIG. 8 is a graph showing the killing effect of CAR-T cells on brain glioma cells U87 in example 7;
FIG. 9 is a graph showing the killing effect of CAR-T cells on glioma cell U251 in example 7;
FIG. 10 is a bar graph of TNF alpha secretion by CAR-T cells in example 8;
FIG. 11 is a bar graph of IFNγ secretion by CAR-T cells in example 8.
Detailed Description
The following embodiments are further described with reference to the accompanying drawings, but the following examples are merely simple examples of the present invention and do not represent or limit the scope of the invention, which is defined by the claims.
The specific techniques or conditions are not identified in the examples and are described in the literature in this field or are carried out in accordance with the product specifications. The reagents or equipment used were conventional products available for purchase by regular vendors, with no manufacturer noted.
Example 1
In this example, phage nanobody libraries were constructed and panning were performed and primary screening was performed using ELISA, as follows:
(1) Construction of phage nanobody library
Immunizing alpaca by adopting IL13Ra2 extracellular recombinant protein, and extracting peripheral blood after ELISA detection of serum titer; separating lymphocytes, extracting total RNA, performing reverse transcription to obtain cDNA, and amplifying VHH genes by nested PCR; VHH gene was inserted into pShort phagemid and electrotransformed
Figure BDA0003444640390000063
Separating and purifying the phage by PEG8000/NaCI precipitation method after the competent cells are amplified to obtain an antibody library; adjusting concentration, packaging, and freezing in refrigerator at-80deg.C;
(2) Screening of phage nanobody libraries
Firstly, taking 293T cells and an antibody library for co-incubation to carry out negative screening, and then taking supernatant, and respectively incubating with IL13Ra2 positive cells 293T-IL13Ra2 cells and 293T cells; washing for 4 times by adopting a pre-cooled PT buffer solution at 4 ℃; infecting NEB alpha 5F' cells, adding helper phage, and culturing overnight; coating a plate by a Drop method, and counting the enrichment degree the next day; separating and purifying the phage by PEG8000/NaCI precipitation method, and then carrying out the next round of screening; after enrichment, the VHH region was amplified using the obtained phage as a template, and subjected to second generation sequencing to obtain 5 anti-IL13Ra2 nanobodies, the amino acid sequences of which were respectively designated VHH-1, VHH-7, VHH-8, VHH-13 and VHH-16 as shown in SE1ID No.22, SE1ID No.23, SE1ID No.24, SE1ID No.25 and SE1ID No. 26.
Example 2
This example expresses and purifies the anti-IL13Ra2 nanobody (VHH-mIgG 2a Fc nanobody) screened in example 1 and determines the affinity of the antibody. In order to further identify the antibodies obtained by screening, it was necessary to express the antibodies by mammalian cells, and therefore, a plasmid vector carrying a mouse Fc tag-expressing VHH, designated C-4pCP. Stuffer-mCg a-FC, was constructed first, comprising the following steps:
(1) The VHH fragment was amplified using PCR, the reaction system of which is shown in Table 1, and the amplification procedure is shown in Table 2;
TABLE 1
Figure BDA0003444640390000062
TABLE 2
Figure BDA0003444640390000061
Figure BDA0003444640390000071
(2) The enzyme digestion system is shown in Table 3, the enzyme digestion temperature is 37 ℃ and the time is 6 hours, and the carrier after enzyme digestion is used
Figure BDA0003444640390000072
Purifying by using a PCR purification kit, dissolving the recovered DNA in 45 mu L of water, and detecting the concentration of the DNA;
TABLE 3 Table 3
Reagent(s) Usage amount
C-4pCP.Stuffer-mCg2a-FC 5μg
10 Xenzyme cutting Buffer (10 Xreaction Buffer) 5μL
FspA I 2μL
PfI 23II 2μL
ddH 2 O Make up to 50 mu L
(3) Connecting the PCR amplification product into an enzyme-digested linearization vector by adopting a homologous recombination mode, wherein the system is shown in a table 4, and the condition is that the water bath is carried out for 30min at 37 ℃;
TABLE 4 Table 4
Reagent(s) Volume (mu L)
Exnase II 1
Exnase II buffer 2
Linearization carrier (linearized vector) 4
Insert (Insert fragment) 3
(4) Adding all homologous recombination reaction systems into DH5 alpha competent cells, and transforming DH5 alpha competent cells under the transformation conditions shown in Table 5;
TABLE 5
Procedure Temperature (temperature) Time
Ice bath
0℃ 5min
Heat shock 42℃ 1min
Ice bath 0℃ 3min
Adding 500 μl of LB medium, and shaking culturing at 220rpm 37℃ 1.5h
mu.L was pipetted and spread on LB/Amp plates 37℃ Overnight
(5) The transformation plate is selected to be pre-identified by monoclonal PCR, and the conditions of a PCR identification system are shown in Table 6; the conditions are that the pre-denaturation is carried out for 3min at 95 ℃; denaturation at 95℃for 30s, annealing at 55℃for 30s, elongation at 72℃for 30s,35 cycles; extending at 72 ℃ for 5min, preserving at 4 ℃, and sending to a sequencing company for sequencing identification, wherein the sequencing result meets the expectations, and successfully constructing a plasmid vector with the mouse Fc tag for expressing VHH in the examples of the specification.
