diff --git a/index.html b/index.html index f7c6dd0..dab6d95 100644 --- a/index.html +++ b/index.html @@ -1 +1,326 @@ -11111 + + + + +
+ + + + +Directed evolution, a laboratory simulation of Darwinian natural selection, employs random mutagenesis and recombination coupled with human-designed selection pressures to sift through proteins for those exhibiting desired characteristics, thereby facilitating the artificial evolution of molecules and cells.
Drawing inspiration from the traditional Chinese 'Sword Tomb', this study introduces a novel evolutionary strategy to refine the Natural Killer (NK) cell receptor repertoire, akin to selecting the most fitting sword for each battle against tumor, to counteract the diverse and elusive nature of tumor cells.
1. Project: Team member recruitment
2. Time: 2024.03.11-2024.03.16
3. Member: Jianhua Luo
4. Result: We selected 7 students out of 60 as regular players. They are Song Zhang, Yaqi Gao, Xinyu Zhu, Xiaoyuan Chen, Yinran Luo, Hanyue Liu, Xudong Tang. Moreover, we choose Yaqi Gao and Xudong Tang as the delegates to participate the iDEC festival in Cambridge University.
1. Experiment: The introduction of the cell therapy background
2. Time: 2024.04.01-2024.04.06
3. Member: Song Zhang, Yaqi Gao, Xinyu Zhu, Xiaoyuan Chen, Yinran Luo, Hanyue Liu, Xudong Tang
4. Summary: Anti-CD19 CAR-T cells have made remarkable achievements in the research of hematological malignancies. In addition to CAR-T, many other immune cells such as NK cells are also ideal choices for the treatment of tumors.
5. Attachment: The introduction of the cell therapy background
1. Experiment: Lecture research and report
2. Time: 2024.04.07-2024.04.27
3. Member: Song Zhang, Yaqi Gao, Xinyu Zhu, Xiaoyuan Chen, Yinran Luo, Hanyue Liu, Xudong Tang
4. Summary: We learned three lectures and made report in the team meeting. After that each player summarized the lecture they read.
5. Attachment: Apr. 02 Lecture research and report
1. Experiment: Experiment technique learning
2. Time: 2024.04.08-2024.04.22
3. Member: Song Zhang, Yaqi Gao, Xinyu Zhu, Xiaoyuan Chen, Yinran Luo, Hanyue Liu, Xudong Tang
4. Summary: We asked players learn the project relevant experiment technique and made summary. They were Construction of the plasmids of CAR, lentivirus packaging and NK-92 expansion.
5. Attachment: Apr. 03 Experiment technique learning
1. Experiment: Barcode designing
2. Time: 2024.04.23-2024.04.30
3. Member: Song Zhang, Yaqi Gao
4. Summary: We designed the barcode sequence with Python. There are five parts of the barcode sequence, and they are the forward primer, the reverse primer, a TaqMan Probe and two spacer sequences.
5. Attachment: Apr. 04 Barcode designing
1. Experiment: 23 sequences design
2. Time: 2024.05.01-2024.05.03
3. Member: Yaqi Gao, Song Zhang, Xudong Tang
4. Summary: We designed two types of specific ICD CAR sequences. They were the tandem IL2R-ICD CAR and Charge-attracting IL2R-ICD CAR.
5. Attachment: May. 01 23 sequences design
1. Experiment: Construction of the plasmids for CAR
2. Time: 2024.05.03-2024.05.05
3. Member: Yaqi Gao, Song Zhang, Xudong Tang, Xinyu Zhu
4. Summary: The 23 sequences were constructed and purified using the Rapid DNA Ligation Kit (Genstar), NucleoBond Xtra Midi Maxi EF plasmid purification kit (Macherey-Nagel 740420.10) to lay the foundation for subsequent CAR-NK construction
5. Attachment:May. 02 Construction of the plasmids for CAR
1. Experiment: Lentivirus Package
2. Time: 2024.05.06-2024.05.09
3. Member: Xinyu Zhu, Xudong Tang, Song Zhang
4. Summary: We aimed to produce lentivirus for subsequent gene transduction. Lentiviral plasmids were transfected into Lenti-293T cells using the calcium phosphate method, and virus supernatants were collected, concentrated, and purified for storage and future use.
5. Attachment: May. 03 Lentivirus Package
1. Experiment: Infection of NK92 cell line
2. Time: 2024.05.10-2024.05.11
3. Member: Hanyue Liu, Xudong Tang, Xiaoyuan Chen
4. Summary: We aimed to prepare NK92 cells at 1×107 cells/mL, plate them, and infect them with the virus using polybrene, followed by incubation at 37°C for 12-16 hrs.
5. Attachment: May. 04 Infection of NK92 cell line
1. Experiment: NK92 expansion and flow cytometry sorting
2. Time: 2024.05.12-2024.05.14
3. Member: Xiaoyuan Chen, Yinran Luo, Xudong Tang
4. Summary: We aimed to culture NK92 cells, passage them, and label with FLAG-APC antibody. Cells were sorted based on CAR expression, expanded, and analyzed by flow cytometry.
5. Attachment: May. 05 NK92 expansion and flow cytometry sorting
1. Experiment: Secondary flow cytometry sorting
2. Time: 2024.05.21-2024.05.22
3. Member: Xiaoyuan Chen, Yinran Luo, Song Zhang, Xinyu Zhu
4. Summary: We aimed to perform a second flow cytometry sorting on NK92 cells for further purification, following initial expansion and FLAG-APC antibody labeling.
5. Attachment: May. 06 Secondary flow cytometry sorting
1. Experiment: NK cells co-cultured with tumor cells
2. Time: 2024.05.21-2024.05.28
3. Member: Xiaoyuan Chen, Xinyu Zhu, Yaqi Gao
4. Summary: We planned to co-culture NK92 cells with tumor cells, divided into three groups (A, B, C) for comparison, and assess NK92 expansion by photographing and analyzing EGFP fluorescence.
5. Attachment: May. 07 NK cells co-cultured with tumor cells
1. Experiment: Cell proliferation assessment
2. Time: 2024.05.10-2024.05.29
3. Member: Xinyu Zhu, Song Zhang, Yinran Luo
4. Summary: We monitored cell growth by collecting samples daily (Day 1-Day 7), counting cells by an automated cell counter, calculating fold expansion, and plotting a growth curve to analyze proliferation and kinetics.
