The present invention belongs to the field of biomedical technology, and particularly relates to a hypoxia-triggered artificial transcription factor. The present invention further relates to a hypoxia-triggered transcription control system including the hypoxia-triggered artificial transcription factor, and use thereof in the treatment of hypoxic tumors.
Immune cell therapies for tumors, such as T cell receptor T cells (TCR-T cells) and chimeric antigen receptor T cells (CAR-T cells), have shown good anti-tumor effects in clinical studies. However, in clinical trials and applications, new questions continue to arise with cell therapies. In addition to the difficulty of entering dense solid tumors, common adverse reactions include off-target effects due to tumor-specific antigen deficiency, potentially fatal neurotoxicity, allergic reactions and other side effects, which limit the clinical applications of cell therapies for solid tumors. Therefore, it is necessary to further develop an immune cell therapy that can be regulated and precisely targeted at tumors, in order to reduce the damages to normal tissues.
It has been found that hypoxia is a common feature of many solid tumors due to insufficient blood supply caused by abnormal vascular structures in solid tumors and excessive proliferation of tumor cells, creating a relatively hypoxic tumor microenvironment, with an oxygen level in tumor tissues being often less than 2%. Aiming at the hypoxic microenvironment in solid tumors, the development of tumor killing technologies that activate TCR-T cells, CAR-T cells or genetically engineered T cells through hypoxia signals is expected to improve the specificity of tumor therapy.
Previous studies have shown that fusing an oxygen-dependent degradation domain (ODD) of hypoxia-inducible factors 1-alpha (HIF-1α) into a chimeric antigen receptor can degrade the chimeric antigen receptor in a normoxic environment and enrich the chimeric antigen receptor in a hypoxic environment, thereby recognizing and killing tumor cells. However, an induction level of the chimeric antigen receptor fused with the oxygen-dependent degradation domain in the hypoxic environment is still at a low level, which is not conducive to the control of tumors with large antigenic heterogeneity, especially solid tumors with low tumor antigen expression. In addition, the current ODD-regulated hypoxia-sensitive CAR-T cells still have a high level of background leakage and can produce a certain level of non-hypoxia-dependent killing, resulting in safety concerns. In addition, although CAR-T cells driven by hypoxia-triggered promoters have a certain ability to respond to oxygen concentration, they also have a higher level of background leakage, thereby resulting in a higher level of non-hypoxia-dependent killing, which may produce on-target off-tumor toxicity caused by targeting non-tumor cells.
Therefore, it is necessary to further develop more sensitive and efficient hypoxia-triggered artificial transcription factors, so as to develop safer and more effective hypoxia-sensitive treatment methods, which can not only improve the safety of cell therapy, but also effectively control tumor growth.
An object of the present invention is to provide a hypoxia-triggered transcription control system against the defects of the prior art. The hypoxia-triggered transcription control system provided by the present invention includes two sets of transcription control units linked upstream and downstream. The present invention further provides use of the hypoxia-triggered transcription control system. The transcription control system provided by the present invention is composed of two sets of linked transcription control units, forming upstream and downstream two-stage amplifications for achieving high sensitivity to hypoxia. In addition, the system can realize the preparation of a medicament for tumor treatment and precision treatment by carrying different genes of interest. Compared with the prior art, the hypoxia-triggered transcription control system of the present invention is efficiently and inducibly expressed under hypoxia, thereby improving the strictness and sensitivity of hypoxia triggering. In addition, with the help of the transcription control system of the present invention, the higher expression of genes of interest can be driven by using a small amount of transcription factors, thereby further amplifying its sensitivity to hypoxia.
The objects of the present invention are achieved by the following technical solutions.
In an aspect, the present invention provides a hypoxia-triggered artificial transcription factor (HATF), the HATF comprising:
In another aspect, the present invention further provides a nucleic acid sequence encoding the HATF.