TABLE 6
Figure BDA0003444640390000073
Figure BDA0003444640390000081
293E cells were passaged approximately 24h prior to plasmid transfection to a cell density of approximately 0.6X10 6 cells/mL; when the cell density is 1.2X10 6 cell/mL, activity>At 95%, 293E cells were transfected with PEI at a ratio of 150. Mu.g DNA/100mL 293E, wherein the ratio of plasmid DNA to PEI was 1:2;
37℃、130rpm、8%CO 2 shaking culture for 6 days, collecting cell culture supernatant at 3000rpm for 30min, filtering the collected supernatant containing target antibody with 0.45 μm filter, and adding sample to purification column MabSelect TM SuRe TM The column was washed with 5 times PBS, the protein eluted with 0.1M Gly-HCl (pH 3.0) and neutralized with 1/10 volume of Tris-HCl at pH 8.5, followed by dialysis overnight at 4℃and then determination of A with Nanodrop 2000 280 The antibody purity was determined by SEC-HPLC.
In addition, in this example, the affinity of the purified VHH antibody was also determined by Biacore.
Biacore is a bioanalytical sensing technology developed based on surface plasmon resonance (surface Plasmon resonance, SPR) and can detect and track the whole change process of binding and dissociating molecules in a solution and molecules fixed on the surface of a chip, record the whole change process in the form of a sensor map and provide dynamics and affinity data.
In the measurement, the antibodies were immobilized on the chip surface, the mobile phase was a solution containing the antigen (IL 13Ra 2), and the measurement results are shown in Table 7 and FIGS. 1A to 1E, wherein the affinities of the 5 antibodies all reached sub-nanomolar levels of 1.61E-8, 3.24-8, 1.07E-9, 1.66E-8 and 2.57E-9, respectively.
TABLE 7
Nanobody Ka(1/Ms) Kd(1/s) KD(M)
VHH-1(SEQ ID NO.23) 5.53E+04 1.56E-03 1.61E-08
VHH-7(SEQ ID NO.24) 6.54E+04 6.6E-04 3.24E-08
VHH-8(SEQ ID NO.25) 4.95E+04 6.98E-03 1.07E-09
VHH-13(SEQ ID NO.26) 6.83E+04 4.33E-03 1.66E-08
VHH-16(SEQ ID NO.27) 6.25E+04 3.64E-03 2.57E-09
Example 3
This example describes a flow assay for the nanobody of example anti-IL13Ra 2.
293T (IL 13Ra2 negative, IL13Ra 2) - ) 293T-Mesothelin cells were mixed with purified anti-IL13Ra2 nanobodies, incubated for 30min in an ice bath, then incubated with APC-labeled goat anti-mouse IgG antibody for 30min, and examined using a flow cytometer, as shown in FIG. 2, indicating that the anti-IL13Ra2 nanobodies of the present invention recognize the cell surface IL13Ra2 antigen.
Example 4
This example prepares lentiviral vectors expressing a chimeric antigen receptor targeting IL13Ra2 (IL 13Ra2 CAR).
First, a lentiviral vector pSIN03IL13Ra2CAR carrying an IL13Ra2CAR chimeric antigen receptor was constructed, the vector profile of which is shown in FIG. 3, and the schematic of which is shown in FIG. 4, comprising a CD8 alpha signal peptide, a nanobody against IL13Ra2 (anti-IL 13Ra2 VHH), a CD8 alpha hinge region, a transmembrane region, and an immunoreceptor tyrosine activation motif (CD 3 zeta).
Wherein the amino acid sequence of the signal peptide (SEQ ID NO. 28) is:
MALPVTALLLPLALLLHAARP。
the amino acid sequence of the anti-IL13Ra2VHH is shown as SEQ ID NO.23, SEQ ID NO.24, SEQ ID NO.25, SEQ ID NO.26 or SEQ ID NO. 27.
The amino acid sequences of the CD 8. Alpha. Hinge region and the transmembrane region (SEQ ID NO. 29) are:
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYC。
the amino acid sequence of the intracellular region of 4-1BB (SEQ ID NO. 30) is:
KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL。
the CD3 zeta amino acid sequence (SEQ ID NO. 31) is:
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR。
the preparation method comprises the following steps:
(1) A PCR reaction system was prepared according to Table 8 (reagents in Table were derived from TOYOBO Inc.), each anti-IL13Ra2 nanobody fragment was amplified, and the reaction was performed according to the PCR procedure shown in Table 9, with primer sequences:
MS1-F(SEQ ID NO.32):
tgccgctggccttgctgctccacgccgccaggccgcaggtgcagctggtggag;
MS1-R(SEQ ID NO.33):cgctggcgtcgtggtgctagacactgtcacctg;
MS9-R(SEQ ID NO.34):cgctggcgtcgtggtagagctcactgtcacctg。
TABLE 8
Reagent(s) Volume (mu L)
10×buffer 5
2mM dNTP 5
25mM MgSO 4 3
10μM MF1-F 1
10 mu M MS1-R or MS9-R 1
Template DNA (cDNA clone) 1
Sterile deionized water (PCR grade water) 33
KOD-Plus-Neo high-fidelity PCR enzyme 1
TABLE 9
Figure BDA0003444640390000091
(2) A PCR reaction system was prepared according to Table 10, and the resulting amplified product was preceded by a CD 8. Alpha. Signal peptide, and reacted according to the PCR procedure shown in Table 9, using the primers:
BamH-CD8αsig-F(SEQ ID NO.35):
GCTGCAGGTCGACTCTAGAGGATCCCGCCACCATGGCCTTACCAGTGACCGCCTTGCTCCTGCCGCTGGCCTTGC;
table 10
Reagent(s) Volume (mu L)
10×buffer 5
2mM dNTP 5
25mM MgSO 4 3
10 mu M upstream primer (BamH-CD 8. Alpha. Sig-F) 1
10 mu M downstream primer (MS 1-R or MS 9-R) 1
Template DNA (VHH fragment PCR reaction liquid) 4
Sterile deionized water (PCR grade water) 30
KOD-Plus-Neo high-fidelity PCR enzyme 1
After the reaction is finished, the PCR product is subjected to 1% agarose gel electrophoresis, the fragment of about 500bp is recovered, and the quantity is fixed by an ultraviolet absorption method;
