Xiaojun Xu’s research while affiliated with Sun Yat-sen University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (59)


Erratum: Busulfan Plus Fludarabine Compared With Busulfan Plus Cyclophosphamide for AML Undergoing HLA-Haploidentical Hematopoietic Cell Transplantation: A Multicenter Randomized Phase III Trial
  • Article

March 2025

·

6 Reads

Journal of Clinical Oncology

Yiwen Ling

·

·

·

[...]

·

Qifa Liu

Proportion of CD3⁻CD56⁺ NK cells in human PBMNCs. Flow cytometric analysis of CD3⁻CD56⁺ NK cells using annexin V/FITC staining before (Left) and after (Right) CD56⁺ cell positive selection. Data was presented as the mean ± SD of three independent experiments. NK, natural killer; PBMNCs, peripheral blood mononuclear cells.
Effects of ACM treatment on the metabolism of KG-1α and HL-60 cells in vitro. (A) KG-1α and (B) HL-60 cells were treated with different concentrations of ACM (1-1,280 nmol/l) for 24, 48, 72 and 96 h, and cell viability was evaluated using the CCK-8 assay. AML cells were pre-incubated with 1-1,280 nmol/l ACM, blank control (Control: without ACM treatment) or negative control (DMSO). Data was presented as the mean ± SD of three independent experiments. ****P < 0.0001. ACM, aclacinomycin; CCK-8, cell counting kit-8; AML, acute myeloid leukemia; DMSO, dimethyl sulfoxide.
Effects of combination treatment (allogeneic NK cells and ACM) on the metabolism of KG-1α and HL-60 cells. (A) The effects of different concentrations of ACM on NK cell viability after 96 hours of treatment were assessed using the CCK-8 assay. The control group consisted of cells without ACM treatment, and DMSO was used as a negative control. (B) KG-1α and (C) HL-60 cells were treated with ACM (0-320 nmol/l) and NK cells (effector to target ratio was 20) for 24, 48, 72 and 96 h, and cell viability was analyzed using an LDH assay. AML cells were pre-incubated with 10-320 nmol/l of ACM or Control (without ACM treatment). Data are presented as the mean ± SD of three independent experiments. NK, natural killer; ACM, aclacinomycin; LDH, lactate dehydrogenase; AML, acute myeloid leukemia.
Representative flow cytometry images of (A) KG-1α cells, (C) HL-60 and (E) PBMNC following combination treatment with allogeneic NK cells and ACM. ACM (40 nmol/l) and allogeneic NK cells (effector to target ratio was 20) were administered alone or together to KG-1α cells and the incubation time ranged from 24 to 96 h. Afterwards, the apoptosis of these cells was determined by FCM. Quantitative analysis of the apoptotic ratio of the (B) KG-1α, (D) HL-60 and (F) PBMNC after the combination treatment of allogeneic NK cells and ACM. Data are presented as the mean ± SD. *P<0.05, **P<0.01, ***P<0.001 and ****P<0.0001.
Modulation of the expression of ICD pathway-related molecules in KG-1α cells following combination treatment with ACM and allogeneic NK cells. (A) Western blotting of CRT, ATP and HMGB1. (B-D) Quantification analysis of the western blotting results for CRT, ATP and HMGB1 respectively. ACM (40 nmol/l) and allogeneic NK cells (effector to target ratio was 20) were administered alone or together to the KG-1α cells and the incubation time ranged from 24 to 96 h. The levels of CRT, ATP and HMGB1 were then analyzed by western blotting, followed by ImageJ quantification. CRT, calreticulin; ATP, adenosine triphosphate; HMGB1, high mobility group box 1.

