Wei-Dong Han’s research while affiliated with Chinese PLA General Hospital (301 Hospital) 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 (57)


Fig. 1 Immunosuppressive microenvironment in solid tumors. MDSC myeloid-derived suppressor cell, Treg regulatory T cell, TAM tumor-associated macrophage, TAF tumor-associated fibroblast
Fig. 2 Intracellular costimulatory domain of a CAR construct used in CAR-T cells tested in clinical trials. The data were obtained from https:// clinicaltrials.gov, which was accessed on January 30, 2019. a Diagram of clinical trials of CAR-T cells from different generations. There are 342 registered trials categorized as CAR-T cell trials (second generation: 133, third generation: 20, fourth generation: 5, NA, not available). b Diagram of clinical trials of CAR-T cells using different costimulatory molecules. A total of 156 available CAR constructs with different costimulatory molecules were specifically indicated, and a pie diagram is presented that shows the percentages of trials using cells with different costimulatory domains (4-1BB: 64.7%, CD28: 19.2%, CD28+4-1BB: 9%, CD28+OX40: 2%, CD28+TLR2: 0.6%, CD28+4-1BB+CD27: 2%, CD27: 0.6%, iMyD88/CD40: 0.6%, NKG2D and DAP10: 1.2%)
Application of engineered T cells in clinical trials for treating hematological malignancies
Published clinical studies using CAR-T cells for treating solid tumors
Genetically engineered T cells for cancer immunotherapy
  • Literature Review
  • Full-text available

September 2019

·

645 Reads

·

194 Citations

Signal Transduction and Targeted Therapy

·

Xue Li

·

Wei-Lin Zhou

·

[...]

·

T cells in the immune system protect the human body from infection by pathogens and clear mutant cells through specific recognition by T cell receptors (TCRs). Cancer immunotherapy, by relying on this basic recognition method, boosts the antitumor efficacy of T cells by unleashing the inhibition of immune checkpoints and expands adaptive immunity by facilitating the adoptive transfer of genetically engineered T cells. T cells genetically equipped with chimeric antigen receptors (CARs) or TCRs have shown remarkable effectiveness in treating some hematological malignancies, although the efficacy of engineered T cells in treating solid tumors is far from satisfactory. In this review, we summarize the development of genetically engineered T cells, outline the most recent studies investigating genetically engineered T cells for cancer immunotherapy, and discuss strategies for improving the performance of these T cells in fighting cancers.

Download

Fig. 1. Anatomical features of CAR constructs. CAR: chimeric antigen receptor; V H : variable heavy chain; V L : variable light chain; ZAP70: zeta-chain-associated protein kinase 70; LAT: linker for activation of T cells; scFv: single-chain variable fragment; TCR: T-cell receptor; CD: cluster of differentiation; SUPRA: split, universal, and programmable.
Table 1 (continued )
Table 2 CAR-T trials for the treatment of hematologic malignancy.
Table 2 (continued )
Table 3 CAR-T targets for treatment of solid tumors.
Chimeric antigen receptor modified T-cells for cancer treatment

September 2018

·

790 Reads

·

23 Citations

Chronic Diseases and Translational Medicine

T cells engineered with the chimeric antigen receptor (CAR) are rapidly emerging as an important immunotherapy for hematologic malignancies. The anti-cluster of differentiation (CD)19 CAR-T cell therapy has been remarkably successful against refractory/relapsed acute lymphoblastic leukemia (ALL), and a complete remission rate as high as 90% was observed, in both children and adults. Although the achievement of clinical efficacy using CAR-T cell therapy for solid tumors has encountered several obstacles that were associated with the multiple mechanisms contributing to an immunosuppressive microenvironment, investigators are exploring more optimized approaches to improve the efficiency of CAR-T in solid tumors. In addition, cytokine release syndrome (CRS) and neurotoxicity following CAR-T cell therapy can be severe or even fatal; therefore, the management of these toxicities is significant. Herein, we briefly review the structure of CAR-T and some novel CAR designs, the clinical application of CAR-T cell therapies, as well as the assessment and management of toxicities.