(3) A PCR reaction system was prepared according to Table 11, and after completion of the preparation, a PCR reaction was performed according to the PCR procedure shown in Table 9 to amplify the CD 8. Alpha. Range-TM-41 BB-CD3Z fragment, using the primers as follows:
CD8αH-F(SEQ ID NO.36):ACCACGACGCCAGCGCCGCGAC;
Vector-R(SEQ ID NO.37):TCGATAAGCTTGATATCG;
TABLE 11
Reagent(s) Volume (mu L)
10×buffer 5
2mM dNTP 5
25mM MgSO 4 3
10 mu M upstream primer CD8 alpha H-F 1
10 mu M downstream primer Vector-R 1
Template DNA (HD CD19 CAR) 1
Sterile deionized water (PCR grade water) 33
KOD-Plus-Neo high-fidelity PCR enzyme 1
After the PCR is finished, carrying out 1% agarose gel electrophoresis, recovering fragments about 780bp, and quantifying by an ultraviolet absorption method;
(4) Carrying out BamHI and EcoRI double digestion on 5 μg of HD SIN03 CD19 CAR plasmid constructed in a laboratory, and recovering the vector after water bath reaction for 2h at 37 ℃;
the 3 fragments recovered above and the vector backbone were linked by recombinase, the recombination reaction system is shown in Table 12, and after completion of the preparation, the mixture was subjected to a water bath reaction at 37℃for 0.5h, and transformed into E.coli stbl3 competent cells by a conventional method.
Table 12
Reagent(s) Usage amount
HD CD19 CAR 184.54ng
CD8α singal IL13Ra2 VHH 31.32ng
CD8α hinge-TM-41BB-CD3Z 29.72ng
5×CE buffer 2μL
Exnase TM II 1μL
Sterile deionized water (PCR grade water) Make up to 10 mu L
And selecting a monoclonal from the solid culture medium, culturing overnight, carrying out PCR identification, preparing a PCR reaction system as shown in table 13, carrying out a PCR procedure as shown in table 14, selecting a positive clone after the PCR is finished, and further carrying out sequencing identification, wherein the sequencing result meets the expectations.
TABLE 13
Reagent(s) Volume (mu L)
Taq PCR Master Mix 10
10μM F Seq-trEF1a-F 1
10μM R Vector-R 1
Template DNA bacterial liquid 1
Sterile deionized water (PCR grade water) 7
TABLE 14
Figure BDA0003444640390000101
Figure BDA0003444640390000111
Example 5
In this example, lentiviral packaging, concentration and titer detection were performed on the lentiviral vector HD SIN03-IL13 Ra2CAR prepared in example 4, comprising the steps of:
(1) Lentivirus package
At 1.6X10 7 Cell count 293T cells were seeded in 15cm dishes at 37℃with 5% CO 2 The virus is prepared for packaging after overnight culture, and the culture medium is DMEM containing 10% of fetal calf serum; the lentiviral vector 14.5. Mu.g HD SIN03-IL13 Ra2CAR, 16.7. Mu.g helper plasmid pMDLg-RRE, 16.7. Mu.g helper plasmid pRSV-REV and 6.5. Mu.g envelope plasmid VSVg were dissolved in 2mL serum-free DMEM medium and mixed well;
163.2. Mu.g PEI (1. Mu.g/. Mu.L) was dissolved in 2mL serum-free DMEM medium, vortexed at 1000rpm for 5 seconds and incubated at 25℃for 5min; adding PEI mixed solution into the DNA mixed solution, immediately vortex mixing or gently mixing after adding, and incubating for 20min at 25 ℃ to form a transfection complex; then, 4mL of the transfection complex is added into 25mL of DMEM medium containing 293T cells in a dropwise manner, and after 4 hours, the fresh medium is replaced; after 48 hours, collecting the virus liquid supernatant;
(2) Lentivirus concentration
Filtering the virus supernatant with a 0.45 μm filter membrane, collecting the filtered virus supernatant into a 50mL centrifuge tube, adding 1/4 PEG-NaCl virus concentrate, mixing the mixture upside down, and standing the mixture at 4 ℃ overnight; centrifuging at 4 ℃ at 3500rpm for 30min; removing supernatant, adding appropriate amount of RPMI 1640 medium (containing 10% FBS), and dissolving the resuspended virus precipitate; split charging 50 μl of the concentrated lentiviral suspension into each portion, storing in a finished tube, and storing at-80deg.C;
(3) Lentivirus titer detection
500 mu L K562 cells (1X 10) 5 Individual cells) were inoculated into 24-well plates, and the concentrated lentiviruses were added to the cell suspension in volumes of 1. Mu.L, 0.2. Mu.L and 0.04. Mu.L, respectively, and polybrene was added to a final concentration of 5. Mu.g/mL, 37℃at 5% CO 2 After overnight incubation, fresh medium was changed;
after 72h infection, 400 Xg was centrifuged for 5min, the supernatant was discarded to collect cells, 100. Mu.L PBS+2% FBS was added to resuspend the cells, PE coat anti-alpaca VHH antibody was added and incubated on ice for 30min; after 2 times of washing with the flow buffer, 300 mu L of the flow buffer is added to resuspend cells, and a flow cytometer is adopted to detect the infection efficiency; the titer was calculated as follows: titer (TU/mL) =cell number (10 5 ) X positive rate/viral volume (mL).
Example 6
This example uses lentivirus transduced T lymphocytes prepared in example 5, comprising the steps of:
(1) Diluting the anti-human CD3 antibody and the anti-human CD28 antibody with PBS (phosphate buffered saline) to obtain final concentrations of 1 mug/mL and 0.5 mug/mL respectively, coating an orifice plate, and standing at 4 ℃ in a refrigerator overnight; discarding the antibody coating liquid in the pore plate, and washing the plate by 1 mLPBS;