+2

Aclacinomycin enhances the killing effect of allogeneic NK cells on acute myeloid leukemia cells by inducing immunogenic cell death
  • Article
  • Full-text available

February 2025

·

19 Reads

Introduction Natural killer (NK) cells, which exert spontaneous cytotoxicity against infectious diseases and cancer, also play an important role in leukemia therapy. Despite the success of NK-based therapy in the treatment of myeloid leukemia, the potential use of NK alloreactivity in these hematologic malignancies remains elusive. The aim of the present study was to investigate whether allogeneic NK cells combined with aclacinomycin (ACM) could enhance anti-leukemic functionality against an acute myeloid leukemia (AML) cell line and to clarify the underlying mechanism. Methods KG-1α and HL-60 AML cell lines were subjected to different treatments. The effects of different drug combinations on cytotoxicity, cell viability, and apoptotic status were examined. Results The results showed that the combination of ACM (40 nmol/l) and allogeneic NK cells (ratio 20:1) was significantly cytotoxic to AML cells and increased the apoptosis of AML cells, especially after 72 h of treatment. Subsequent analyses revealed that the expression of immunogenic cell death (ICD)-related molecules calreticulin, adenosine triphosphate, and high mobility group box 1, as well as NK cell effector production—perforin and granzyme B—was markedly increased in the combination treatment group. These findings suggest that ACM enhances the anti-leukemic activity of allogeneic NK cells through the ICD pathway. Discussion These results demonstrated that allogeneic NK cells had enhanced functional responses when stimulated with ACM in vitro, exhibiting superior effector cytokine production and cytotoxicity compared to the control, which contained conventional NK cells. In conclusion, the present study suggested that the combination of ACM and allogeneic NK cells is a promising therapeutic strategy against AML.

Download


Loss of Trim31 Worsens Cardiac Remodeling in a Mouse Model of Heart Failure by Enhancing the Activation of the NLRP3 Inflammasome

December 2024

·

8 Reads

·

1 Citation

Inflammation

Tripartite motif-containing protein 31 (Trim31) is known to be involved in various pathological conditions, including heart diseases. Nonetheless, its specific involvement in heart failure (HF) has yet to be determined. In this study, we examined the function and mechanism of Trim31 in HF by using mice with cardiac-specific knockout (cKO) of Trim31. The HF mouse model was induced via the subcutaneous injection of isoproterenol (ISO). We observed a decrease in Trim31 expression in the heart tissues of mice with HF. Compared with wild-type (WT) mice, Trim31 cKO mice presented more severe characteristics of HF, including worsened cardiac dysfunction, hypertrophy, and fibrosis. However, these symptoms in Trim31 cKO mice were significantly reversed when they received an intramyocardial injection of recombinant adeno-associated virus (AAV) expressing Trim31. Excessive activation of the NLRP3 inflammasome, manifested by increased levels of NLRP3, ASC, cleaved Caspase-1, cleaved GSDMD, IL-1β, and IL-18, was observed in Trim31 cKO mice with HF. However, Trim31 overexpression effectively reversed the NLRP3 inflammasome activation in Trim31 cKO mice with HF. Selective inhibition of the NLRP3 inflammasome with the NLRP3 inhibitor MCC950 effectively reversed the worsened cardiac dysfunction, hypertrophy, and fibrosis observed in Trim31 cKO mice with HF. Overall, the findings from this study reveal a crucial role of Trim31 in HF. Trim31 deficiency may contribute to the progression of HF by promoting cardiac hypertrophy, fibrosis, and inflammation by facilitating the activation of the NLRP3 inflammasome. Therefore, Trim31 may hold significant potential as a therapeutic target for the treatment of HF.


Cumulative incidences of leukemia relapse (A), non‐relapse mortality (B), overall survival (C), and leukemia‐free survival (D) in the prophylactic and control groups.
A prophylactic tyrosine kinase inhibitor strategy based on measurable residual disease pre‐transplantation for Ph acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation: A prospective multicenter cohort study