Lentiviral vector and clinical protocol. a Design of the CAR-encoding lentiviral vector. b Protocol for CAR T-cell infusion and conditioning chemotherapy. c Characterization of lentiviral transduced T cells
Patient responses after infusion. a After infusion, the number of CAR copies in the peripheral blood continued to increase and reached their highest value on day 9; the number of CAR copies remained high even after the administration of methylprednisolone (200 mg/d for 3 days) and cyclophosphamide (0.8 g/d for 2 days). The number of CD19⁺ B cells was significantly reduced on day 8 after the infusion and remained low. b Response of the right axilla lymph nodes (arrow) according to CT scans at baseline and 3 weeks after anti-CD19 CAR T-cell infusion
Manifestations of cytokine release syndrome. a Serum cytokine levels were determined by a fluorescence-activated cell sorter. The fold changes are relative to the pre-infusion peripheral blood samples (baseline). IL-10, IL-6 and CRP levels were significantly increased when the T-cells proliferated rapidly. b Changes in liver function. AST and ALT levels were increased during the rapid proliferation of T cells
Kinetics and characteristics of abnormal T cells. a Changes in the white cell and lymphocyte counts in the peripheral blood. At day 10 after the infusion, the lymphocytes began to proliferate rapidly and could not be controlled by methylprednisolone. The lymphocyte levels peaked on day 13, with an absolute number of lymphocytes of 77 × 10⁹/L. The administration of cyclophosphamide ended the proliferation. On day 38, the lymphocytes rapidly proliferated again, and cyclophosphamide again ended the proliferation. b Flow cytometry was used to detect changes in cell composition and revealed that CD3⁺CD8⁺ cells made up the majority of lymphocytes. c Changes in the lymphocyte subsets. d Flow cytometry analyses of CD3⁺CD8⁺IFN⁺, CD3⁺CD8⁺GrB⁺ and CD3⁺CD8⁺Ki-67⁺ expression on T cells from the peripheral blood of the patient. On day 13 after the infusion, the T cells from this patient secreted IFN and granzyme at significantly higher levels than those at baseline. CD3⁺CD8⁺Ki-67⁺ represents T-cell proliferation. e Lung injury was discovered by computed tomography scans performed before the infusion and on day 18 and day 38 post infusion. The frosted glass appearance was diffusely distributed in the lungs on day 18 when the T-cell count in the peripheral blood was increased. f A bumpy red rash was found on the skin of the neck and chest. Pathologic examination of the skin rash showed that a small number of CD3⁺ and CD8⁺ cells were located in the dermis
Exploration of the mechanisms underlying excessive T-cell proliferation. a Peripheral blood and bone marrow smears stained with May-Grünwald-Giemsa solution (magnification, ×1000). A large number of naive lymphocytes and one mature lobulated granulocyte nucleus are shown in the peripheral blood. The naive lymphocytes are larger, cytoplasm-rich neutrophils with visible small particles, larger nuclei, an irregular cell shape and a biased side; they also have loose chromatin. In the bone marrow smears, a mix of immature and mature lymphocytes appear; the naive lymphocytes are visibly mitotic. b The polyclonal T-cell receptor (TCR, vβ) was identified in the peripheral blood by flow cytometry on day 13 after the infusion. c Karyotype analysis of bone marrow nucleated cells showed that 20 medium-sized mitotic cells were observed, and four abnormal karyotypes were found, including 46, XY, add (11) (p15)/46, idem (3) (p21)/47, XY, +2/46 and XY [16]. d The DNA of 12 types of viruses was screened by real-time PCR. CMV cytomegalovirus; EBV Epstein-Barr virus; HBV hepatitis B virus; JCV John Cunningham polyomavirus; BKPyV BK polyomavirus; VZV varicella-zoster virus; HSV herpes simplex virus; HHV human herpesvirus; B19V parvovirus B19; HCV hepatitis C virus
Excessive activated T-cell proliferation after anti-CD19 CAR T-cell therapy