5. Attachment: May. 08 Cell proliferation assessment
1. Experiment: Enrichment and screening of dominant sequences
2. Time: 2024.05.28-2024.05.29
3. Member: Xinyu Zhu, Hanyue Liu, Xiaoyuan Chen
4. Summary: We performed PCR on samples from Experiment Record 7 using pre-designed barcodes to assess CAR-NK cell expansion, and then create diagrams for comparative analysis with the results from Record 7.
5. Attachment: May. 09 Enrichment and screening of dominant sequences
1. Experiment: LDH assay for measuring cytotoxic activity
2. Time: 2024.05.26-2024.05.31
3. Member: Xinyu Zhu, Yaqi Gao, Xiaoyuan Chen
4. Summary: We used an LDH assay kit to measure the cytotoxicity of CAR-NK cells against various tumor cells at different effector-to-target.
5. Attachment: May. 10 LDH assay for measuring cytotoxic activity
1. Experiment: Western Blot
2. Time: 2024.06.01-2024.06.03
3. Member: Hanyue Liu, Xudong Tang, Yaqi Gao
4. Summary: Western blot is a technique for detecting specific proteins in a sample, involving separation by gel electrophoresis, transfer to a membrane, and identification using antibody probes.
5. Attachment: Jun. 01 Western Blot
1. Experiment: LDH assay
2. Time: 2024.06.04-2024.06.06
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: The LDH assay was used for quantifying cytotoxic activity by measuring the levels of lactate dehydrogenase enzyme released from intact cells into the culture medium upon cell membrane disruption, indicating cell death or damage.
5. Attachment: Jun. 02 LDH assay
1. Experiment: Investigation of NKR
2. Time: 2024.06.07-2024.06.09
3. Member: Song Zhang, Yaqi Gao, Xinyu Zhu, Yinran Luo, Xudong Tang, Xiaoyuan Chen, Hanyue Liu
4. Summary: We investigated NK cell receptors by reviewing literature and online resources to identify their corresponding ligands, clinical batch numbers, applications, and significance, providing a comprehensive understanding of NK cell receptor diversity and function.
5. Attachment: Jun. 03 Investigation of NKR
1. Experiment: Literature review of tumor ligands
2. Time: 2024.06.10-2024.06.12
3. Member: Yinran Luo, Song Zhang, Yaqi Gao, Xinyu Zhu
4. Summary: The process involves retrieving the expression levels of ligands for KIR and KAR in various cancer cell lines using target classification on the Protein Atlas website for comprehensive analysis.
5. Attachment: Jun. 04 Literature review of tumor ligands
1. Experiment: NKR sequence design
2. Time: 2024.06.13-2024.06.15
3. Member: Yaqi Gao, Song Zhang
4. Summary: This experiment involves applying the intracellular sequence from the first screening round to the second, modifying the NKR sequence, and constructing a new library.
5. Attachment: Jun. 05 NKR sequence design
1. Experiment: Construction of the plasmids for CAR
2. Time: 2024.06.16-2024.06.18
3. Member: Xiaoyuan Chen, Xudong Tang, Hanyue Liu
4. Summary: This experiment involves CAR plasmids by cloning NKR fragments into pRRLSIN-cPPT-PGK-GFP WPRE plasmids, digesting with restriction enzymes, ligating with T4 ligase, transforming into Stbl3, and purifying plasmids after sequencing verification.
5. Attachment: Jun. 06 Construction of the plasmids for CAR
1. Experiment: Lentivirus Package
2. Time: 2024.06.19-2024.06.20
3. Member: Song Zhang, Xinyu Zhu, Hanyue Liu
4. Summary: Lentivirus packaging involves culturing Lenti-293T cells, preparing a plasmid mixture, and transfecting with calcium phosphate using the CPT Transfection Kit, followed by incubation in a CO2 incubator to facilitate virus production.
5. Attachment: Jun. 07 Lentivirus Package
1. Experiment: NK92 cell infection
2. Time: 2024.06.21-2024.06.22
3. Member: Hanyue Liu, Yaqi Gao
4. Summary: The experiment involves inoculating NK92 cells, calculating and adding the required cell suspension to a 96-well plate, incubating to 50% confluence, then infecting with virus using polybrene to enhance infection, adjusting for MOI and virus titer.
5. Attachment: Jun. 08 NK92 cell infection
1. Experiment: Cell expansion and Flow cytometry sorting
2. Time: 2024.06.23-2024.06.24
3. Member: Song Zhang, Xudong Tang, Hanyue Liu
4. Summary: NK92 cells suspension from the stock is transferred to a tissue culture flask. Then it is incubated to expand. After the expansion, flow cytometry is used to sort the cells.
5. Attachment: Jun. 09 Cell expansion and Flow cytometry sorting
1. Experiment: Second round of flow cytometry sorting
2. Time: 2024.06.25-2024.06.26
3. Member: Song Zhang, Xudong Tang, Hanyue Liu
4. Summary: NK92 cells suspension from the stock is transferred to a tissue culture flask. Then it is incubated to expand. After the expansion, flow cytometry is used to sort the cells.
5. Attachment: Jun. 10 Second round of flow cytometry sorting
1. Experiment: Detection of the NKR
2. Time: 2024.06.27-2024.06.29
3. Member: Song Zhang, Yaqi Gao, Xudong Tang, Hanyue Liu
4. Summary: After the process of the co-culture, we detected the scFvs (including KIRs and KARs) on the NK cells’ surface.
5. Attachment: Jun. 11 Detection of the NKR
1. Experiment: Detection of the NKR
2. Time: 2024.06.29-2024.06.30
3. Member: Song Zhang, Yaqi Gao, Xudong Tang, Hanyue Liu
4. Summary: After the process of the co-culture, we detected the scfvs (including KIRs and KARs) on the NK cells’ surface.
5. Attachment: Jun. 12 Detection of the NKR
1. Experiment: The cytotoxic activity detected by LDH
2. Time: 2024.07.01-2024.07.07
3. Member: Song Zhang, Xudong Tang, Hanyue Liu, Xiaoyuan Chen, Xinyu Zhu, Yinran Luo
4. Summary: We detected the cytotoxic activity of NK cells which after two rounds of evolution by LDH, targeting at four kinds of cell lines.
5. Attachment: Jul. 01 The cytotoxic activity detected by LDH
1. Experiment: Activating signal pathway detected by Western Blotting
2. Time: 2024.07.08-2024.07.14
3. Member: Song Zhang, Xudong Tang, Hanyue Liu, Xiaoyuan Chen, Xinyu Zhu, Yinran Luo, Yaqi Gao
4. Summary: NK92 cells transduced with αMSLN eCAR sequence exhibited significant higher expression of p-JAK1, p-JAK3, p-ERK, p-STAT1, p-STAT3, p-STAT5, p-AKT.