In yet another aspect, the present invention provides a recognition element (RE) of the HATF, the RE comprising a core sequence selected from:
Preferably, the RE comprises a plurality of copies of the core sequence and a minimal promoter; more preferably, the plurality of copies are two copies, three copies, four copies, five copies, six copies, seven copies, eight copies, nine copies, or ten copies; and further preferably, the recognition element (RE) comprises five or six copies of the core sequence and a minimal promoter.
Still further preferably, the RE comprises a sequence selected from:
Most preferably, the RE is shown in SEQ ID NO: 3.
The HATF provided by the present invention is a novel artificial transcription factor. The inventors of the present invention have found that co-regulation by this HATF and its matching RE can increase the expression of a gene of interest by a factor of more than one hundred, which is far greater than the upregulation achieved by the conventional artificial transcription factor and its reaction element.
In another aspect, the present invention provides a hypoxia-triggered transcription control system. The transcription control system comprises a nucleic acid sequence encoding a HATF, and a RE.
The hypoxia-triggered transcription control system according to the present invention comprises two sets of transcription control units linked upstream and downstream, wherein the upstream transcription control unit comprises a hypoxia-triggered transcription reaction element (HRTE) for controlling the HATF, and a nucleic acid sequence encoding the HATF; and the downstream transcription control unit comprises an RE and a gene of interest (GOI).
The HATF is triggered by hypoxia during both RNA transcription and protein translation, that is, the hypoxia-triggered artificial transcription factor is not expressed or expressed at a low level under normoxia, but is efficiently and inducibly expressed under hypoxia, thereby improving the strictness and sensitivity of hypoxia triggering.
In the hypoxia-triggered transcription control system according to the present invention, the transcription control units linked upstream and downstream are nonlinearly connected in series, and located at two vectors; and preferably, the transcription control system has a combined form as shown in any one of the following formulas:
Alternatively, the transcription control units linked upstream and downstream are linearly connected in series, and located at the same vector; and preferably, the transcription control system has a combined form as shown in any one of the following formulas:
In the hypoxia-triggered transcription control system according to the present invention, the HRTE is selected from a flanking region of a hypoxia-inducible gene, such as a vascular endothelial growth factor gene, an erythropoietin gene or a glycolytic enzyme gene, and a core sequence of the HRTE is 5′-(A/G)CGT(G/C)-3′.
In the hypoxia-triggered transcription control system according to the present invention, the HRTE comprises a core sequence selected from:
Preferably, the HRTE includes a plurality of copies of the core sequence and a minimal promoter; more preferably, the plurality of copies are two copies, three copies, four copies, five copies, six copies, seven copies, eight copies, nine copies or ten copies; and
Still further preferably, the HRTE comprises a sequence selected from:
Most preferably, the HRTE is shown in SEQ ID NO: 5.
In a specific embodiment, the artificial transcription factor HATF according to the present invention has an amino acid sequence shown in SEQ ID NO: 1, and the recognition element RE of the HATF has a key nucleic acid sequence shown in SEQ ID NO: 3. Co-regulation by the HATF and the RE can increase the expression of a gene of interest (red fluorescent protein mCherry) by a factor of one hundred, which is much superior to the 10-fold up-regulation achieved by the conventional artificial transcription factor (GAL4) and its reaction element (UAS).