June 2018

·

196 Reads

·

8 Citations

Gene Therapy

Excessive activated T-cell proliferation was observed in vivo in one patient after an anti-CD19-chimeric antigen receptor (CAR) T-cell infusion. The patient, who had chemotherapy refractory and CD19+diffuse large B-cell lymphoma (DLBCL), received an anti-CD19 CAR T-cell infusion following conditioning chemotherapy (fludarabine/cyclophosphamide). The lymphocyte count in the peripheral blood (PB) increased to 77 × 109/L on day 13 post infusion, and the proportion of CD8+actived T cells was 93.06% of the lymphocytes. Then, the patient suffered from fever and hypoxaemia. Significant increases in serum cytokine, lactate dehydrogenase, aspartate aminotransferase (AST), alanine transaminase (ALT), and glutamic-oxalacetic transaminase (γ-GT) levels were observed. A high-throughput sequencing analysis for T-cell receptors (TCRs) and whole-genome sequencing were used to explore the mechanisms underlying this excessive T-cell proliferation. TCR diversity was demonstrated, but no special gene mutation was found. The patient was found to be infected with the John Cunningham polyomavirus (JCV). It cannot be ruled out the bystander activation pathway induced by JCV infections related the excessive activated T-cell proliferation. Although the clinical and laboratory data do not fully explain the reason for excessive T-cell proliferation after the anti-CD19 CAR T-cell infusion, the risk of this type of toxicity should be emphasized. This study was registered at www.clinicaltrials.gov as NCT01864889.




Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma

January 2017

·

369 Reads

·

202 Citations

Journal of Hematology & Oncology

Background: Cholangiocarcinoma (CCA) is one of the most fatal malignant tumors with increasing incidence, mortality, and insensitivity to traditional chemo-radiotherapy and targeted therapy. Chimeric antigen receptor-modified T cell (CART) immunotherapy represents a novel strategy for the management of many malignancies. However, the potential of CART therapy in treating advanced unresectable/metastatic CCA is uncharted so far. Case presentation: In this case, a 52-year-old female who was diagnosed as advanced unresectable/metastatic CCA and resistant to the following chemotherapy and radiotherapy was treated with CART cocktail immunotherapy, which was composed of successive infusions of CART cells targeting epidermal growth factor receptor (EGFR) and CD133, respectively. The patient finally achieved an 8.5-month partial response (PR) from the CART-EGFR therapy and a 4.5-month-lasting PR from the CART133 treatment. The CART-EGFR cells induced acute infusion-related toxicities such as mild chills, fever, fatigue, vomiting and muscle soreness, and a 9-day duration of delayed lower fever, accompanied by escalation of IL-6 and C reactive protein (CRP), acute increase of glutamic-pyruvic transaminase and glutamic-oxalacetic transaminase, and grade 2 lichen striatus-like skin pathological changes. The CART133 cells induced an intermittent upper abdominal dull pain, chills, fever, and rapidly deteriorative grade 3 systemic subcutaneous hemorrhages and congestive rashes together with serum cytokine release, which needed emergent medical intervention including intravenous methylprednisolone. Conclusions: This case suggests that CART cocktail immunotherapy may be feasible for the treatment of CCA as well as other solid malignancies; however, the toxicities, especially the epidermal/endothelial damages, require a further investigation. Trial registration: ClinicalTrials.gov NCT01869166 and NCT02541370 .


Figure 2. hASCs rarely express CCR7. (A) Human CCR7 mRNA expression level in hASCs was analyzed by RT-PCR. hPBCs served as positive control. (B) Human CCR7 protein on cell surface of hASCs was detected by FCM technique, and hPBCs served as positive control. hASCs-human adipose-derived stem cells; hPBCs-human peripheral blood cells; CCR7-CC chemokine receptor 7; RT-PCR-reverse transcription-polymerase chain reaction; FCM-flow cytometry; *** p<0.001. 
Figure 3. Rat CCR7 gene was introduced into hASCs. (A) Plasmid structure exploited for packaging lentivirus and WPRE is an element in the plasmid. (B) The rat CCR7 gene was inserted into the control plasmid of (A). (C) WPRE mRNA level in rCCR7-hASCs and GFP-hASCs was examined by RT-PCR, rPBCs and hASCs served as negative control. (D) Rat CCR7 mRNA level in GFP-hASCs and rCCR7-hASCs. rPBCs served as positive control and hASCs served as negative control. (E) Rat CCR7 protein expression on cells detected by FCM technique. hASCs-human adipose-derived stem cells; rPBCs-rat peripheral blood cells; CCR7-CC chemokine receptor 7; GFP-green fluorescent protein; WPRE-WoodchUck hepatitis post-transcriptional regulatory element; RT-PCR-reverse transcription-polymerase chain reaction; FCM-flow cytometry; *** p<0.001. 
Figure 6. rCCR7 enabled hASCs to migrate to SLOs efficiently. GFP-hASCs (1.5×10 4 /g) or rCCR7-hASCs (1.5×10 4 /g) were intravenously injected into Lewis rats. Rats injected with PBS only served as negative control. At 3 days after transfusion, the GFP cells in the rat spleen and lymphoid nodes were detected by FCM. (A) Rate of GFP-positive cells in spleen. (B) Rate of GFP-positive cells in lymphoid nodes. (C) Representative FCM result in A and B. SLOs-secondary lymphoid organs; hASCs-human adipose-derived stem cells; CCR7-CC chemokine receptor 7; GFP-green fluorescent protein; PBS-phosphate-buffered saline; G-A injection-GFP-hASCs-injected rat; C-A injection-rCCR7-hASCs-injected rat; FCM-flow cytometry; * P<0.05; ** P<0.01. 
Upregulation of CC Chemokine Receptor 7 (CCR7) Enables Migration of Xenogeneic Human Adipose-Derived Mesenchymal Stem Cells to Rat Secondary Lymphoid Organs