5. Attachment: Jul. 02 Detect the activating signal pathway by Western Blotting
1. Experiment: Surface molecules detected by the flow cytometry
2. Time: 2024.07.15-2024.07.21
3. Member: Song Zhang, Xudong Tang, Hanyue Liu, Xiaoyuan Chen, Xinyu Zhu, Yinran Luo, Yaqi Gao
4. Summary: We used flow cytometry to detect the molecules on the cell’s surface.
5. Attachment: Jul.03 Surface molecules detected by the flow cytometry
1. Experiment: Human pancreatic tumor organoids culture and passage
2. Time: 2024.08.01-2024.08.09
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: The experiment aims to culture and passage human pancreatic tumor organoids. It involves processing tumor tissue samples, digestion, centrifugation, seeding in plates with Matrigel, monitoring growth, and subculturing the organoids at approximately 70% confluence.
5. Attachment: Aug.01 Human pancreatic tumor organoids culture and passage
1. Experiment: Co-culture of NK92 and human pancreatic tumor organoids
2. Time: 2024.08.10-2024.08.13
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: This experiment involves co-culturing NK-92 cells with human pancreatic tumor organoids to assess cytotoxicity, using a 48-well plate with a Matrigel layer and culture medium supplemented with Y-27632 and PGE2 for optimal growth conditions.
5. Attachment: Aug.02 Co-culture of NK92 and human pancreatic tumor organoids
1. Experiment: Assess human pancreatic tumor organoid lysis
2. Time: 2024.08.14-2024.08.16
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: The experiment aims to assess the damage of human pancreatic tumor organoids using LDH Detection Reagent. It involves preparing LDH assay working solution, setting up controls, treating cells with test compounds, collecting samples, and measuring LDH activity to determine cytotoxicity.
5. Attachment: Aug.03 Assess human pancreatic tumor organoid lysis
1. Experiment: Human hepatoma organoids culture and passage
2. Time: 2024.08.17-2024.08.20
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: The experiment details the culturing and passaging of human hepatoma organoids, including preparation of various culture media, tissue digestion, seeding in Matrigel, incubation, and passaging procedures when organoids reach approximately 70% density.
5. Attachment: Aug.04 Human hepatoma organoids culture and passage
1. Experiment: Co-culture of NK92 and human hepatoma organoids
2. Time: 2024.08.21-2024.08.25
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: The main content of the experiment is the co-culture of NK92 cells with human hepatoma organoids to assess their cytotoxicity effect of NK cells.
5. Attachment: Aug.05 Co-culture of NK92 and human hepatoma organoids
1. Experiment: Human hepatoma organoid lysis detection
2. Time: 2024.08.25-2024.08.31
3. Member: Xiaoyuan Chen, Hanyue Liu
4. Summary: The experiment evaluates human hepatoma organoid damage utilizing LDH detection reagent by monitoring LDH release from damaged cells.
5. Attachment: Aug.06 Human hepatoma organoid lysis detection
1. Experiment: Construction of in situ tumor models
2. Time: 2024.09.01-2024.09.09
3. Member: Hanyue Liu, Yinran Luo
4. Summary:Pancreatic cancer cells AsPC1 were injected intraperitoneally into each mouse to construct in situ tumor models.
5. Attachment: Sep.01 Construction of in situ tumor models
1. Experiment: Construction of in situ hepatic tumor models
2. Time: 2024.09.10-2024.09.15
3. Member: Xinyu Zhu, Song Zhang
4. Summary: To evaluate the impact of inhibitory signal-activated CAR-NK cells on in vivo anti-tumor activity, an in-situ tumor-bearing mouse model of liver cancer was established by inoculating Hep G2 cells into NCG mice.
5. Attachment: Sep.02 Construction of in situ hepatic tumor models
1. Experiment: Adoptive cell transfer therapy targeting pancreatic cancer
2. Time: 2024.09.15-2024.09.20
3. Member: Yaqi Gao, Xiaoyuan Chen
4. Summary: NK92 cells and KARs/KIRs CAR-NK cells were delivered intravenously to the Model mice, pre-inoculated with luciferase-expressing HepG2 cells subcutaneously.
5. Attachment: Sep. 03 Adoptive cell transfer therapy targeting pancreatic cancer
1. Experiment: Adoptive cell transfer therapy targeting liver cancer cells
2. Time: 2024.09.21-2024.09.25
3. Member: Song Zhang, Xudong Tang
4. Summary: NK92 cells and KARs/KIRs CAR-NK cells were delivered intravenously to the Model mice, pre-inoculated with luciferase-expressing HepG2 cells subcutaneously.
5. Attachment: Sep.04 Adoptive cell transfer therapy targeting liver cancer cells
1. Experiment: Tumor volume detection
2. Time: 2024.09.25-2024.09.30
3. Member: Yinran Luo, Xinyu Zhu
4. Summary: Tumor diameters were recorded using a digital vernier caliper every 3 days.
5. Attachment: Sep. 05 Tumor volume detection
In local hospitals, we approached the bedside to communicate and interact with patients, introducing the concept of cell therapy. We used cartoon images to present the complex CAR-T therapy to patients. For the patients who were interested in it, we additionally talked about CAR-NK therapy based on our research projects, extremely broadening their understanding of cellular therapies. Although patients had different opinions on CAR-T, most of which believed that cell therapy could provide more help for clinical treatment in the near future. And for us, we successfully let more people know about cell therapy, illuminating beacons of hope for more lives.
With the advancement of medical research, an increasing number of experiments rely on animal studies for pre-clinical research. Therefore, promoting innovation in experimental methods through new technologies is crucial to reducing the use of animals in research. We aim to share our application of synthetic biology in the context of animal experiments, as well as provide some educational insights on the topic.
Hanyue Liu of our team elucidated the principles of directed evolution to the assembly of biomedical researchers, highlighting its role as a sophisticated strategy that emulates natural selection to facilitate the controlled evolution of biomolecules within a laboratory setting. This innovative approach holds the potential to enhance pharmacological interventions and to broaden our repertoire of therapeutic strategies, particularly in the realm of autoimmune disorders. By harnessing directed evolution, we stand on the cusp of creating more targeted and secure treatment modalities that could usher in a new era of hope for those afflicted.
