Review for "CXCR2‐modified CAR‐T cells have enhanced trafficking ability that improves treatment of hepatocellular carcinoma"

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A121-A121
Author(s):  
Nina Chu ◽  
Michael Overstreet ◽  
Ryan Gilbreth ◽  
Lori Clarke ◽  
Christina Gesse ◽  
...  

BackgroundChimeric antigen receptors (CARs) are engineered synthetic receptors that reprogram T cell specificity and function against a given antigen. Autologous CAR-T cell therapy has demonstrated potent efficacy against various hematological malignancies, but has yielded limited success against solid cancers. MEDI7028 is a CAR that targets oncofetal antigen glypican-3 (GPC3), which is expressed in 70–90% of hepatocellular carcinoma (HCC), but not in normal liver tissue. Transforming growth factor β (TGFβ) secretion is increased in advanced HCC, which creates an immunosuppressive milieu and facilitates cancer progression and poor prognosis. We tested whether the anti-tumor efficacy of a GPC3 CAR-T can be enhanced with the co-expression of dominant-negative TGFβRII (TGFβRIIDN).MethodsPrimary human T cells were lentivirally transduced to express GPC3 CAR both with and without TGFβRIIDN. Western blot and flow cytometry were performed on purified CAR-T cells to assess modulation of pathways and immune phenotypes driven by TGFβ in vitro. A xenograft model of human HCC cell line overexpressing TGFβ in immunodeficient mice was used to investigate the in vivo efficacy of TGFβRIIDN armored and unarmored CAR-T. Tumor infiltrating lymphocyte populations were analyzed by flow cytometry while serum cytokine levels were quantified with ELISA.ResultsArmoring GPC3 CAR-T with TGFβRIIDN nearly abolished phospho-SMAD2/3 expression upon exposure to recombinant human TGFβ in vitro, indicating that the TGFβ signaling axis was successfully blocked by expression of the dominant-negative receptor. Additionally, expression of TGFβRIIDN suppressed TGFβ-driven CD103 upregulation, further demonstrating attenuation of the pathway by this armoring strategy. In vivo, the TGFβRIIDN armored CAR-T achieved superior tumor regression and delayed tumor regrowth compared to the unarmored CAR-T. The armored CAR-T cells infiltrated HCC tumors more abundantly than their unarmored counterparts, and were phenotypically less exhausted and less differentiated. In line with these observations, we detected significantly more interferon gamma (IFNγ) at peak response and decreased alpha-fetoprotein in the serum of mice treated with armored cells compared to mice receiving unarmored CAR-T, demonstrating in vivo functional superiority of TGFβRIIDN armored CAR-T therapy.ConclusionsArmoring GPC3 CAR-T with TGFβRIIDN abrogates the signaling of TGFβ in vitro and enhances the anti-tumor efficacy of GPC3 CAR-T against TGFβ-expressing HCC tumors in vivo, proving TGFβRIIDN to be an effective armoring strategy against TGFβ-expressing solid malignancies in preclinical models.Ethics ApprovalThe study was approved by AstraZeneca’s Ethics Board and Institutional Animal Care and Use Committee (IACUC).


2021 ◽  
Vol 11 ◽  
Author(s):  
Wei Jiang ◽  
Tao Li ◽  
Jiaojiao Guo ◽  
Jingjing Wang ◽  
Lizhou Jia ◽  
...  

T cells expressing chimeric antigen receptors, especially CD19 CAR-T cells have exhibited effective antitumor activities in B cell malignancies, but due to several factors such as antigen escape effects and tumor microenvironment, their curative potential in hepatocellular carcinoma has not been encouraging. To reduce the antigen escape risk of hepatocellular carcinoma, this study was to design and construct a bispecific CAR targeting c-Met and PD-L1. c-Met/PD-L1 CAR-T cells were obtained by lentiviral transfection, and the transfection efficiency was monitored by flow cytometry analysis. LDH release assays were used to elucidate the efficacy of c-Met/PD-L1 CAR-T cells on hepatocellular carcinoma cells. In addition, xenograft models bearing human hepatocellular carcinoma were constructed to detect the antitumor effect of c-Met/PD-L1 CAR-T cells in vivo. The results shown that this bispecific CAR was manufactured successfully, T cells modified with this bispecific CAR demonstrated improved antitumor activities against c-Met and PD-L1 positive hepatocellular carcinoma cells when compared with those of monovalent c-Met CAR-T cells or PD-L1 CAR-T cells but shown no distinct cytotoxicity on hepatocytes in vitro. In vivo experiments shown that c-Met/PD-L1 CAR-T cells significantly inhibited tumor growth and improve survival persistence compared with other groups. These results suggested that the design of single-chain, bi-specific c-Met/PD-L1 CAR-T is more effective than that of monovalent c-Met CAR-T for the treatment of hepatocellular carcinoma., and this bi-specific c-Met/PD-L1 CAR is rational and implementable with current T-cell engineering technology.