In the hypoxia-triggered transcription control system according to the present invention, the gene of interest GOI is a functional gene; and
Preferably, the corresponding antigen targets are broad-spectrum universal targets expressed on different cell membranes, including but not limited to: AXL, EGFR, MHC, CD24, CD47, FAP, CD147, HER-2, CD55, CD59, ROR1, ROR2, CD133, CD44v6, CD44v7, CD44v8, CD126, CD171, CEA, EpCAM, TAG72, IL-13Rα, EGFRvIII, GD2, GD3, FRα, PSCA, PSMA, GPC3, CAIX, Claudin18.2, VEGFR2, PD-L1, PD-L2, MSLN, MUC1, c-Met, FOLR1, B7-H3, Trop2, and the like; and
The present invention further provides a nucleic acid sequence, comprising the hypoxia-triggered transcription control system of the present invention; and
The present invention further provides a vector, comprising the nucleic acid sequence; preferably, the vector is selected from a plasmid, a retroviral vector, a lentiviral vector, an adenoviral vector, an adeno-associated viral vector, a vaccinia virus vector, a herpes simplex virus vector, a forest encephalitis virus vector, a poliovirus vector, a Newcastle disease virus vector, a transposon or a combination of one or more thereof; and more preferably, the vector is the lentiviral vector.
The present invention further provides a host cell. The host cell comprises the vector or the nucleic acid molecule is integrated into a chromosome of the host cell.
Preferably, the host cell is an isolated human-derived cell, including an embryonic stem cell, a umbilical cord blood-derived stem cell, an induced pluripotent stem cell, a hematopoietic stem cell, a mesenchymal stem cell, an adipose-derived stem cell, a T cell, an NK cell, an NKT cell or a macrophage; more preferably, the human-derived cell is a genetically engineered immune cell; further preferably, the genetically engineered immune cell is a T cell, an NK cell, an NKT cell or a macrophage; and still further preferably, the genetically engineered immune cell is a T cell.
Most preferably, the genetically engineered immune cell is selected from the group consisting of:
In a preferred embodiment, the genetically engineered immune cell is a CAR-T cell. The hypoxia-triggered transcription control system of the present invention is introduced into the CAR-T cell by transfection or transduction via the vector, thereby inducing the expression of a red fluorescent protein mCherry, a chimeric antigen receptor or a bispecific T cell engager in a hypoxic environment.
The present invention further provides a method for preparing a genetically engineered cell comprising the hypoxia-triggered transcription control system, wherein the method comprises the steps of: introducing a vector comprising the hypoxia-triggered transcription control system of the present invention into a host cell, thereby obtaining the genetically engineered cell.
In a preferred embodiment, the introduction may be performed simultaneously, in a chronological order, or in sequence.
The present invention further provides use of the hypoxia-triggered artificial transcription factor HATF, the recognition element RE of the HATE, the hypoxia-triggered transcription control system, the nucleic acid molecule, the vector and the host cell in the preparation of a medicament or a formulation for treating hypoxic diseases, ischemic diseases, or cancers.
Preferably, the cancer is a solid tumor; and more preferably, the solid tumor is one or more selected from neuroblastoma, lung cancer, breast cancer, esophageal cancer, gastric cancer, liver cancer, cervical cancer, ovarian cancer, kidney cancer, pancreatic cancer, nasopharyngeal cancer, small bowel cancer, large bowel cancer, colorectal cancer, bladder cancer, bone cancer, prostate cancer, thyroid cancer or brain cancer.
A method for treating hypoxic diseases, ischemic diseases or cancers comprises: administrating to a subject in need thereof a therapeutically effective amount of the hypoxia-triggered artificial transcription factor HATF, the recognition element RE of the HATF, the hypoxia-triggered transcription control system, the nucleic acid molecule, the vector, the host cell, a combination of the vector and the cell, or a combination of the above with other therapeutic drugs and technologies, including but not limited to a PD-1 antibody, a PD-L1 antibody, a CTLA-4 antibody, a TIGIT antibody, a Tim-3 antibody, and a LAG-3 antibody.
The present invention provides a hypoxia-triggered artificial control factor and a transcription control system comprising the same. The present invention further provides use of the hypoxia-triggered artificial control factor in the treatment of hypoxic diseases such as solid tumors, especially the use in CAR-T cell therapy of solid tumors.
Compared with the prior art, the present invention has the following advantages.
It should be understood that the above technical features of the present invention and the technical features specifically described below (e.g., in examples) may be combined with each other within the scope of the present invention, thereby constituting a new or preferred technical solution. Due to limited space, these technical features are not repeated one by one.