For those developing targeted drugs for solid tumors, we described the concept and basic workflow of directed mutagenesis. Through a deep understanding of directed mutagenesis, drug developers may design and optimize drug structures more accurately. The heterogeneous tumor micro-environment restricted the efficacy of drugs in the treatment of solid tumors. Directed mutagenesis provides a way to enhance drug adaptability and efficacy, enabling flexible structural modifications.
During visits to university and hospital neighborhoods, we crafted educational materials based on our CAR-NK therapy research, aiming to spread knowledge on its cancer treatment applications. With the support of the residents' committees, we held lectures and Q&As, engaging audiences across ages. During these events, we elaborated on the fundamental principles and advantages of CAR-NK therapy, highlighting its promising direction for future cancer immunotherapy. This community service reinforced our commitment to advancing CAR-NK, confident it will offer hope to more cancer sufferers.
1. Innovative eCAR Technology: The novel chimeric receptor, eCAR, through charge-induced oligomerization enhances lymphocyte proliferation and activation, offering a promising approach for cancer immunotherapy.
2. Enhanced NK Cell Effectiveness: By integrating an NKR library within the eCAR, the engineered NK cells exhibit ligand-dependent cytotoxicity, effectively targeting a wide range of tumor cells and tumor-derived organoids.
3. Broad-Spectrum Cancer Immunotherapy: This study introduces a new strategy for NK cell-based therapy, utilizing an NKR library as a sensor to achieve targeted and efficient destruction of tumor cells across various malignancies.
Developing CAR Constructs for Tumor Antigen-Dependent Proliferation via Directed Evolution
We engineered two classes of CARs configurations targeting CD19, totaling 23 distinct constructs: the Tandem IL2R-ICD CARs incorporate the intracellular domains (ICDs) of the β and/or γ chains serially linked to CD3ζ; the Charge-attracting IL2R-ICD CARs feature a positively charged transmembrane region connecting 4-1BB and CD3ζ, and a negatively charged transmembrane region linked to the IL2R ICD (Fig. 1a). All CARs are coupled with enhanced green fluorescent protein (EGFP) via an internal ribosome entry site (IRES) to identification (Extended Data Fig. 1). These CAR gene fragments were transduced into NK-92 cells using lentivirus, with a second-generation CD19-targeting CAR-NK (BBζ) serving as a positive control (Fig. 1b). Under IL-2 supplemented culturing conditions, NK92, αCD19 NK cells (termed αCD19), and the αCD19 CAR library exhibited comparable proliferation rates. However, in the absence of IL-2 and upon stimulation by Raji cells (CD19 positive), only the αCD19 CAR library demonstrated significant expansion (Fig. 1c-d). The results indicates that the αCD19 CAR library can robustly proliferate in an IL-2 independent manner, responding exclusively to tumor antigen signals.
To pinpoint the specific sequences within the library that facilitate enhanced NK cell proliferation solely in response to tumor antigens, we repeated stimulate NK cells with Raji cells in an IL-2-deprived environment (Extended Data Fig. 2a-c). Post-stimulation, EGFP-positive NK cells were sorted and subjected to single-cell cloning. Subsequent stimulations with Raji cells led to significant enrichment of the constructs 46-BBζ/Fcγ/3β, 46-BBζ/Fcβ/3γ, and 44-BBζ/12β/12γ in comparison to others (Fig. 1e). These results demonstrate that these specific constructs are exceptionally effective in driving robust NK cell proliferation in response to tumor antigen stimulation, independent of exogenous IL-2. These constructs have been designated as eCAR1, eCAR2, and eCAR3, respectively.
Fig. 1 | Design and verification of the synthetic CAR NK-92 cell library. a, Map of lentiviral constructs encoding the IL2R-ICD CAR library. b, Membrane-bound αCD19 CAR-library expression. At 72 h after retroviral transduction, the expression of αCD19 CAR-library on human NK-92 cells was measured by staining with anti-FLAG antibody, followed by flow cytometry analysis. NK-92 cells without transduction were used as negative controls. The histograms shown in grey correspond to the isotype controls and the blue histograms indicate positive fluorescence of αCD19 CAR, whereas the red histograms indicate positive fluorescence of αCD19 CAR-library. Data are representative of at least three independent experiments. c-d, Proliferation activity of CAR-NK cells in response to IL-2 and Raji stimulation (without IL-2). The purple arrow indicates the addition of an equal volume of fresh medium containing 100 IU/mL IL-2. The red arrow indicates the addition of an equal volume of fresh medium without IL-2, containing the same number of non-proliferative Raji cells (***P < 0.001 compared with NK-92). Data are presented as mean ± SD of six independent biological replicates. e, Growth dynamics of the αCD19 CAR-library in the periodical stimulation co-culture. Frequencies of αCD19 CAR-NK were analyzed before the addition of fresh target cells.
Antigen-Specific Proliferation of eCAR-NK in an IL-2- deprived Environment
We further explored the capacity of eCARs to sustain extensive proliferation in an IL-2 independent environment, relying exclusively on tumor antigen signals. To this end, we engineered CAR-NK cells to target specific antigens associated with different malignancies-CD19 and CD20 for hematological cancers, GPC3 for hepatocellular carcinoma, and MSLN for pancreatic cancer, each can be sensed by corresponding eCAR (Fig. 2a). Experimental results revealed that both αCD19 eCAR1 and αCD19 eCAR2 exhibited robust proliferation in response to stimulation by Raji cells, even in the absence of IL-2 (Fig. 2b). In contrast, coculture with CD19-negative tumor cells (K562) without IL-2 supplementation resulted in no proliferation of both αCD19 eCAR1 and αCD19 eCAR2 (Fig. 2c). These observations affirm that eCAR1 and eCAR2 are capable of extensive proliferation driven solely by tumor antigen signals, independent of IL-2.
Given that the proliferative capabilities of αCD19 eCAR1 and αCD19 eCAR2 were indistinguishable, we chose to further validate eCAR1 and henceforth refer to it as eCAR. Similarly, significant proliferation was observed when αCD20 eCAR and αGPC3 eCAR were exposed to their corresponding target cells-Raji (CD20 positive) for αCD20 eCAR and HepG2 (GPC3 positive) for αGPC3 eCAR (Fig. 2d-e). Additionally, under stimulation by the pancreatic cancer cell line AsPC-1 (MSLN-positive), αMSLN eCAR was able to proliferate, whereas stimulation with MSLN-knockout AsPC-1 cells failed to proliferate (Fig. 2f). We next assessed the IL-2R downstream signaling in eCAR-NK cells upon tumor antigen stimulation. Phosphorylation of JAK1/3-STAT1/3/5 and AKT was effectively induced in eCAR-NK by MSLN-positive AsPC-1 cells, a response that was not replicated in either NK-92 or CAR-NK. This activation of signaling pathways was contingent upon the interaction between the tumor antigen and eCAR; stimulation with MSLN-knockout AsPC-1 did not activate these pathways in eCAR-NK cells (Fig. 2g). These findings highlight the specificity of eCAR in driving targeted signaling responses, crucially dependent on antigen recognition in an IL-2-deprived environment.