December 2016

·

61 Reads

·

9 Citations

Medical Science Monitor: International Medical Journal of Experimental and Clinical Research

Background CC chemokine receptor 7 (CCR7) expression is vital for cell migration to secondary lymphoid organs (SLOs). Our previous work showed that inducing CCR7 expression enabled syngeneic mesenchymal stem cells (MSCs) to migrate into SLOs, resulting in enhanced immunosuppressive performance in mice. Given that human adipose-derived stem cells (hASCs) are widely used in clinical therapy, we further investigated whether upregulation of CCR7 enables xenogeneic hASCs to migrate to rat SLOs. Material/Methods hASCs rarely express CCR7; therefore, hASCs were transfected with lentivirus encoding rat CCR7 (rCCR7) plus green fluorescence protein (GFP) or GFP alone. CCR7 mRNA and cell surface expression of rCCR7-hASCs and GFP-hASCs were examined by reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry (FCM), respectively. The phenotype, differentiation, and proliferation capacity of each cell type was also determined. To examine migration, rCCR7-hASCs and GFP-hASCs were injected intravenously into Lewis rats, and the proportion of GFP-positive cells in the spleen and lymph nodes was determined with FCM. Results mRNA and cell surface protein expression of CCR7 was essentially undetectable in hASCs and GFP-ASCs; however, CCR7 was highly expressed in rCCR7-ASCs. rCCR7-hASCs, GFP-hASCs, and hASCs shared a similar immunophenotype, and maintained the ability of multilineage differentiation and proliferation. In addition, the average proportion of GFP-positive cells was significantly higher following transplantation of rCCR7-hASCs compared with GFP-hASCs (p<0.01). Conclusions These results suggest that upregulation of rat CCR7 expression does not change the phenotype, differentiation, or proliferation capacity of hASCs, but does enable efficient migration of hASCs to rat SLOs.


Mesenchymal stem cells in diabetes treatment: Progress and perspectives

July 2016

·

17 Reads

Medical Journal of Chinese People's Liberation Army

Diabetes is a chronic metabolic disorder caused by relative or absolute insulin deficient or reduced sensitivity of target cells to insulin. Mesenchymal stem cells (MSCs) are adult stem cells with multiple differentiation potential, self-renewable and immunoregulatory properties. Accumulating evidences from clinic or animal experiments recent years showed that MSCs infusion could ameliorate hyperglycemia in diabetes. The research progress of MSCs in diabetes treatment is summarized and a corresponding perspective is herewith proposed in present paper. DOI: 10.11855/j.issn.0577-7402.2016.07.16


Lipopolysaccharides priming mesenchymal stem cells accelerate diabetic wound healing via exosomes