The embodiments of the present invention will be described in detail below with reference to the accompanying drawings. In drawings:
The following examples are used to illustrate the present invention, but not to limit the scope of the present invention.
Experimental methods used in the following examples were conventional experimental methods in the art unless otherwise specified. Experimental materials used in the following examples, unless otherwise specified, were purchased from conventional biochemical reagent sales companies, wherein:
DMEM medium and RPMI1640 medium were purchased from Corning, and lymphocyte medium X-VIVO 15 was purchased from Lonza.
A T cell growth medium, which was composed of a basal medium and cytokines, was prepared with reference to the Chinese invention patent CN201910163391.1. The basal medium was a lymphocyte medium X-VIVO 15, and the cytokines, i.e., 5 ng/mL IL-7, 10 ng/mL IL-15 and 30 ng/mL IL-21 were added. The cytokines IL-7 and IL-15 were purchased from R&D, and IL-21 was purchased from Nearshore Protein Technology Co., Ltd.
Fetal bovine serum was purchased from BI Inc.
A TurboFect Transfection Kit was purchased from Thermo Fisher Scientific.
A Lenti-X lentiviral concentrate reagent was purchased from Takara, Inc.
Gene synthesis was purchased from Shanghai Generay Bioengineering Co., Ltd.
A packaging plasmid psPAX2 and an envelope plasmid PMD2.G in blank lentiviral expression plasmids (pXW-EF1α-MCS-P2A-EGFP and pXW-EF1α-MCS) were purchased from Addgene, and pSV1.0-EGFP-ATR-RE, pSV1.0-EGFP-GAL4UAS, pSV1.0-HATF, pSV1.0-Tat and pSV1.0-GAL4-VP64 were purchased from Shanghai SINOBAY Biotechnology Co., Ltd.
Stable 3 chemically competent cells were purchased from Shanghai Weidi Biotechnology Co., Ltd.
An endotoxin-free plasmid miniprep kit and an endotoxin-free plasmid midiprep kit were purchased from OMEGA and Macherey Nagel, respectively.
A luciferase substrate was purchased from Promega Biotechnology Co., Ltd.
HEK293T cells, A549 lung cancer cells, SKOV3 ovarian cancer cells, and NCI-11292 lung cancer cells were purchased from ATCC in the United States. SKOV3-luc and NCI-H292-luc, which stably integrated firefly luciferase genes, were engineered by Shanghai SINOBAY Biotechnology Co., Ltd.
Severe combined immuno-deficient mice (B-NDG) were purchased from Biocytogen (Jiangsu) Gene Biotechnology Co., Ltd.
Replication-deficient adenovirus type 5 (HER2-Luc-Ad) loaded with a human-derived HER2 gene and firefly luciferase was purchased from GeneChem.
Nucleic acid sequences shown in SEQ ID NOs: 2-5 and 9 were synthesized by Shanghai Generay Bioengineering Co., Ltd. and cloned into blank lentiviral expression plasmids (pXW-EF1α-MCS-P2A-EGFP and pXW-EF1α-MCS) to obtain the following recombinant lentiviral expression plasmids:
HEK293T cell treatment: 24 h before transfection, HEK293T cells in the logarithmic growth phase were collected, and inoculated in a 10 cm cell culture dish (6×106 to 8×106 cells); and the cells grew in 10 mL of complete DMEM medium, and cultured in a 37° C., 5% CO2 cell incubator for 18-24 h, followed by plasma transfection till the cell density reached 70-90%.