Fig. 2 | Proliferation activity of the eCAR NK-92 cells. a, Construction of four eCARs targeting different antigens (CD19, CD20, GPC3 and MSLN). b-f, Proliferation activity of eCAR NK-92 cells in response to tumor cells stimulation. Tumor cells treated with mitomycin C, including Raji (20 µM), K562(20 µM), HepG2 (30 µM) and AsPC-1 (30 µM). Data are presented as mean ± SD of five independent biological replicates. g, eCAR NK-92 cell activation pathway of JAK/STAT and MAPK indicated by Western Blot.
Enhanced Cytotoxicity of eCAR-NK in an IL-2-deprived Environments
To deepen our understanding, we proceeded to evaluate the cytotoxic capabilities of eCAR-NK in an IL-2-deprived environment. The results demonstrate that in environments supplemented with IL-2, eCAR-NK and traditional CAR-NK, both targeting specific antigens, exhibit comparable cytotoxic activities (Fig. 3a-d). However, in conditions devoid of IL-2, eCAR-NK continue to exhibit substantial cytotoxicity, while the cytotoxic capacity of traditional CAR-NK is significantly diminished (Fig. 3a-d). Additionally, eCAR-NK cells do not exhibit increased non-specific cytotoxic activity compared to CAR-NK cells. Against MSLN-knockout AsPC-1 cells, both eCAR-NK and CAR-NK display similar and relatively low cytotoxic activities (Extended Data Fig. 3a). Intriguingly, it was observed that eCAR-NK were uniquely capable of secreting the cytotoxic effector molecules interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) in the absence of IL-2 (Fig. 3e and Extended Data Fig. 3b).
We further explored the potential of eCAR to augment the in vivo antitumor efficacy of NK cells. Studies indicates that murine IL-15, with only 70% homology to human IL-15, fails to provide sufficient cytokine signaling for human NK cells (1). Thus, using Human IL-15 transgenic NCG mice (NCG-hIL15) and NCG mice can respectively simulate internal environments with sufficient and insufficient cytokines. Using the NCG-hIL15 mice, we established a Raji xenograft model of hematologic malignancy. After the adoptive transfer of effector cells for treatment, both CD19-specific eCAR-NK and CAR-NK significantly curtailed tumor progression and prolonged the survival of the tumor-bearing mice (Fig. 4a-c). To contrast these findings, we also established a Raji xenograft model in standard NCG mice, which lack human IL-15. In this model, only eCAR-NK cells demonstrated significant antitumor activity and notably extended the survival of tumor-bearing mice (Fig. 4d-f). Moreover, in solid tumor models, which possess more complex TME, eCAR-NK still exhibits robust antitumor activity (Fig. 4g-h). By examining the survival of adoptively transferred cells within the tumors, it was found that eCAR-NK could sustain proliferation over a 21-day observation period, whereas CAR-NK could not (Fig. 4i). Collectively, these results underscore that eCAR NK cells can effectively suppress tumor growth and enhance intratumoral survival, independent of cytokine support, and driven solely by tumor antigen recognition.
Fig. 3 | In vitro antitumor effect of the eCAR NK-92 cells. a-d, Cytotoxic activity of NK-92 (control), CAR or eCAR co-cultured with different tumor cells. Data were assessed by LDH Assay at the indicated E:T ratios and presented as mean ± SD of five independent biological replicates. (ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001 compared with NK-92). Data are mean ± SD. e, Killing activity of NK-92 (control), αMSLN CAR or αMSLN eCAR co-cultured with AsPC-1 (MSLN+/+). The IFN-γ and TNF-α expression levels of eCAR NK-92 cells and the control NK cells against AsPC-1 (MSLN+/+) under the condition of IL-2+ and IL-2- were assessed by flow cytometry.
Fig. 4 | In vivo antitumor effect of the eCAR NK-92 cells. a, NCG-hIL15 mice were transplanted with Raji and the in vivo assess for tumor growth after treatment. The bioluminescence was monitored every 7 days. Image radiance values were normalized using Living Image (Perkinelmer). b-c, Tumor growth (b) and survival (c) of tumor-bearing mice treated with PBS (control), NK-92, αCD19 CAR or αCD19 eCAR. d, NCG mice were transplanted with Raji and the in vivo assess for tumor growth after treatment. The bioluminescence was monitored every 7 days. Image radiance values were normalized using Living Image (Perkinelmer). e-f, Tumor growth (e) and survival (f) of tumor-bearing mice treated with PBS (control), NK-92, αCD19 CAR or αCD19 eCAR. g-h, Tumor volume and tumor weight of tumor-bearing mice treated with PBS (control), NK-92, αMSLN CAR or αMSLN eCAR (ns, not significant, **P < 0.01, ***P < 0.001 compared with NK-92). Data are mean ± SD. i, The eCAR NK-92 infiltration after treatment in subcutaneous pancreatic tumor biopsies was assess by immunofluorescence. The green fluorescence indicated the eCAR NK-92 cells and the blue fluorescence indicated DAPI, scale bar = 20 μm.
Integration of NKR Repertoire into eCAR Achieves Pan-Cancer Recognition
Considering the inherent heterogeneity and antigenic variability of solid tumors, we aimed to integrate an NKR repertoire into the eCAR to broaden the adoptive cells' recognition capabilities across a wide range of tumor antigens. We engineered eCAR by replacing the scFv to Ig-like domain of NKRs, facilitating the display of an NKR library (Fig. 5a, refer to it as eCAR-NKR library). It was found that various tumor stimuli could promote the proliferation of the eCAR-NKR library in the absence of exogenous cytokines (Fig. 5b). We summarized the NKRs and their corresponding ligands and examined the expression of NKR ligands on the surface of various tumor cells (Fig. 5c and Extended Data Fig. 4). Subsequently, we aimed to identify an eNKR from the eCAR-NKR library that could recognize pan-cancer. To achieve this, we stimulated the eCAR-NKR library with different tumor cells in vitro without exogenous cytokines and enriched eNKR. Ultimately, we discovered that NKG2D-eCAR NK cells were consistently enriched under stimulation by various tumors (Fig. 5d-i).