(2) Human PBMC were conditioned to a density of 1X 10 with T cell culture medium (X-VIVO+10% FBS+300U/mL IL-2) 6 Per mL, inoculated into CD3 and CD28 antibody coated well plates for 24h activation; the activated T cells were collected and the cell density was adjusted to 1X 10 6 Lentivirus was added per mL at a multiplicity of infection (multiplicity of infection, MOI) of 10, polybrene was added to a final concentration of 5 μg/mL; at 37℃with 5% CO 2 Replacing fresh culture medium after overnight culture in the environment, and carrying out passage every 3 days;
(3) 5 days after T cell infection, 3X 10 cells were taken 5 Centrifuging at 4 ℃ for 5min at 400 Xg, discarding the supernatant, and washing once with streaming buffer; adding 100 mu L of buffer solution to resuspend cells, adding PE coat anti-alpaca VHH antibody, and incubating on ice for 30min; after 2 washes with buffer, 300 μl buffer was added to resuspend cells;
the expression rate of chimeric antigen receptor of T lymphocytes was measured by flow cytometry, and as a result, as shown in fig. 5, the infection efficiency of each group of CAR-T cells was: 24.8%, 25.8%, 14.8%, 26.2%, and 23.9%, indicating successful construction of CAR-T cells.
In addition, the flow cytometer is used to detect lymphocyte phenotypes in this example, including the steps of:
(1) 5 days after T cell infection, 3X 10 cells were taken 5 Centrifuging at 4 ℃ for 5min at 400g, discarding the supernatant, and washing with PBS (containing 2% by mass of FBS) buffer solution once;
(2) Cells were resuspended in 50. Mu.L of buffer, 1. Mu.L of FITC-labeled Anti-CD3 Ab, percp-Cy5.5 labeled Anti-CD4 Ab and PE-Cy7 labeled Anti-CD8 Ab were added and incubated on ice for 30min; after washing twice with buffer, cells were resuspended in 300. Mu.L of buffer and examined for cell phenotype using flow cytometry, as shown in FIGS. 6A and 6B, CD3 + CD4 + Cell population ratio is 33-37%, CD3 + CD8 + The cell population was 61-66%.
Example 7
In this example, an in vitro toxicity test of CAR-T cells was performed, comprising the steps of:
(1) Target cell inoculation
293-GPF-luci (IL 13)Ra2 negative, IL13Ra2 - )、U251-GFP-luci(IL13Ra2 + )、U87-GFP-luci(IL13Ra2 + ) As target cells, the concentration of target cells was adjusted to 2X 10 5 50. Mu.L of the solution was inoculated into a white 96-well plate;
(2) Effector cell seeding
IL13Ra2 CAR-T and control T cells are effector cells, and CAR-T cells and control T cells are added into a 96-well plate according to the effective target ratio of 0.3:1 and 1:1;
(3) Each group was set with 3 duplicate wells and the average of 3 duplicate wells was taken, wherein each experimental group and each control group were as follows:
experimental group: each target cell + CAR-T;
control group: inoculating only target cells;
(4) The detection method comprises the following steps:
after 18h of co-culture of effector cells with target cells, 100. Mu.L was added to each well
Figure BDA0003444640390000121
Reagents (Promega, cat#E2520), after 5min of reaction, the bioluminescence signal was detected using a multifunctional microplate reader;
(5) The CAR-T killing efficiency calculation formula is as follows:
killing efficiency% = (control-experimental)/control x 100%.
The results are shown in figures 7-9, the CAR-T cells constructed by the invention have no killing effect on IL13Ra2 negative 293T cells and have killing activity on IL13Ra2 positive tumor cells, and the CAR-T cells constructed by the invention have high-efficiency tumor killing activity and high specificity.
Example 8
In this example, the secretion of the CAR-T cytokines TNF- α and IFN- γ was detected using the kits of Human TNF- α ELISA Kit (co-products, cat# EK 182-96) and Human IFN- γ ELISA Kit (co-products, cat# EK 180-96), respectively.
1. Cell culture supernatant
Cell cultures with an effective target ratio of 1:1 were centrifuged at 400 Xg for 10min to remove sediment, and the supernatant was stored at-80℃for examination.
2. Reagent preparation
All reagents, samples were returned to 25℃before testing, and if crystallization occurred in the concentrated reagents, the incubation was performed at 37℃until the crystals were completely dissolved, and 1 Xof wash solution and 1 Xof assay buffer were prepared according to the instructions of use.
3. Standard substance and sample preparation
Standard substance: standard stock was 2-fold diluted with 5%1640 medium for a total of 8 dilution gradients, including zero concentration.
Sample: samples were diluted in ratio using 5%1640 medium.
4. Detection step
(1) Soaking the ELISA plate: adding 300 mu L of 1 Xwashing liquid, standing and soaking for 30s, discarding the washing liquid, and beating the micro-pore plate on water-absorbing paper;
(2) Adding a standard substance: standard wells were filled with 100 μl of 2-fold diluted standard and blank wells were filled with 100 μl of standard diluent (serum/plasma samples);
(3) Adding a sample: sample wells were added with 100 μl of cell culture supernatant;
(4) Adding a detection antibody: 50. Mu.L of diluted detection antibody (1:100 dilution) was added to each well;
(5) Incubation: sealing plates by using sealing plates, vibrating at 300rpm, and incubating at 25 ℃ for 2 hours;
(6) Washing: liquid was discarded, and 300. Mu.L of wash solution was added to wash the plate 6 times per well;
(7) And (3) enzyme adding and incubation: mu.L of diluted horseradish peroxidase-labeled streptavidin (1:100 dilution) was added to each well;