HEK293T cell transfection: 1 mL of basal DMEM medium was added to a 15 mL centrifuge tube, and a transfected mixed solution was prepared according to a mass ratio of lentiviral expression plasmids to packaging plasmids to envelope plasmids of 1:3:1, with a total plasmid amount of 15 μg/dish. 30 μL of TurboFect transfection reagent was added according to a ratio of plasmids (μg) to a transfection reagent (μL) of 1:2, incubated at room temperature for 15-20 min, then added to a dish plated with HEK293T cells, and continued to be cultured in a 37° C., 5% CO2 cell incubator for 48 h; a virus supernatant was then collected, and centrifuged at 1000×g, 4° C. for 10 min; precipitates at the bottom of the tube were discarded; and a virus supernatant was collected.
A 0.45 μm filter was used to further filter the virus supernatant collected by centrifugation, added with a Lenti-X lentiviral concentration reagent in ⅓ of the volume of the virus supernatant, inverted and mixed uniformly several times, incubated at 4° C. overnight, and centrifuged at 2000×g, 4° C. for 45 min, wherein a white precipitate was seen at the bottom of the centrifuge tube, which is concentrated virus particles. The supernatant was discarded carefully, and the white precipitate was resuspended with a blank RPMI1640 medium in 1/50- 1/100 of the volume of the original virus supernatant, aliquoted and cryopreserved at −80° C. for later use.
Jurkat T cells were inoculated at 1×105/well on a 96-well U-bottom plate, and the collected lentiviral concentrate was diluted in 10-fold increments. 100 μL of virus diluent was added to the corresponding wells, added with an infection promoting agent, i.e., protamine sulfate, to adjust the concentration to 10 μg/mL, centrifuged at 1000×g, 32° C. and infected for 90 min, cultured overnight followed by replacement with a fresh RPMI1640 complete medium, and continued to be cultured for 48 h; and the proportion of fluorescence-positive cells was detected by a flow cytometry. The virus titer was calculated using the following formula:
Virus titer (TU/mL)=1×105×proportion of fluorescence-positive cells/100×1000×corresponding dilution factor.
The following lentiviral vectors obtained by concentration:
Lentiviral vectors LV-EF1α-GFP-HATF-HRTE-RE-BiTE (MOI=3) containing 2-6 copies of RE, which were obtained by concentration, were respectively added to a 6-well flat-bottom plate plated with 3×105 A549 cells on alternate days, added with an infection promoting agent, i.e., polybrene, to adjust the working concentration to 10 μg/mL, cultured overnight followed by replacement with a fresh complete medium R10 (RPMI1640+10% FBS+1% PS), and continued to be cultured. After the cells grew to 80% coverage, they were expanded and cultured for later use.
In order to verify that the HATF of the present invention had a better ability to induce the expression of exogenous genes than the conventional GAL4-VP64 artificial transcription factor, plasmid transfection experiments were performed for HEK293T cells. The HEK293T cells were plated in a 48-well plate at 5×104 cells/well and transfected the next day after cell attachment. Only 0.25 sg of reaction element plasmid pSV1.0-EGFP-ATR-RE or pSV1.0-EGFP-GAL4UAS which was transfected in a single transfection group; 0.25 μg of pSV1.0-EGFP-RE plasmid and pSV1.0-HATF or pSV1.0-Tat, or 0.25 μg of pSV1.0-EGFP-GAL4UAS plasmid and pSV1.0-GAL4-VP64 which were transfected in an experimental group, were added with a TurboFect transfection reagent in a ratio of plasmids (μg):transfection reagent (μL) of 1:2, incubated at room temperature for 15-20 min, added to a cell culture plate, and cultured in a 37° C., 5% CO2 cell incubator for 48 h; and the expression of a gene of interest mCherry was then detected by flow cytometry.