November 2024

·

12 Reads

Relapse is the major cause of treatment failure in Philadelphia chromosome‐positive (Ph⁺) acute lymphoblastic leukemia (ALL) undergoing allogeneic hematopoietic stem cell transplantation (allo‐HSCT). This study aimed to evaluate the effect of a prophylactic tyrosine kinase inhibitor (TKI) strategy on relapse in this population. Patients were assigned to prophylactic or control groups based on measurable residual disease (MRD) pre‐transplantation. The primary endpoint was the cumulative incidence of relapse. A total of 110 patients with Ph⁺ ALL undergoing allo‐HSCT were enrolled in this prospective study. Thirty‐eight patients with positive MRD pre‐transplantation were included in the prophylactic group, and 72 with negative MRD pre‐transplantation were included in the control group. The 4‐year cumulative incidence of relapse was 25.3% (95% CI: 12.1%–41.0%) and 20.3% (11.6%–30.7%; HR = 1.272, 95% CI: 0.551–2.940, p = .549), and non‐relapse mortality was 10.5% (3.3%–22.7%) and 9.7% (4.2%–17.9%; HR = 1.094, 95% CI: 0.320–3.738, p = .928) in the prophylactic and control groups. The 4‐year overall survival was 71.8% (53.2%–84.1%) and 84.1% (72.9%–90.9%; HR = 1.746, 95% CI: 0.741–4.112, p = .196), and leukemia‐free survival was 64.1% (45.8%–77.7%) and 70.0% (57.6%–79.4%; HR = 1.212, 95% CI: 0.607–2.421, p = .585) in the prophylactic and control groups. Our results suggest that prophylactic TKI post‐HSCT in patients with positive MRD pre‐transplantation can produce outcomes comparable to negative MRD pre‐transplantation without TKI post‐HSCT.



Fig. 2 Viability and proliferation of NT-T, N and N15 CAR-T cells. (A) Proliferation multiplicity of NT-T, N and N15 CAR-T cells in culturing with IL-2 maintenance. (B) Secretion of IL-15/IL-15Rα in cultures without IL-2 maintenance detected by IL-15 ELISA kit. (C) The percentage of viable T cells in cultures without IL-2 maintenance. IL-15 concentration in N15 group was higher than that in NT or N group, but no statistically different between NT and N groups (P>0.05). (D) The numbers of viable T cells in cultures without IL-2 maintenance. The data were presented as the mean of 3 independent tests from 5 individual donors. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001
Fig. 6 Frequency of cytokine expressing T cells from NT-T, N and N15 CAR-T cell groups in co-culturing with B-LCLs or PBMCs without IL-2 maintenance measured by ICS. (A-E) In co-culturing with B-LCLs; (F-H) In co-culturing with PBMCs. The data were presented as the mean of 3 independent tests from 5 individual donors. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001
Fig. 7 Examination for clinical outcomes of EBV-PTID mouse models treated by three types of T cells. (A) Schematic schedule of treatment and examination for B-LCLs xenografted NSG mice (EBV-PTID mouse models). (B) Weight curve (n = 6). (C) MR imaging of treated and control mice (n = 3). The red solid line arrows showed the tumor signals; red dashed arrows showed the suspected tumor signals. (D) Pathological observation for representative livers of treated and control mice. The white solid line arrows showed the tumor signals; white dashed arrows showed the suspected tumor signals. (E) EBV DNA load in caudal venous blood of treated and control mice detected by qPCR (n = 6). (F) Kaplan-Meier survival curve (n = 6). * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001
Fig. 8 Evaluation for effects of three types of T cells for treating EBV-PTID mouse models. (A) B-LCL tumor burden in the liver, lung, spleen, kidney, bone marrow and peripheral blood of mice detected by flow cytometry (n = 6). (B) EBV DNA load in the liver, lung, spleen, kidney, bone marrow and peripheral blood of mice detected by qPCR (n = 6). (C) T cells resided in the liver, lung, spleen, kidney, bone marrow and peripheral blood evaluated by flow cytometry (n = 6). (D) H&E staining of spleen tissue sections. Scale = 400 μm. The bottom left region is of 200-fold magnification. (E) Measurement of cytokines in blood of mice by Multiplex Human Cytokines Analysis (n = 6). Significantly high level of IL-15, IFN-γ, Perforin and Granulysin were detected in N15 CAR-T cell group. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001
Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder

August 2024

·

16 Reads

·

6 Citations

Experimental Hematology and Oncology

Epstein-Barr virus (EBV) related post-transplant lymphoproliferative disorder (EBV-PTLD) is a life-threatening complication after hematopoietic stem cell transplantation (HSCT) or solid organ transplantation (SOT), for which no standard therapeutic means have been developed. Significant increase expression of natural killer group 2 member D ligands (NKG2DLs) was observed on B-lymphoblastoid cells of EBV-PTLD, indicating NKG2DLs as potential therapeutic targets for treatment of EBV-PTLD. In this study, the recombinant constructs of NKG2D CAR and IL-15/IL-15Rα-NKG2D CAR were generated with a retroviral vector and then transduced to human T cells to produce NKG2D CAR-T and IL-15/IL-15Rα-NKG2D CAR-T cells, respectively. B-lymphoblastoid cell lines (B-LCLs) and the xenografted mouse models were established to evaluate the efficacy of these CAR-T cells. IL-15/IL-15Rα-NKG2D CAR-T cells exhibited superior proliferation and antigen-specific cytotoxic effect compared to NKG2D CAR-T, as IL-15/IL-15Rα signaling promoted the expansion of less differentiated central memory T cells (TCM) and increased expression of CD107a and IFN-γ. Moreover, EBV DNA load was dramatically reduced, and 80% B-LCL cells were eliminated by IL-15/IL-15Rα-NKG2D CAR-T cells after co-culturing. In-vivo study confirmed that IL-15/IL-15Rα-NKG2D CAR-T cell therapy significantly enhanced antiviral efficacy in mice, as the serum load of EBV after IL-15/IL-15Rα-NKG2D CAR-T cell infusion was 1500 times lower than the untreated control (P < 0.001). The enhanced efficacy of IL-15/IL-15Rα-NKG2D CAR T cells was probably due to the IL-15/IL-15Rα signaling improved homing and persistence of NKG2D CAR-T cells in vivo, and increased the production of IFN-γ, Perforin, and Granulysin. In conclusion, NKG2D CAR-T cells co-expressing IL-15/IL-15Rα promoted the central memory CAR T cell proliferation and improved the homing and persistence of CAR T cells in vivo, resulting in enhanced anti-tumor and anti-viral effects in treating EBV-PTLD. Supplementary Information The online version contains supplementary material available at 10.1186/s40164-024-00553-z.




Figure 2. Venetoclax affects ASCT2-mediated glutamine uptake. (A) Representative TMRM fluorescence images of each group, with red fluorescence representing mitochondria and blue fluorescence representing the cell nuclei. Scale bar, 20 µm. (B) Detection of mitochondrial membrane potential after treatment with different concentrations of Venetoclax for 24 h. (C) After 24 h Venetoclax treatment in vitro, the expression of ATF4, CHOP, ASCT2, IDH2 and Rb was measured through western blotting. ATF4 and CHOP expression was markedly increased in the Venetoclax group compared with that in the control group. (D) The transcriptional levels of IDH2 and ASCT2 at different time points after drug administration were detected using reverse transcription-quantitative PCR (n=3). Results are presented as the means ± standard deviation. * P<0.05 vs. 0 µM venetoclax. ** P<0.01 vs. 0 h post treatment. TMRM, tetramethylrhodamine methyl ester; CHOP, CCAAT/enhancer-binding protein homologous protein; ATF4, activating transcription factor 4; ASCT2, alanine-serine-cysteine transporter 2; IDH2, isocitrate dehydrogenase 2; Rb, retinoblastoma.
Figure 4. GSH can further promote the synergistic effect of VEN and azacitidine. (A) Representative flow cytometry histogram of cell cycle and S phase ratio after treatment with the indicated concentrations of VEN for 24 h. (B) SKM-1 cells were treated with the indicated concentrations of VEN for 24 h before being harvested, and had GSH concentration measured using liquid chromatography-mass spectrometry. (C) Representative flow cytometry histogram of cell cycle 24 h after treatment. (D) S phase ratio after treatment with indicated concentrations of VEN and 1 mM GSH for 24 h. (E) Different concentrations of VEN and azacitidine were added to the SKM-1 culture medium, whereas (F) 1 mM GSH was added during treatment, before cell viability was measured using a Cell Counting Kit-8 assay 48 h after treatment. The values in the heat map represent the mean values from three replicates; darker shades indicate a higher OD450 value. * P<0.05 and ** P<0.01 vs. 0 µM venetoclax in Fig. 4A and B. **** P<0.0001. VEN, Venetoclax; GSH, Glutathione.
Glutathione promotes the synergistic effects of venetoclax and azacytidine against myelodysplastic syndrome‑refractory anemia by regulating the cell cycle