(8) Incubation: using a new sealing plate membrane sealing plate, oscillating at 300rpm, incubating at 25 ℃ for 45min, and washing;
(9) And (3) color development of the substrate: 100 mu L of chromogenic substrate TMB is added into each hole, and incubated for 20min at 25 ℃ in the dark;
(10) Adding a stop solution: 100 mu L of stop solution is added to each well;
(11) Detecting and reading: within 30min, performing dual-wavelength detection by using an enzyme-labeled instrument, and measuring the OD value at the maximum absorption wavelength of 450nm and the reference wavelength; the OD value after calibration was 450nm minus the measurement at the reference wavelength.
TNF- α, IFN- γ factor secretion results are shown in FIGS. 10 and 11, respectively, wherein spontaneous is a single CAR-T cell group, no cytokines were detected; no cytokines were detected in the CAR-T cell co-cultured group with 293T cells; after co-culture with target cells over-expressing MSLN, the TNF-alpha secreted by the CAR-T cells exceeds 200pg/mL, and the IFN-gamma exceeds 3000pg/mL, and the constructed CAR-T cells release cytokines to tumor cells positive for IL13Ra2, but have no obvious cytokine secretion to IL13Ra2 negative cells.
In conclusion, the nano antibody with high affinity for resisting IL13Ra2 can be screened and prepared, can be combined with IL13Ra2 in a specific way, and can be used as an antigen binding domain to construct a chimeric antigen receptor and a CAR-T cell, and the obtained CAR-T cell has obvious killing activity and specificity for tumor cells positive to IL13Ra2 and can secrete cytokines for killing tumors, so that the nano antibody can be effectively applied to immunotherapy, and has important significance for developing tumor therapeutic drugs.
The applicant declares that the above is only a specific embodiment of the present invention, but the scope of the present invention is not limited thereto, and it should be apparent to those skilled in the art that any changes or substitutions that are easily conceivable within the technical scope of the present invention disclosed by the present invention fall within the scope of the present invention and the disclosure.
Sequence listing
<110> Hua Dao (Shanghai) biological medicine Co., ltd
<120> an anti-IL13Ra2 nanobody and use thereof
<130> 2021-12-28
<160> 37
<170> PatentIn version 3.3
<210> 1
<211> 8
<212> PRT
<213> artificial sequence
<400> 1
Gly Phe Thr Leu Asp Tyr Tyr Ala
1 5
<210> 2
<211> 8
<212> PRT
<213> artificial sequence
<400> 2
Gly Ala Asn Leu Lys Asn Tyr Ala
1 5
<210> 3
<211> 8
<212> PRT
<213> artificial sequence
<400> 3
Arg Phe Thr Leu Asp Tyr Tyr Ala
1 5
<210> 4
<211> 8
<212> PRT
<213> artificial sequence
<400> 4
Ile Ser Ser Ser Glu Gly Ser Thr
1 5
<210> 5
<211> 8
<212> PRT
<213> artificial sequence
<400> 5
Leu Arg Val Arg Tyr Ala Asn Thr
1 5
<210> 6
<211> 8
<212> PRT
<213> artificial sequence
<400> 6
Ile Ser Ser Ser Gly Gly Ser Thr
1 5
<210> 7
<211> 8
<212> PRT
<213> artificial sequence
<400> 7
Leu Arg Val Arg Tyr Ala Asn Ser
1 5
<210> 8
<211> 18
<212> PRT
<213> artificial sequence
<400> 8
Ala Ala Asp Ser Thr Gly Arg Cys Trp Ala Arg Pro Leu Tyr Glu Tyr
1 5 10 15
Asp Tyr
<210> 9
<211> 20
<212> PRT
<213> artificial sequence
<400> 9
Ala Ala Arg Pro Gln Gln Pro Ser Ala Asp Cys Ser Leu Leu Ala Asn
1 5 10 15
Asp Tyr Asp Asn
20
<210> 10
<211> 20
<212> PRT
<213> artificial sequence
<400> 10
Ala Ala Arg Pro Gln Gln Pro Ser Ala Asp Cys Ser Leu Ser Ala Asn
1 5 10 15
Asp Tyr Asp Asn
20
<210> 11
<211> 25
<212> PRT
<213> artificial sequence
<400> 11
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser
20 25
<210> 12
<211> 25
<212> PRT
<213> artificial sequence
<400> 12
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Ala His Pro Gly Gly
1 5 10 15
Arg Leu Arg Val Thr Cys Thr Ala Ser
20 25
<210> 13
<211> 25
<212> PRT
<213> artificial sequence
<400> 13
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Ser Val Gln Ala Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Thr Ser Ser
20 25
<210> 14
<211> 17
<212> PRT
<213> artificial sequence
<400> 14
Ile Ala Trp Phe Arg Gln Ala Pro Gly Lys Arg Arg Glu Gly Val Ser
1 5 10 15
Cys
<210> 15
<211> 17
<212> PRT
<213> artificial sequence
<400> 15
Val Gly Trp Phe Arg Gln Arg Pro Gly Lys Gln Arg Glu Glu Val Ala
1 5 10 15
Cys
<210> 16
<211> 17
<212> PRT
<213> artificial sequence
<400> 16
Val Gly Trp Phe Arg Arg Arg Pro Gly Lys Gln Arg Glu Glu Val Ala
1 5 10 15
Cys
<210> 17
<211> 38
<212> PRT
<213> artificial sequence
<400> 17
Asn Tyr Ala Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn
1 5 10 15
Ala Lys Asn Thr Val Tyr Leu Gln Met Asn Ser Leu Lys Pro Glu Asp
20 25 30
Thr Ala Val Tyr Tyr Cys
35
<210> 18
<211> 37
<212> PRT
<213> artificial sequence
<400> 18
Asn Lys Ala Pro Ser Val Arg Glu Arg Val His Val Phe Arg Glu Glu
1 5 10 15
Asn Asn Asn Leu Val Tyr Met Leu Met Ser Asp Leu Thr Pro Glu Asp
20 25 30
Thr Gly Ile Tyr Tyr
35
<210> 19
<211> 38
<212> PRT
<213> artificial sequence
<400> 19
Asn Tyr Ala Asn Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn
1 5 10 15
Ala Lys Asn Thr Val Tyr Leu Gln Met Asn Ser Leu Lys Pro Glu Asp
20 25 30
Thr Ala Val Tyr Tyr Cys
35
<210> 20
<211> 38
<212> PRT
<213> artificial sequence
<400> 20
Asn Lys Ala Pro Ser Val Arg Glu Arg Val His Val Phe Arg Glu Glu
1 5 10 15
Lys Asn Asn Leu Val Tyr Met Leu Met Ser Asp Leu Thr Pro Glu Asp
20 25 30
Thr Gly Ile Tyr Tyr Cys