As shown in
Three different T cells integrating EF1α-GFP-RE-mCherry, EF1α-BFP-HATF-HRTE and EF1α-GFP-RE-mCherry, and EF1α-GFP-HATF-HRTE-RE-mCherry genes were prepared by using the lentiviral expression plasmid in Example 1, the lentiviral vector preparation method in Example 2 and the method in Example 3, which were a RE-mCherry reaction element, a hypoxia-sensitive transcription control system in a binary unit form (co-infected by dual vectors) and a hypoxia-sensitive transcription control system in a unary unit form (injected by a single vector). The above three genetically engineered T cells were cultured under a normoxic condition (21% O2) and a hypoxic condition (1% O2) for 24 h; the cells were collected after the culture and eluted with FACS buffer (1×PBS of 2% FBS) once, centrifuged at 500×g for 5 min, then resuspended and mixed uniformly with 300 μL of FACS buffer after the centrifugation; and then, the expression of the red fluorescent protein mCherry was detected by flow cytometer.
The results were shown in
Two engineered T cells carrying EF1α-HER2 CAR-P2A-EGFP and EF1α-GFP-HATF-HRTE-RE-CAR genes, named HER2 CAR-T cells (positive control) and hypoxia-sensitive HER2 CAR-T cells respectively, were prepared using the lentiviral expression plasmid in Example 1, the lentiviral vector preparation method in Example 2 and the method in Example 3. The above two genetically engineered T cells were cultured under a normoxic condition (21% O2) or a hypoxic condition (1% O2) for 24 h respectively; the cells were collected after the culture and eluted with FACS buffer once, added with 2 μg/mL flow cytometry antibody PE-anti-DYKDDDDK, incubated at room temperature in the dark for 20 min, eluted twice with FACS buffer after the incubation, and resuspended and mixed uniformly with 300 μL of FACS buffer; and then, the expression of HER2 CAR molecules was detected by flow cytometer.
The results were shown in
The tumor cell killing efficiency was evaluated by a luciferase-based cytotoxicity assay. First, 1×104 SKOV3-Luc (firefly luciferase gene-modified human ovarian cancer cells) or NCI-H292-Luc (firefly luciferase gene-modified human lung cancer cells) were inoculated on a 96-well flat-bottom black plate with 100 μL of medium per well, and cultured in a 37° C., 5% CO2 cell incubator for 18 h. On the second day, the genetically engineered T cells in Example 6 and non-transduced T cells cultured at the same time were added to the wells containing target cells at ratios of effector cells to target cells of 1:2, 1:1 and 2:1, and were cultured under a normoxic condition (21% O2) or a hypoxic condition (1% O2) for 24 h, respectively; and the luciferase activity value of the target cells was detected by a GloMax®96 microplate luminescence detector after co-culture.
The formula for calculating the cell kill rate was as follows:
The results were shown in
The results were shown in
Local hair removal on the back of B-NDG mice raised in a sterile isolator was performed with a hair removal cream or an animal shaver one day in advance, such that the skin of a tumor cell inoculation site was exposed.
Each mouse was fixed with the left hand, that is, the head, neck and back skin of the mouse were grasped with the left hand at the same time; and after a part to be hair-removed on the right side of the back was fully exposed by turning its back to the left, this part was disinfected by an alcohol cotton ball with the right hand. A 1 mL insulin syringe was used to blow and mix uniformly the pre-prepared SKOV3 or NCI-H292 tumor cells, and then suck 125 μL of cell suspension (5×106 tumor cells), and a tip of a needle was pierced subcutaneously and diagonally into the mouse at an angle of 30°-40° from the skin; and the cell suspension was slowly injected to avoid cell spillage. After the injection of 125 μL of cell suspension was completed, the needle was left for 2-3 seconds and then quickly withdrawn, and a clearly visible bulge could be seen under the skin at the injection site.
After cell inoculation, the tumorigenesis and health status of the mice were observed every 2-3 days, and the baseline tumor volume was measured with a vernier caliper after tumorigenesis, and subsequent experiments were carried out.
5×106 hypoxia-sensitive HER2 CAR-T cells and control cells were intravenously infused, the tumor size was measured every 2-3 days, and a long diameter and a short diameter of the tumor were measured with a vernier caliper, respectively; and the tumor volume was calculated according to the following formula: volume=(long diameter×short diameter2)/2.