October 2023

·

43 Reads

·

1 Citation

Experimental and Therapeutic Medicine

Azacitidine is a DNA methyltransferase inhibitor that has been used as a singular agent for the treatment of myelodysplastic syndrome-refractory anemia with excess blast-1 and -2 (MDS-RAEB I/II). However, recurrence and overall response rates following this treatment remain unsatisfactory. The combination of azacitidine and venetoclax has been used for the clinical treatment of a variety of hematological diseases due to the synergistic killing effect of the two drugs. Venetoclax is a BCL-2 inhibitor that can inhibit mitochondrial metabolism. In addition, azacitidine has been shown to reduce the levels of myeloid cell leukemia 1 (MCL-1) in acute myeloid leukemia cells. MCL-1 is an anti-apoptotic protein and a potential source of resistance to venetoclax. However, the mechanism underlying the effects of combined venetoclax and azacitidine treatment remains to be fully elucidated. In the present study, the molecular mechanism underlying the impact of venetoclax on the efficacy of azacitidine was investigated by examining its effects on cell cycle progression. SKM-1 cell lines were treated in vitro with 0-2 µM venetoclax and 0-4 µM azacytidine. After 24, 48 and 72 h of treatment, the impact of the drugs on the cell cycle was assessed by flow cytometry. Following drug treatment, changes in cellular glutamine metabolism pathways was analyzed using western blotting (ATF4, CHOP, ASCT2, IDH2 and RB), quantitative PCR (ASCT2 and IDH2), liquid chromatography-mass spectrometry (α-KG, succinate and glutathione) and ELISA (glutamine and glutaminase). Venetoclax was found to inhibit mitochondrial activity though the alanine-serine-cysteine transporter 2 (ASCT2) pathway, which decreased glutamine uptake. Furthermore, venetoclax partially antagonized the action of azacitidine through this ASCT2 pathway, which was reversed by glutathione (GSH) treatment. These results suggest that GSH treatment can potentiate the synergistic therapeutic effects of venetoclax and azacitidine combined treatment on a myelodysplastic syndrome-refractory anemia cell line at lower concentrations.


Citations (36)


... In the context of cardiac remodeling, the activation of the NLRP3 inflammasome promotes not only cardiac inflammation and fibrosis but also aggravates pathological myocardial hypertrophy, consequently exacerbating symptoms of HF (99). Elevated levels of G protein-coupled receptor kinase 2 (GRK2) were identified in hypertrophied myocardial tissue (100,101). ...

Reference:

Active ingredients of traditional Chinese medicine inhibit NOD-like receptor protein 3 inflammasome: a novel strategy for preventing and treating heart failure
Loss of Trim31 Worsens Cardiac Remodeling in a Mouse Model of Heart Failure by Enhancing the Activation of the NLRP3 Inflammasome

Inflammation

... IL-15 retroviral construct (Fig. 1A). Recombinant retroviruses were produced by co-transfecting retroviral plasmids and packing plasmids (PegPam3 plasmid and RDF plasmid) into HEK-293 T/17 cells as previous described [26,27]. Human PBMCs were isolated by density gradient centrifugation from the "Buffy Coat" of healthy blood donors at the Guangzhou Blood Centre using Human Peripheral Blood Lymphocyte Separation Solution (Tianjin Haoyang Biological Manufacture Co., Ltd, China). ...

Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder

Experimental Hematology and Oncology

... To the best of our knowledge, it is not known whether these XPO1 mutations and XPO1 mislocation can be detected or have any clinical impact in human AML. However, a recent study described the generation of an induced pluripotent stem cell line from an AML patient; this cell line had maintained mutations of XPO1 as well as PALB2, and it showed characteristics similar to embryonic stem cells [60]. This observation suggests that XPO1 mutations have the capacity to contribute to malignant transformation also in AML, but additional studies are definitely needed. ...