35
<210> 21
<211> 11
<212> PRT
<213> artificial sequence
<400> 21
Arg Gly Gln Gly Thr Gln Val Thr Val Ser Ser
1 5 10
<210> 22
<211> 11
<212> PRT
<213> artificial sequence
<400> 22
Trp Gly Gln Gly Ile Gln Val Thr Val Ser Glu
1 5 10
<210> 23
<211> 125
<212> PRT
<213> artificial sequence
<400> 23
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Leu Asp Tyr Tyr
20 25 30
Ala Ile Ala Trp Phe Arg Gln Ala Pro Gly Lys Arg Arg Glu Gly Val
35 40 45
Ser Cys Ile Ser Ser Ser Glu Gly Ser Thr Asn Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Val Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Lys Pro Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Ala Asp Ser Thr Gly Arg Cys Trp Ala Arg Pro Leu Tyr Glu Tyr
100 105 110
Asp Tyr Arg Gly Gln Gly Thr Gln Val Thr Val Ser Ser
115 120 125
<210> 24
<211> 127
<212> PRT
<213> artificial sequence
<400> 24
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Ala His Pro Gly Gly
1 5 10 15
Arg Leu Arg Val Thr Cys Thr Ala Ser Gly Ala Asn Leu Lys Asn Tyr
20 25 30
Ala Val Gly Trp Phe Arg Gln Arg Pro Gly Lys Gln Arg Glu Glu Val
35 40 45
Ala Cys Leu Arg Val Arg Tyr Ala Asn Thr Asn Lys Ala Pro Ser Val
50 55 60
Arg Glu Arg Val His Val Phe Arg Glu Glu Asn Asn Asn Leu Val Tyr
65 70 75 80
Met Leu Met Ser Asp Leu Thr Pro Glu Asp Thr Gly Ile Tyr Tyr Cys
85 90 95
Ala Ala Arg Pro Gln Gln Pro Ser Ala Asp Cys Ser Leu Leu Ala Asn
100 105 110
Asp Tyr Asp Asn Trp Gly Gln Gly Ile Gln Val Thr Val Ser Glu
115 120 125
<210> 25
<211> 125
<212> PRT
<213> artificial sequence
<400> 25
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Leu Asp Tyr Tyr
20 25 30
Ala Ile Ala Trp Phe Arg Gln Ala Pro Gly Lys Arg Arg Glu Gly Val
35 40 45
Ser Cys Ile Ser Ser Ser Gly Gly Ser Thr Asn Tyr Ala Asn Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Val Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Lys Pro Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Ala Asp Ser Thr Gly Arg Cys Trp Ala Arg Pro Leu Tyr Glu Tyr
100 105 110
Asp Tyr Arg Gly Gln Gly Thr Gln Val Thr Val Ser Ser
115 120 125
<210> 26
<211> 125
<212> PRT
<213> artificial sequence
<400> 26
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Ser Val Gln Ala Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Thr Ser Ser Arg Phe Thr Leu Asp Tyr Tyr
20 25 30
Ala Ile Ala Trp Phe Arg Gln Ala Pro Gly Lys Arg Arg Glu Gly Val
35 40 45
Ser Cys Ile Ser Ser Ser Glu Gly Ser Thr Asn Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Val Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Lys Pro Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Ala Asp Ser Thr Gly Arg Cys Trp Ala Arg Pro Leu Tyr Glu Tyr
100 105 110
Asp Tyr Arg Gly Gln Gly Thr Gln Val Thr Val Ser Ser
115 120 125
<210> 27
<211> 127
<212> PRT
<213> artificial sequence
<400> 27
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Ala His Pro Gly Gly
1 5 10 15
Arg Leu Arg Val Thr Cys Thr Ala Ser Gly Ala Asn Leu Lys Asn Tyr
20 25 30
Ala Val Gly Trp Phe Arg Arg Arg Pro Gly Lys Gln Arg Glu Glu Val
35 40 45
Ala Cys Leu Arg Val Arg Tyr Ala Asn Ser Asn Lys Ala Pro Ser Val
50 55 60
Arg Glu Arg Val His Val Phe Arg Glu Glu Lys Asn Asn Leu Val Tyr
65 70 75 80
Met Leu Met Ser Asp Leu Thr Pro Glu Asp Thr Gly Ile Tyr Tyr Cys
85 90 95
Ala Ala Arg Pro Gln Gln Pro Ser Ala Asp Cys Ser Leu Ser Ala Asn
100 105 110
Asp Tyr Asp Asn Trp Gly Gln Gly Ile Gln Val Thr Val Ser Glu
115 120 125
<210> 28
<211> 21
<212> PRT
<213> artificial sequence
<400> 28
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala Ala Arg Pro
20
<210> 29
<211> 69
<212> PRT
<213> artificial sequence
<400> 29
Thr Thr Thr Pro Ala Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala
1 5 10 15
Ser Gln Pro Leu Ser Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly
20 25 30
Gly Ala Val His Thr Arg Gly Leu Asp Phe Ala Cys Asp Ile Tyr Ile
35 40 45
Trp Ala Pro Leu Ala Gly Thr Cys Gly Val Leu Leu Leu Ser Leu Val
50 55 60
Ile Thr Leu Tyr Cys
65
<210> 30
<211> 42
<212> PRT
<213> artificial sequence
<400> 30
Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro Phe Met
1 5 10 15
Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys Arg Phe
20 25 30
Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu
35 40
<210> 31
<211> 112
<212> PRT
<213> artificial sequence
<400> 31
Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Lys Gln Gly
1 5 10 15
Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr
20 25 30
Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys
35 40 45
Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys
50 55 60
Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg
65 70 75 80
Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala
85 90 95
Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
100 105 110
<210> 32
<211> 53
<212> DNA
<213> artificial sequence
<400> 32
tgccgctggc cttgctgctc cacgccgcca ggccgcaggt gcagctggtg gag 53
<210> 33
<211> 33
<212> DNA
<213> artificial sequence
<400> 33
cgctggcgtc gtggtgctag acactgtcac ctg 33
<210> 34
<211> 33
<212> DNA
<213> artificial sequence
<400> 34
cgctggcgtc gtggtagagc tcactgtcac ctg 33
<210> 35
<211> 75
<212> DNA
<213> artificial sequence
<400> 35
gctgcaggtc gactctagag gatcccgcca ccatggcctt accagtgacc gccttgctcc 60
tgccgctggc cttgc 75
<210> 36
<211> 22
<212> DNA
<213> artificial sequence
<400> 36
accacgacgc cagcgccgcg ac 22
<210> 37
<211> 18
<212> DNA
<213> artificial sequence
<400> 37
tcgataagct tgatatcg 18