The results were shown in
Genetically engineered A549 cells in Example 4 were plated in a 12-well flat-bottom plate one day in advance, with 3×105 cells per well and a total volume of 1 mL.
On the second day, the plate was cultured under a normoxic condition (21% O2) or a hypoxic condition (1% O2) for 24 h; the cells were collected after culture, resuspended with 1×BS, diluted into 1× with 4×SDS Loading Buffer, and fully blown and mixed uniformly; and the sample was placed in a pot, and heated and boiled with an induction cooker for 10 min to fully denature the protein.
According to the molecular weight (72.5 kD) of the hypoxia-sensitive artificial transcription factor and the molecular weight (54.7 kD) of BiTE of a protein of interest, upper and lower layers of SDS-PAGE gel with a concentration of 10% were prepared, and the sample was separated by electrophoresis in the SDS-PAGE gel for 2 h; and then the sample was transferred from the gel to a PVDF membrane, which is activated by methanol in advance, by means of wet transfer.
According to the instructions of a protein Marker, the PVDF membrane containing the protein of interest and a reference protein was cut and placed in a 5% skimmed milk powder blocking solution, and incubated on a shaker for at least 1 h at room temperature.
The blocked PVDF membrane was taken out and placed in a mouse anti-His-tag primary antibody diluent, with a dilution ratio of primary antibody of 1:1000, and placed on a shaker at 4° C. overnight.
The primary antibody diluent was recovered and cryopreserved at −20° C. for later use. The PVDF membrane was taken out, and oscillated and eluted with PBST on the shaker for three times for 8 min each time, and a corresponding goat anti-mouse IgG-HRP secondary antibody diluent, with a dilution ratio of secondary antibody of 1:3000, was added after elution was completed, and incubated on the shaker at room temperature for 1 h.
After the PVDF membrane was taken out, it was eluted with PBST for three times, and then exposed after color development.
The results were shown in
The tumor cell killing efficiency detection method and the killing efficiency calculation were the same as those in Example 7. First, 1×104 SKOV3-Luc (firefly luciferase gene-modified human ovarian cancer cells) or NCI-H292-Luc (firefly luciferase gene-modified human lung cancer cells) were inoculated on a 96-well flat-bottom black plate with 100 μL of medium per well, and cultured in a 37° C., 5% CO2 cell incubator for 18 h. On the second day, the hypoxia-sensitive CD47/CD3-BiTE-T cells in Example 8 and non-transduced T cells cultured at the same time were added to the wells containing target cells at ratios of effector cells to target cells of 1:2, 1:1, and 2:1, and were cultured under a normoxic condition (21% O2) or a hypoxic condition (1% O2) for 20 h, respectively; and the luciferase activity value of the target cells was detected by a GloMax®96 microplate luminescence detector after co-culture.
The results were shown in
First, 5-106 NCI-H292-Luc (human lung cancer cells modified by firefly luciferase gene) were inoculated subcutaneously on the right side of the back of B-NDG mice, and each injected with 125 μL of tumor cell suspension; hypoxia-sensitive BiTE-T cells prepared in Example 3 and non-transduced T cells cultured at the same time were injected intravenously or intratumorally day 6 after tumor inoculation; and 5×106 positive cells were infused into each tumor-bearing mouse. After the tumor cells were inoculated, a long diameter and a short diameter of the tumor were measured by a vernier caliper, and the tumor volume was calculated according to the following formula: tumor volume=(long diameter×short diameter2)/2.
The results were shown in
The above examples are exemplary, and should not be construed as limiting the present invention. A person of ordinary skill in the art can make changes, modifications, substitutions and variations to the above examples within the scope of the present invention.
Number | Date | Country | Kind |
---|---|---|---|
202110930268.5 | Aug 2021 | CN | national |
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/CN2021/141636 | 12/27/2021 | WO |