July 2016

·

98 Reads

Medical Journal of Chinese People's Liberation Army

Objective To study the therapeutic effect of exosome derived from lipopolysaccharides (LPS) priming mesenchymal stem cells (MSCs) for diabetic wound healing. Methods Human umbilical cord MSCs were treated with LPS (100ng/ml) for 2 days, the supernatant were then collected, and exosomes were harvested by density gradient centrifugation and identified. Diabetic cutaneous wounds were prepared and the animals were divided into the following three groups: control group, untreated MSCs derived exosome (un-exosome) treatment group and LPS primed MSCs derived exosome (LPS-exosome) treatment group. Exosomes (60μg) were injected dispersively into the wound edge daily for 10 days. After treatment, the therapeutic results were evaluated by gross observation of the wounds, the expression levels of inflammation related factors and macrophage subtype markers in the injured sites were detected by qRT-PCR at day 3, 7 and 14 after treatment. Results Compared with control group, the diabetic wound healing was obviously improved in LPS-exosome treatment group after treatment for 7 and 14 days, with faster wound close, depressed expression of pro-inflammatory factors IL-1, IL-12 and M1 macrophage surface marker iNOS, and upregulation of anti-inflammatory factors IL-10, TGF-β and M2 macrophage surface marker CD163, the differences were significant (P<0.05). Conclusions LPS-exosome may balance macrophage plasticity, restrain chronic inflammation and accelerate diabetic cutaneous wound healing. © 2016, People’s Military Medical Press. All rights reserved.


Fig.3Effect of hUC-MSCs cultured under normoxia and hypoxia on macrophage polarization Representative photomicrographs of macrophage after cocultivation for 24h stained for iNOS (green) and Arg-1(red).
Hypoxic pretreatment of human umbilical cord mesenchymal stem cells regulating macrophage polarization

July 2016

·

84 Reads

Medical Journal of Chinese People's Liberation Army

Objective To investigate the effect of human umbilical cord mesenchymal stem cells (hUC-MSCs) on macrophage polarization under hypoxia. Methods hUC-MSCs were obtained by explants adherent culture and cultured under normoxia (21% O2) and hypoxia (5% O2). The multi-directional differentiation of hUC-MSCs was observed by osteogenic and adipogenic differentiation induction. Live/death staining was performed to detect the cell viability, and ELISA was executed to detect the protein content in supernatant of hUC-MSCs. Transwell chamber was employed to co-culture the hUC-MSCs cultured under normoxia and hypoxia and macrophage (THP-1) stimulated by lipopolysaccharide (IPS), then the polarization of THP-1 was detected by immunofluorescence, and the secretions of inflammatory factor and anti-inflammatory factor of THP-1 were detected by ELISA. Results hUC-MSCs cultured under hypoxia showed the ability of osteogenic and adipogenic multi-directional differentiation. Live/death staining showed the high cell viability of hUC-MSCs cultured under normoxia and hypoxia. The expression levels of prostaglandin E2 (PGE2) and indoleamine 2,3-dioxygenase (IDO) were significantly higher in the hUC-MSCs cultured under hypoxia than in those cultured under normoxia. hUCMSCs cultured under hypoxia promoted the polarization of THP-1 to M2, obviously reduced the expression of TNF-α and IL-1β, and increased the expression of IL-10 significantly. Conclusion hUC-MSCs cultured under hypoxia may promote the polarization of THP-1 to M2 and improve the viability of anti-inflammatory. DOI: 10.11855/j.issn.0577-7402.2016.07.01


Citations (35)


... T cells play a critical and indispensable role in the adaptive immune response against cancer cells, as they possess the extraordinary ability to recognize and eliminate malignant cells (13,14). TCRs are essential for initiating T-cell-mediated antitumor functions because they trigger immune responses, including cytokine production, that impede tumor growth (14,15). ...

Reference:

Characterization of the T-cell receptor repertoire associated with lymph node metastasis in colorectal cancer
Genetically engineered T cells for cancer immunotherapy

Signal Transduction and Targeted Therapy

... [9] These and other promising results led to the approval of tisagenlecleucel (Kymriah® by Novartis Pharmaceutical) and axicabtagene ciloleucel (Yescarta® by Kite Pharma) for treatment of patients with R/R B-ALL and relapsed large B cell Non-Hodgkin lymphoma (NHL) respectively, in 2017. [10] Recently two additional anti CD19 products Breyanzi® (lisocabtagene maraleucel), for large B-cell lymphomas and Tecartus® (brexucabtagene autoleucel, formerly KTE-X19), for Mantle cell lymphoma have also been approved. Abecma® (Idecabtagene vicleucel), the first anti-BCMA CAR-T product for relapsed and refractory Multiple Myeloma from Bristol Myers Squibb's and Bluebird Bio has been approved by the FDA. ...