2020 ◽  
Vol Volume 13 ◽  
pp. 5707-5708
Author(s):  
Hezhi Wang ◽  
Xueshuai Ye ◽  
Yi Ju ◽  
Ziqi Cai ◽  
Xiaoxiao Wang ◽  
...  

2020 ◽  
Vol 50 (5) ◽  
pp. 712-724 ◽  
Author(s):  
Guangna Liu ◽  
Wei Rui ◽  
Hongli Zheng ◽  
Daosheng Huang ◽  
Fei Yu ◽  
...  

2017 ◽  
Vol 7 ◽  
Author(s):  
Zhiwu Jiang ◽  
Xiaofeng Jiang ◽  
Suimin Chen ◽  
Yunxin Lai ◽  
Xinru Wei ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A149-A149
Author(s):  
Yuan Qin ◽  
Anna Qin ◽  
Anna Musket ◽  
Joseph Lee ◽  
Zhi Yao ◽  
...  

BackgroundHepatocellular carcinoma (HCC) is the leading cause of cancer mortality worldwide. While HBV/HCV infection is the primary cause of HCC, overexpression of MET, the receptor of hepatocyte growth factor (HGF), occurs in 50% HCC patients, and is an indicator of poor prognosis. Although the multi-target MET tyrosine kinase inhibitor cabozantinib is FDA approved for treating advanced HCC, the long-term efficacy versus toxicity remains unknown. Our study is to develop specific MET-targeting chimeric antigen receptor T (CAR-T) cells for treating HCC with MET overexpression.MethodsBased on a well-established anti-MET monoclonal antibody, we synthesized and cloned the single-chain variable fragment (ScFv) sequence into two retroviral based 2nd generation CAR vectors (MET-CAR.CD28.ζ. and MET-CAR.4-1BB.ζ.). A MET-CAR without CD3ζ domain (MET-CARΔ) served as a negative control. To produce MET-CAR-T cells, healthy PBMCs were stimulated with anti-CD3/CD28 antibodies in the presence of IL-7/IL-15 followed by transduction with MET-CAR viral particles. T cell transduction efficacy was determined using flow cytometry. HCC cell lines with variable MET expression from high/positive (MHCC97H, C3A, and JHH5) to MET low/negative (SNU398) were used to determine MET-specific CAR T cells specificity and effector function using MTS assay. We also collected media from the tumor-T cell co-cultures and determined IL-2 and IFNγ secretion using ELISA. Finally, real-time confocal imaging (24 h) was performed to record the progress of MET-CAR T cell mediated killing activity against MHCC97H/mCherry cells.ResultsWe show that both MET-CAR.CD28.ζ and MET-CAR.4-1BB.ζ -T cells significantly killed MHCC97H, C3A, and JHH5 cells in antigen dependent manner. MET-CAR T cell killing is MET dependent as we observed no killing of MET-negative SNU398 cells. In addition, MET-CAR.4-1BB.ζ and MET-CAR.CD28.ζ- T cells secreted IL-2 and IFNγ when co-cultured with MHCC97H, C3A, JHH5 cells, but not SNU398. Confocal imaging studies showed that both MET-specific CAR T cells migrated toward MHCC97H/mCherry cells, formed aggregations, and induced tumor cell death, while MET-CARΔ T cells failed to do so.ConclusionsHere we demonstrate that MET-CAR.4-1BB.ζ and MET-CAR.CD28.ζ- T cells specifically recognize and kill MET-positive HCC cells in vitro. While animal studies are required to validate the efficacy in vivo, our study has produced a novel therapeutic CAR T cell target for treating malignant HCC and other type of cancers with MET overexpression.AcknowledgementsThis independent research was supported by the Gilead Sciences Research Scholars Program in Liver Disease- The Americas, and Department of Defense (DoD) Ideal Award (to QX)Ethics ApprovalThe study was approved by East Tennessee State University’s Ethics Board, approval number #0619.3s.


Sign in / Sign up

Export Citation Format

Share Document