Fig. 5 | Design and characterization of eCAR-NKR library. a, Construction of eCAR-NKR library. Map of lentiviral constructs encoding the eCAR-NKR library expression. At 72 h after retroviral transduction, the expression of eCAR-NKR library on human NK-92 cells was measured by staining with anti-FLAG antibody, followed by flow cytometry analysis. NK-92 cells without transduction were used as negative controls. The red histograms indicate positive fluorescence of eCAR-NKR library. Data are representative of at least three independent experiments. b, The expansion level of eCAR-NKR library in response to different tumor stimulations. c, The expression levels of different tumor ligands in different tumor cell lines. Data were normalized using z-score. d-i, The enrichment of eCAR-NKR library was assessed by NGS after stimulation of different tumor cells.
eCAR-NKR improves antitumor effect target solid tumors
We proceeded to validate the antitumor efficacy of the eCAR-NKR library in vivo using various solid tumor mouse models, including GPC3-positive cancers (HepG2 and Huh-7), HER2-positive cancers (BT-474 and MKN45), and MSLN-positive pancreatic cancers (AsPC-1 and SK-OV-3). Post-treatment observations revealed that the eCAR-NKR library significantly curtailed tumor growth across these models (Fig. 6a-c). Furthermore, we established patient-derived tumor organoid xenograft (ODX) models to closely mimic clinical scenarios. The administration of the eCAR-NKR library to these ODX models effectively halted tumor progression, underscoring the library’s potent therapeutic potential (Fig. 6d-e).
Fig. 6 | In vivo antitumor effect of the eCAR-NKR library. a, Tumor growth of tumor-bearing mice with GPC3-positive cancers (HepG2 and Huh-7) treated with NK-92 (control), αGPC3 CAR or eCAR-NKR library. b, Tumor growth of tumor-bearing mice with HER2-positive cancers (BT-474 and MKN45) treated with NK-92 (control), αHER2 CAR or eCAR-NKR library. c, Tumor growth of tumor-bearing mice with MSLN-positive pancreatic cancers (AsPC-1 and SK-OV-3) treated with NK-92 (control), αMSLN CAR or eCAR-NKR library. d, Tumor growth of tumor-bearing mice with two types of patient-derived tumor organoid xenografts (HCC) treated with NK-92 (control) or eCAR-NKR library. e, Tumor growth of tumor-bearing mice with three types of patient-derived tumor organoid xenografts (STAD) treated with NK-92 (control) or eCAR-NKR library. Data are presented as mean ± SD.
Principle
Plasmid construction is a fundamental technique in molecular biology used to create recombinant DNA molecules for various applications, including gene expression, functional studies, and protein production. The process involves inserting a gene of interest into a plasmid vector—a circular DNA molecule that replicates independently within a host cell. This is achieved through restriction enzyme digestion, which cuts both the plasmid and the gene at specific sites, and ligation, which joins them together. The newly constructed plasmid is then transformed into a host organism, such as bacteria or yeast, where it can replicate and express the inserted gene, enabling further analysis and experimentation.
Method
1. The CAR sequence was synthesized by Genewiz (Suzhou, China), introducing an Xbal restriction site at the 5’ end of the sequence and a BamHI restriction site at the 3’ end of the sequence. The CARs target fragment and vector were double-digested with restriction enzymes.
2. The reactions were maintained at 37°C hour, inactivated at 65 °C for 5 min, and then stored at 4˚C. The products were proceeded by using 1.5% agarose gel electrophoresis at 120V for 45 min and then the target bands were recovered.
3. The linear vectors and the target segments were recycled.
4. The whole system was incubated at 16 °C for 30 min, inactivated at 65°C for 10 min, and then stored at 4 °C. The connection products were transformed into Stbl3 by T4 ligase (Rapid DNA Ligation Kit, Genstar). After selected clones were identified correctly through sequencing, we shaked the bacteria (500 mL volume) in the 2L flask and used the NucleoBond Xtra Midi Maxi EF plasmid purification kit (macherey-nagel 740420.10) to extract the plasmids.
Fig. Schematic diagram of plasmid construction
Principle
The lentiviral vector packaging system consists of two main components: the packaging component and the vector component. The packaging component is created by removing cis-acting sequences necessary for HIV-1 genome packaging, reverse transcription, and integration, allowing it to provide essential proteins for viral particle production. In contrast, the vector component contains these required cis-acting sequences along with a polyclonal site regulated by a heterologous promoter where a target gene can be inserted. By co-transfecting packaging cells with multiple plasmids of the vector components, replication-defective lentiviral particles carrying target genes can be produced.
Method
1. Cell Preparation: Seed HEK293T cells in a 10 cm dish at a density of 3-4 × 106 cells per dish in DMEM supplemented with 10% FBS and 1% Pen-Strep. Incubate the cells at 37 °C in a 5% CO₂ atmosphere until they reach 70-80% confluence (approximately 24 hours).
2. Transfection: Prepare the DNA-PEI complex. For each 10 cm dish, use 10 µg of the lentiviral transfer plasmid, 5 µg of pMD2.G, and 7.5 µg of psPAX2. Dilute the plasmids in 500 µL of serum-free DMEM. Separately, dilute 60 µL of 1 mg/mL PEI in 500 µL of serum-free DMEM. Combine the DNA solution with the PEI solution, mix gently, and incubate at room temperature for 20 min to allow the formation of DNA-PEI complexes.
3. Transfection Addition: Add the DNA-PEI complex dropwise to the HEK293T cells. Swirl the dish gently to distribute the complex evenly. Incubate the cells at 37°C in a 5% CO₂ atmosphere.
4. Medium Change: After 6-8 hours, replace the transfection medium with fresh DMEM supplemented with 10% FBS and 1% Pen-Strep to reduce toxicity.
5. Virus Harvesting: Collect the supernatant containing lentiviral particles at 48 and 72 hours post-transfection. Pool the supernatants and centrifuge at 500 × g for 10 min to remove cell debris.
6. Supernatant Filtration: Filter the supernatant through a 0.45 µm filter to ensure the removal of any remaining cellular debris.