Chimeric antigen receptor modified T-cells for cancer treatment

Chronic Diseases and Translational Medicine

... This method can be used to quantify cell numbers early after injection. However, it is important to point out that this method is not as reliable for quantifying CAR-T cell numbers over time, as CAR-T cells have been shown to proliferate significantly after CAR interaction with their respective antigen [36]. During cell division, the PFC label should be divided between daughter cells. ...

Excessive activated T-cell proliferation after anti-CD19 CAR T-cell therapy

Gene Therapy

... The CAR-T treatment of (2) B-NHL also includes other special adverse effects, including capillary leakage syndrome, impaired respiratory function, gastrointestinal bleeding, oncolytic syndrome and other [11][12][13]. ...

Long-term safety and efficacy of CART-20 cells in patients with refractory or relapsed B-cell non-Hodgkin lymphoma: 5-years follow-up results of the phase I and IIa trials

Signal Transduction and Targeted Therapy

... While this technology has been highly effective in treating certain hematological malignancies, its application in solid tumors like ICC is challenging due to the lack of effective targets and significant tumor heterogeneity [133] . Recent studies, however, indicate that combining CAR-T cell therapy with PD-1 or PD-L1 inhibitors can significantly enhance the antitumor effects in solid tumors, including ICC [134] . For instance, Feng et al. observed clinical remission in some patients with advanced ICC treated with CAR-T cells targeting EGFR and CD133 [135] . ...

Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma

Journal of Hematology & Oncology

... In a further study, we tested the expression of CC chemokine receptor 1 (CCR1) and CXC chemokine receptor 4 (CXCR4) of cryopreserved UCMSCs that were closely related to migration and distribution [41][42][43]. As shown in Figure 8A,B, compared to fresh UCM-SCs, the recovered UCMSCs from B-E cryopreservation showed normal expression of these receptors. ...

Upregulation of CC Chemokine Receptor 7 (CCR7) Enables Migration of Xenogeneic Human Adipose-Derived Mesenchymal Stem Cells to Rat Secondary Lymphoid Organs

Medical Science Monitor: International Medical Journal of Experimental and Clinical Research

... MicroRNAs (miRNAs) are a group of small, single-stranded noncoding RNAs that have significant involvement in the control of gene expression by translational repression or degradation of target mRNA. These miRNAs have a profound impact on essential activities in a wide range of physiological processes [10][11][12]. Dysregulation of microRNAs (miRNAs) has been implicated in a diverse array of human disorders and can exert influence on the initiation and advancement of tumors through modulation of proto-oncogenes and tumor suppressor genes [13,14]. Current research findings indicate that the manifestation of microRNAs (miRNAs) is correlated with the progression or prognostication of colorectal cancer (CRC) [15,16]. ...

MicroRNAs: Novel immunotherapeutic targets in colorectal carcinoma

World Journal of Gastroenterology

... The remaining two patients showed stable disease (SD). The median progression-free survival (PFS) time was 6 months, and these data were obtained after a median follow-up period of 8 months (144). ...

Treatment of CD20-directed Chimeric Antigen Receptor-modified T cells in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: an early phase IIa trial report

Signal Transduction and Targeted Therapy

... In vitro transcription/translation and GST pull-down assays GST and GST fusion proteins were prepared as described previously [23,53]. 35 S-labeled proteins were produced by using a TNT-coupled in vitro transcription and translation (IVT) system (Promega), with the expression vectors for p65 and its derivatives, p50 and c-Rel in pcDNA3 or pcDNA3.1. ...

FHL2 interacts with and acts as a functional repressor of Id2 in human neuroblastoma cells
  • Citing Article
  • June 2009

Chinese Journal of Cancer Research

... It was a remarkable fact that the anti-CD30 CAR-T cell therapy had no toxicity in this study (23) . Furthermore, Wang et al. (24) performed anti-CD30 CAR-T cell therapy on 11 patients with r/r HL, infusing CAR-T cells per kg of weight. It was indicated that 9 cases (82%) responded to treatment, 1 case (9%) maintained continuous CR, 1 case (46%) achieved PR, and 3 cases (27%) were stable. ...

Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin's lymphoma: an open-label phase 1 trial
  • Citing Article
  • October 2015

The Lancet