Claims (19)

1. An anti-IL13Ra2 nanobody is characterized in that the amino acid sequence of CDR1 of the nanobody is shown as SEQ ID NO. 1;
the amino acid sequence of CDR2 of the nano antibody is shown as SEQ ID NO. 4;
the amino acid sequence of the CDR3 of the nano antibody is shown as SEQ ID NO. 8.
2. A nucleic acid molecule comprising a gene encoding the nanobody of claim 1 against IL13Ra 2.
3. A chimeric antigen receptor, wherein the chimeric antigen receptor comprises a signal peptide, an antigen binding domain, a hinge region, a transmembrane region, and a signal transduction domain;
the antigen binding domain comprises the nanobody of anti-IL13Ra2 of claim 1.
4. The chimeric antigen receptor according to claim 3, wherein the signal peptide comprises a CD8 a signal peptide.
5. The chimeric antigen receptor according to claim 3, wherein the hinge region comprises a CD8 a hinge region.
6. The chimeric antigen receptor according to claim 3, wherein the transmembrane region comprises any one or a combination of at least two of a CD8 a transmembrane region, a CD28 transmembrane region, or a DAP10 transmembrane region.
7. The chimeric antigen receptor according to claim 3, wherein the signaling domain comprises an immunoreceptor tyrosine activation motif.
8. The chimeric antigen receptor according to claim 3, wherein the signaling domain further comprises a co-stimulatory molecule comprising any one or a combination of at least two of 4-1BB, CD28 intracellular region, OX40, ICOS or DAP10 intracellular region.
9. The chimeric antigen receptor according to claim 3, wherein the chimeric antigen receptor comprises a CD8 a signal peptide, the nanobody of anti-IL13Ra2 of claim 1, a CD8 a hinge region, a CD8 a transmembrane region, and an immunoreceptor tyrosine activation motif.
10. An expression vector comprising a gene encoding the chimeric antigen receptor of any one of claims 3-9.
11. The expression vector according to claim 10, wherein the expression vector is any one of a lentiviral vector, a retroviral vector or an adeno-associated viral vector comprising a gene encoding the chimeric antigen receptor according to any one of claims 3 to 9.
12. A recombinant lentivirus comprising the expression vector of claim 10.
13. A chimeric antigen receptor immune cell, characterized in that it expresses the chimeric antigen receptor of any one of claims 3 to 9.
14. The chimeric antigen receptor immune cell according to claim 13, wherein the chimeric antigen receptor immune cell comprises the expression vector of claim 10 and/or the recombinant lentivirus of claim 12.
15. The chimeric antigen receptor-immune cell according to claim 13, wherein the chimeric antigen receptor-immune cell comprises any one or a combination of at least two of T cells, B cells, NK cells, mast cells or macrophages.
16. A pharmaceutical composition comprising the chimeric antigen receptor immune cell of claim 13.
17. The pharmaceutical composition of claim 16, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable adjuvant.
18. The pharmaceutical composition of claim 17, wherein the adjuvant comprises any one or a combination of at least two of a carrier, diluent, excipient, filler, binder, wetting agent, disintegrant, emulsifier, co-solvent, solubilizer, osmotic pressure regulator, surfactant, coating material, colorant, pH regulator, antioxidant, bacteriostat, or buffer.
19. Use of the nanobody of anti-IL13Ra2 of claim 1, the nucleic acid molecule of claim 2, the chimeric antigen receptor of claim 3, the expression vector of claim 10, the recombinant lentivirus of claim 12, the chimeric antigen receptor immune cell of claim 13, or the pharmaceutical composition of claim 16 for the preparation of a medicament for the treatment of a tumor expressing IL13Ra 2.
CN202111681616.6A 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof Active CN114409782B (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CN202310611114.9A CN116589585B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202111681616.6A CN114409782B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610963.2A CN116514982B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610970.2A CN116813773A (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310611121.9A CN116444671B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111681616.6A CN114409782B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof

Related Child Applications (4)

Application Number Title Priority Date Filing Date
CN202310610970.2A Division CN116813773A (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310611114.9A Division CN116589585B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310611121.9A Division CN116444671B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610963.2A Division CN116514982B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof

Publications (2)

Publication Number Publication Date
CN114409782A CN114409782A (en) 2022-04-29
CN114409782B true CN114409782B (en) 2023-06-30

Family

ID=81271191

Family Applications (5)

Application Number Title Priority Date Filing Date
CN202310611121.9A Active CN116444671B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610963.2A Active CN116514982B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610970.2A Pending CN116813773A (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310611114.9A Active CN116589585B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202111681616.6A Active CN114409782B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof

Family Applications Before (4)

Application Number Title Priority Date Filing Date
CN202310611121.9A Active CN116444671B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610963.2A Active CN116514982B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310610970.2A Pending CN116813773A (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof
CN202310611114.9A Active CN116589585B (en) 2021-12-29 2021-12-29 anti-IL13Ra2 nano antibody and application thereof

Country Status (1)

Country Link
CN (5) CN116444671B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114805581B (en) * 2022-06-29 2022-10-14 上海优替济生生物医药有限公司 Antibodies targeting IL13RA2, chimeric antigen receptors and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112236151A (en) * 2018-03-14 2021-01-15 西雅图儿童医院(Dba西雅图儿童研究所) IL-13 receptor alpha 2(IL13RA2) chimeric antigen receptor for tumor-specific T cell immunotherapy
CN113621073A (en) * 2021-07-14 2021-11-09 上海易慕峰生物科技有限公司 Novel chimeric receptor composition, recombinant vector, cell and application thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010190572A (en) * 2007-06-01 2010-09-02 Sapporo Medical Univ Antibody directed against il13ra2, and diagnostic/therapeutic agent comprising the antibody
MX2019006631A (en) * 2016-12-12 2019-11-12 Seattle Childrens Hospital Dba Seattle Childrens Res Inst Chimeric transcription factor variants with augmented sensitivity to drug ligand induction of transgene expression in mammalian cells.
CN108456250A (en) * 2017-02-17 2018-08-28 科济生物医药(上海)有限公司 Target antibody and its application of IL-13RA2
WO2018156711A1 (en) * 2017-02-22 2018-08-30 H. Lee Moffitt Cancer Center And Research Institute, Inc. Il13ra2-binding chimeric antigen receptors
CN109503716B (en) * 2018-10-08 2021-04-27 浙江生研生物科技有限公司 Bispecific chimeric antigen receptor molecule and application thereof in tumor treatment
CN112521502A (en) * 2019-09-19 2021-03-19 北京慧能安生物科技有限公司 anti-IL-13R alpha 2 nano antibody and coding sequence and application thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112236151A (en) * 2018-03-14 2021-01-15 西雅图儿童医院(Dba西雅图儿童研究所) IL-13 receptor alpha 2(IL13RA2) chimeric antigen receptor for tumor-specific T cell immunotherapy
CN113621073A (en) * 2021-07-14 2021-11-09 上海易慕峰生物科技有限公司 Novel chimeric receptor composition, recombinant vector, cell and application thereof

Also Published As

Publication number Publication date
CN116589585A (en) 2023-08-15
CN116514982A (en) 2023-08-01
CN116514982B (en) 2023-12-19
CN116589585B (en) 2023-11-24
CN116444671A (en) 2023-07-18
CN116813773A (en) 2023-09-29
CN116444671B (en) 2024-02-23
CN114409782A (en) 2022-04-29

Similar Documents

Publication Publication Date Title
JP7280828B2 (en) Antibodies targeting BCMA and uses thereof
WO2020108645A1 (en) Cd19-and bcma-based combined car-t immunotherapy
CN112812186B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN114409782B (en) anti-IL13Ra2 nano antibody and application thereof
CN112812185B (en) Monoclonal antibody for resisting B cell maturation antigen and application thereof
CN113265001B (en) Nanometer antibody for resisting B cell mature antigen and application thereof
CN112830971B (en) anti-BCMA antigen binding fragment and application thereof
WO2021057866A1 (en) Single domain antibody and chimeric antigen receptor comprising antibody structure
CN116063532B (en) Antibodies that bind anti-mesothelin and uses thereof
CN115894697B (en) Nanometer antibody for resisting guanylate cyclase 2C and application thereof
CN115925948B (en) anti-CD 22 nano antibody and application thereof
CN112979808B (en) Antibody for resisting B cell mature antigen and application thereof
WO2023024084A1 (en) Chimeric antigen receptor and use thereof
CN115873802A (en) Chimeric antigen receptor immune cell and preparation method and application thereof
WO2023169555A9 (en) Chimeric antigen receptor targeting bcma and gprc5d and application thereof
WO2023273762A1 (en) Spatial conformational epitope mediating efficient retention of cd3 within cells and application thereof
US20230242666A1 (en) Methods and Compositions for the Reduction of Chimeric Antigen Receptor Tonic Signaling
CN116675773A (en) anti-MXR 7 antibody and application thereof
CN116693688A (en) anti-Intestinal protein OCI-5 antibody and application thereof
CN116410323A (en) anti-GPC 3 antibody and application thereof
CN116574181A (en) anti-Glypican 3 antibody and application thereof
CN116640217A (en) anti-GTR 2-2 antibody and application thereof
CN116284389A (en) anti-AFP/HLA 02 TCR-like antibodies and uses thereof
CN117430719A (en) MSLN &amp; CD70 double-target chimeric antigen receptor 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