7. Virus Concentration: Concentrate the viral particles by ultracentrifugation at 25,000 rpm for 2 hours at 4°C. Carefully remove the supernatant and resuspend the viral pellet in an appropriate volume of PBS or DMEM.
8. Virus Storage: Aliquot the concentrated virus and store at -80 °C until use. Avoid repeated freeze-thaw cycles.
Principle
Using artificially controlled environmental conditions to simulate the in vivo environment, cells grow and divide in vitro. In this process, cells are regulated and influenced by many factors, such as temperature, humidity, pH, nutrients, etc. Through proper regulation and control of these factors, the normal growth and division of cells can be maintained, and the biological characteristics of cells can be maintained.
Method
1. Cell resuscitation
(1) Put the required equipment, such as 10 mL pipette, pipette gun, 1 mL gun head, 50 mL centrifuge tube, culture bottle, etc. on the ultra-clean working table. Start the operation after 30 min of ultraviolet light irradiation and 30 min of ventilation.
(2) Preheat the culture medium.
(3) Disinfect the utensils with 75% alcohol and put them into the ultra-clean bench.
(4) The frozen cells were taken out from the liquid nitrogen tank.
(5) The frozen cells were placed in a water bath at 37 °C and rapidly melted by rapid shaking.
(6) Disinfect the utensils with 75% alcohol and put them into the ultra-clean bench.
(7) Take 9 mL medium to 50 mL centrifuge tube.
(8) The thawed cell suspension was pipetted into a centrifuge tube with a 1mL tip.
(9) Centrifugation at 1000 rpm/min for 5 min.
(10) Disinfect the utensils with 75% alcohol and put them into the ultra-clean bench.
(11) Aspirate the supernatant.
(12) The cells were resuspended in 1 mL medium, and the cell suspension was transferred to the culture flask. Add an appropriate amount of medium, gently shake the culture flask to evenly distribute the cells and mark them.
(13) The restored cell density and state were observed under a microscope.
(14) Put the cells back into the incubator for static culture.
2. Exchange medium
(1) The PBS buffer solution (autoclaved) and the complete medium were prepared in advance and wiped with alcohol before moving into the biological safety cabinet.
(2) Change the medium: the adherent cells were completely changed, the old medium was removed, washed with PBS for 2-3 times, and then the fresh medium was added. The suspension cells were replaced with half of the medium, half of the old medium was removed, and then fresh medium was added. The culture could also be transferred to a centrifuge tube, and the old medium was removed by centrifugation, and then the new medium was added to re-suspend the cells.
3. Cell passages
(1) Complete medium, PBS, trypsin, petri dishes, centrifuge tubes, etc. were prepared in advance, and transferred into a biological safety cabinet after alcohol wiping.
(2) The old medium was discarded, and then washed 2-3 times with PBS, 1 mL trypsin was added, and the cells were digested. When the cells became round under the microscope, 1 mL medium was added immediately to terminate the digestion, and then the cells were scanned layer by layer. The cell suspension was added to the centrifuge tube, 1000 rpm, 5 min.
(3) After centrifugation, the medium was discarded, the new medium was added to blow the re-suspended cells, and then added to the culture dish, 0.5 or 1 mL cell suspension was added to each culture dish.
Principle
Organoid culture technology refers to a group of cells with self-renewal and self-organisation capabilities cultured in an in vitro environment. These cells can be derived from primary cells, embryonic stem cells or multipotent stem cells. They possess organ-like structure and function and can mimic in vivo tissue structure and function to the greatest extent possible. They can also be passaged long-term and used as an experimental model for patient individualization.
Typical organoid culture protocols commence with the isolation of embryonic stem cells or pluripotent stem cells, which are subsequently cultured in a supportive medium to facilitate three-dimensional cell growth. The signaling pathways that mediate organoid formation are the same as those used for organ development and maintenance of homeostasis in vivo. Consequently, a variety of cytokines, growth factors and small molecule compounds must be introduced during the culture process in order to activate or inhibit specific cell signaling pathways. The primary component of the culture is Matrix Gel, a natural soluble basement membrane matrix extracted from extracellular matrix protein-rich mouse tumors. It consists of a range of extracellular matrix components, including laminin, collagen type IV and nestin, and provides support, tensile strength and scaffolding support for tissues and cells (Fig.).
Fig. Hepatocellular carcinoma organoid construction
Method
1. The process of digesting human liver tumor tissue
(1) The tumor tissue should be chopped into pieces measuring between 0.5 and 1 mm³ under sterile conditions and placed into a 100 mm petri dish.
(2) The chopped tissue should then be submerged into a 15 mL centrifuge tube, with approximately 10 mL of ice-cold washing medium added. The tissue should then be gently agitated with a 10 mL pipette to precipitate the tumor tissue. The supernatant, including blood cells and floating fat mass, should be discarded, and this step should be repeated once.
(3) A volume of approximately 4-5 mL of pre-warmed human liver digest should be added per gram of tissue. The solution should then be placed on a test-tube rotary shaker in an incubator at 37 °C, mixed thoroughly, and allowed to digest.
(4) Following a 30-minute digestion period, the solution should be agitated to ascertain the presence of single cells. In the event of a low concentration of single cells, the solution should be returned to 37 °C and the digestion process continued. The suspension should be monitored at 10-minute intervals for a maximum of 90 min, with digestion halted when the suspension contains 80-100% single cells.
(5) Upon completion of the digestion process, the solution should be filtered through a 70 µm filter and then through a second filter, which should be pre-cooled, before being added to the washing medium.
(6) Centrifuge at 4 ℃ and 300×g for 5 min, discard the supernatant, and add pre-cooled washing medium to 15 mL.
(7) Centrifuge at 300×g for 5 min at 4 °C, discard the supernatant, and then add pre-cooled washing medium to 15 mL.
2. Inoculation of liver cancer cells
(1) The supernatant should be removed, and the cells should be resuspended in BME2 matrix gel. The cells should then be inoculated into 24-well plates with the matrix gel kept at a low temperature throughout the process.
(2) Add 500 µL of medium per well in the 24-well plate and incubate in a cell culture box.
(3) The medium should be replaced with a fresh one after a period of three to four days.
Principle
Immunodeficient mice are a kind of mice whose immune system function is inhibited or lost through genetic modification, which makes them an important experimental tool in many biomedical research fields. Because of their incomplete immune system, these mice can receive xenotransplantation of cells or tissues, such as humans, for a variety of research applications.
Method
The luciferase gene-labeled HCC cell line in the logarithmic growth phase was digested and prepared into 5×107 cell/mL cell suspension by HBSS.