Generation and characterization of the iPS cell line (SYSUSHi001-A) derived from the peripheral blood mononuclear cells (PBMCs) of a 33-year-old patient with acute myeloid leukemia (AML)
  • Citing Article
  • February 2024

Stem Cell Research

... 35,103 Different mechanisms of resistance to venetoclax have been identified in recent years, including downregulation of the proapoptotic proteins BCL-2 interacting mediator of cell death and BCL-2-associated X protein secondary to venetoclax exposition, acquisition of MCL-1 or BCL-XL dependence of myeloid blasts, and acquisition of BCL-2 mutations. [104][105][106] Possible strategies to overcome venetoclax resistance are currently being tested [107][108][109][110] and have been reviewed extensively in Griffioen et al. 111 Finally, interest in testing venetoclax therapies in other potential pediatric myeloid settings is emerging, including the management of AML molecular relapse after HCT 80 and different diseases such as chronic myeloid leukemia 112 and juvenile myelomonocytic leukemia. 28,113 Future studies will also have to dissect the optimal duration of venetoclax treatment and the number of cycles to be administered. ...

Glutathione promotes the synergistic effects of venetoclax and azacytidine against myelodysplastic syndrome‑refractory anemia by regulating the cell cycle

Experimental and Therapeutic Medicine

... To investigate the mechanism of YOD1 in ccRCC development, we overexpressed YOD1 in A498 and 786-O cells and assessed various YOD1-regulated signaling pathways documented in prior studies [18,[20][21][22][23][24][25]. The result showed that only a slight elevation in Trim33 protein expression was detected ( Supplementary Fig. 2B). ...

miR-221/222 induce instability of p53 By downregulating deubiquitinase YOD1 in acute myeloid leukemia

Cell Death Discovery

... This study showed that outcomes with BuFlu are independent of the donor type and the subsequently adopted GvHD prophylaxis. Moreover, authors found a reduced incidence of grade 3 regimenrelated toxicities (0% in BuFlu vs. 9% in BuCy2), thus confirming that BuFlu is less toxic when compared to BuCy2, even in patients receiving post-transplant cyclophosphamide [20]. ...

Busulfan Plus Fludarabine Compared With Busulfan Plus Cyclophosphamide for AML Undergoing HLA-Haploidentical Hematopoietic Cell Transplantation: A Multicenter Randomized Phase III Trial
  • Citing Article
  • June 2023

Journal of Clinical Oncology

... CAR-T cells exhibit greater proliferation, migration, and cytotoxicity than second-generation Nectin4 CAR-T cells, in which Nectin4-7.19 CAR-T consist of a scFv derived from an antibody against human Nectin4 [24]. VHHs have also been used as the antigen-recognition domains of CAR-T cells due to the small size, high stability, and ability to recognize hidden antigen epitopes. ...

Development of Nectin4/FAP-targeted CAR-T cells secreting IL-7, CCL19, and IL-12 for malignant solid tumors

... Chen et al. 4 indicated that HOXA3-10 genes may serve as possible therapeutic targets and prognostic markers of AML since the upregulation of these genes may contribute to the initiation of AML and their existence may serve as an indicator of adverse prognosis. Ye et al. 108 investigated the relationship between fatty acid metabolism (FAM), TME, as well as the prognosis of AML patients and performed a functional enrichment assay to assess the significance of FAM in the immunosurveillance against AML. Their scRNA-seq analysis revealed that the levels of FAM-related genes were elevated in the population rich in LSCs, and these genes were then utilized to develop a prognostic model capable of accurately predicting the outcome of AML patients and changes in the immunosurveillance based on TME. ...

Fatty acid metabolism predicts prognosis and NK cell immunosurveillance of acute myeloid leukemia patients

... However, third-generation CARs carry risks of off-target effects and excess cytokine production, with limited clinical data available. Consequently, fourth-generation CARs have been developed to address these concerns [43,44] Four Generation Fourth-generation CARs, derived from second-generation CARs, integrate domains regulating cytokine expression alongside costimulatory domains to bolster antitumor responses. These CARs, engineered to secrete IL-12, IL-18, or IL-21, enhance CAR-T cell cytotoxicity, proliferation, and memory T cell differentiation, thereby improving killing efficacy and sustaining antitumor effects in preclinical models [45][46][47] ...

Combination of 4-1BB and DAP10 promotes proliferation and persistence of NKG2D(bbz) CAR-T cells

... lymphoid leukemia. UNC13B was demonstrated to regulate tumor cell resistance to arsenic trioxide (11). These findings suggest a potential involvement of UNC13B in chemotherapeutic drug resistance in tumor cells. ...

UNC13B Promote Arsenic Trioxide Resistance in Chronic Lymphoid Leukemia Through Mitochondria Quality Control