1. The NCG mice were anesthetized and opened about 1 cm horizontally under the xiphoid process, and part of the liver was exposed by squeezing the left and right abdominal cavity. 100 μL of the prepared cells were injected into the liver of the nude mice with 1 mL insulin injection needle. At the same time, the needle was drawn and the assistant used Vetbond Tissue Adhesive to quickly close the pinhole to prevent bleeding and tumor cell outflow to form intraperitoneal implantation.
2. The liver was gently pushed back into the abdominal cavity with a cotton rod wet with normal saline, and the abdomen was closed.
3. Subsequent tumor volume was monitored by small animal imaging.
Principle
The TME significantly influences the biological characteristics of tumor cells. Subcutaneous implanted tumors are removed from their primary tissue microenvironment, resulting in development that deviates from clinical reality. Additionally, this model exhibits poor drug responsiveness, low metastatic rates, and survival curve data that do not correlate with clinical outcomes. To address these limitations, an in-situ transplantation model of tumor cell lines was constructed using primary cultured tumor cells. This model enables the assessment of drug efficacy and disease progression through various methods, including the observation of clinical symptoms in mice, survival times, tumor volume measurements, and the use of bioluminescent imaging to track luciferase-labeled tumor cells. This approach allows for real-time monitoring of tumor growth, distribution, and metastasis. Given the unique tumor growth microenvironment of liver cancer, coupled with the liver being the largest immunologically privileged organ in the human body, in situ tumor formation represents the optimal model for exploring adoptive immunotherapy for liver cancer.
Fig. The procedures of the in-situ pancreatic tumor-bearing mouse model construction
Method
1. Cell Culture
Pancreatic cancer cells tumor cell were cultured in DMEM medium supplemented with 10% fetal bovine serum, 100 mg/mL penicillin, and 100 mg/mL streptomycin and maintained in a 37 °C incubator with 5% CO₂.
2. Construction of the In Situ Pancreatic Cancer Tumor Model
(1) Cell preparation
Tumor cells were seeded in 10 cm culture dishes using DMEM medium (containing 10% fetal bovine serum and 1% penicillin-streptomycin) and incubated at 37 °C in a 5% CO₂ atmosphere until the cells reached the logarithmic growth phase.
(2) Harvesting cells with trypsin
1 mL of 0.25% trypsin was added to each 10 cm dish for digestion for 1 min. Afterward, 2 mL of DMEM containing 10% fetal bovine serum was added to terminate the digestion. The cell suspension was transferred to a 15 mL centrifuge tube, and 5 mL of PBS was added to wash the dish. The solution was collected into the 15 mL tube, centrifuged at 1000 rpm/min for 5 min, and the supernatant was discarded. The tumor cell cells were then resuspended in PBS, centrifuged again at 1000 rpm/min for 5 min, and the wash was repeated twice.
(3) Cell counting
The harvested tumor cell cells were resuspended in PBS, counted, and adjusted to a final concentration of 2×10⁷ cell/mL.
(4) Anesthesia of mice
Each mouse received an intraperitoneal injection of the anesthetic pentobarbital (150-200 μL), and the injection site was disinfected with 75% ethanol prior to administration.
(5) Injection of tumor cell
After anesthesia, the abdomen of the mouse was disinfected with povidone-iodine using a cotton swab. A longitudinal incision of approximately 1 cm was made in the left upper abdomen with ophthalmic surgical scissors. The spleen was gently elevated using a sterile cotton swab to visualize it, followed by careful exploration of the pancreas. Once the pancreas was located, the tip of the tail was grasped with curved ophthalmic forceps and gently pulled to expose the pancreas (care was taken to avoid rupturing the spleen). A 27-gauge needle was used with a 1 mL syringe to gently insert the needle into the pancreas from the tail end towards the head region, ensuring that it did not penetrate through the pancreas to prevent leakage of pancreatic fluid, which could lead to the death of the mouse. Each mouse received an injection of 50 μL containing 2×10⁷ cell/mL of tumor cell cells (successful injections were indicated by a visible small blister formation; if the pancreas was punctured, leakage of the cell suspension would be observed). After the injection, the direction of the needle was slightly adjusted, and the cell suspension was slowly injected while withdrawing the needle. Applying gentle pressure with a sterile cotton swab at the injection site for a moment effectively prevented leakage. The spleen and pancreas were then gently returned to the abdominal cavity, and the skin and peritoneum were sutured using 3-0 surgical sutures. Post-surgery, the mice were placed in a warm environment illuminated by incandescent light until they awakened naturally.
Principle
Patient-derived organoid xenograft (PDOX) mouse model is a human xenograft model established based on the patient-derived organoid (PDO) model in immune-deficient mice. This method involves extracting cells from a patient's tumor tissue, culturing them into three-dimensional organoid samples in the laboratory, and then transplanting them into mice to form a xenograft model.
Method
1. Mouse Preparation: Use immunodeficient mice (e.g. NOD/SCID or NSG mice) to minimize immune rejection. Anesthetize the mice with isoflurane.
2. Surgical Procedure: Sterilize the surgical area and set up a sterile field. Make a small incision in the appropriate organ (e.g. mammary gland for breast cancer, liver for liver cancer or skin). Implant the minced tumor pieces or cells directly into the organ. For orthotopic placement, ensure proper alignment with the native tissue.
Principle
The action of lactate dehydrogenase resulted in the reduction of NAD+ to form NADH. Subsequently, the combination of NADH and INT (2-p-iodophenyl-3-nitrophenyl tetrazolium chloride) was catalyzed by thioctylamine dehydrogenase (diaphorase) to form NAD+ and formazan. This process produced an absorption peak at 490 nm. Absorption peaks were generated at 490 nm, thus enabling the quantification of lactate dehydrogenase activity by colourimetry. Furthermore, the absorbance was found to be linearly and positively correlated with the activity of lactate dehydrogenase. Lactate dehydrogenase activity in cell lysates was assayed to evaluate the in vitro and ex vivo killing activity of CAR-NK cells. The schematic diagram of the principle is shown in Figure.
Fig. Principle of LDH Assay
Method
The cytotoxicity function of CAR-NK cells was assessed by co-culture tumor cells at an E:T (effector to target) ratio of 1:1. For lactate dehydrogenase (LDH), CytoTox96 cytotoxicity assay was used according to the manufacturer's instructions (Promega, G1780). Cytotoxicity (%) = [LDHE:T -LDHE]/LDHMax× 100%.