Cell intrinsic PD-1 checkpoint blockade releases the brake on human chimeric antigen receptor (CAR) T cells for solid tumors

2016 ◽  
Vol 5 (S7) ◽  
pp. S1328-S1332 ◽  
Author(s):  
Joseph A. Fraietta ◽  
J. Joseph Melenhorst ◽  
Bruce L. Levine
2021 ◽  
Author(s):  
tian chi ◽  
yan zou

Chimeric antigen receptor (CAR) T cell therapy has been successful in treating hematological malignancy, but solid tumors remain refractory. Here, we demonstrated that knocking out transcription factor IKZF3 in HER2-specific CAR T cells targeting breast cancer cells did not affect proliferation or differentiation of the CAR T cells in the absence of tumors, but markedly enhanced killing of the cancer cells in vitro and in a xenograft model. Furthermore, IKZF3 KO had similar effects on the CD133-specific CAR T cells targeting glioblastoma cells. AlphaLISA and RNA-seq analyses indicate that IKZF3 KO increased the expression of genes involved in cytokine signaling, chemotaxis and cytotoxicity. Our results suggest a general strategy for enhancing CAR T efficacy on solid tumors.


Cancers ◽  
2020 ◽  
Vol 12 (10) ◽  
pp. 2818
Author(s):  
Anna Pasetto

In the recent years, immunotherapy has achieved impressive results utilizing different approaches, from (chimeric antigen receptor) CAR-T cells directed against CD19 for the treatment of leukemia and lymphomas to the Nobel prize-winning strategy of check-point inhibition for the treatment of several solid tumors [...]


Cancers ◽  
2019 ◽  
Vol 11 (2) ◽  
pp. 191 ◽  
Author(s):  
Benjamin Heyman ◽  
Yiping Yang

Chimeric antigen receptor T cells (CAR T Cells) have led to dramatic improvements in the survival of cancer patients, most notably those with hematologic malignancies. Early phase clinical trials in patients with solid tumors have demonstrated them to be feasible, but unfortunately has yielded limited efficacy for various cancer types. In this article we will review the background on CAR T cells for the treatment of solid tumors, focusing on the unique obstacles that solid tumors present for the development of adoptive T cell therapy, and the novel approaches currently under development to overcome these hurdles.


2020 ◽  
Author(s):  
Le Qin ◽  
Ruocong Zhao ◽  
Dongmei Chen ◽  
Xinru Wei ◽  
Qiting Wu ◽  
...  

Abstract Background: Chimeric antigen receptor T cells (CAR-T cells) therapy has been well recognized for treating B cell-derived malignancy. However, the efficacy of CAR-T cells against solid tumors remains dissatisfactory, partially due to the heterogeneity of solid tumors and T cell exhaustion in tumor microenvironment. PD-L1 is up-regulated in multiple solid tumors, resulting in T cell exhaustion upon binding to its receptor PD-1. Methods: Here, we designed a dominant-negative form of PD-1 , dPD1z, a vector containing the extracellular and transmembrane regions of human PD-1, and a CAR vector against PD-L1, CARPD-L1z, a vector employs a high-affinity single-chain variable fragment (scFv) against human PD-L1. These two vectors shared the same intracellular structure, including 4-1BB and TLR2 co-stimulatory domains, and the CD3ζ signaling domain. Results: dPD1z T and CARPD-L1z T cells efficiently lysed PD-L1 + tumor cells and had enhanced cytokine secretion in vitro and suppressed the growth of non-small cell lung cancer (NSCLC), gastric cancer and hepatoma carcinoma in patient-derived xenograft (PDX). However, the combination of anti-mesothelin CAR-T cells (CARMSLNz T) with dPD1z T or CARPD-L1z T cells did not repress tumor growth synergistically in PDX, as CARMSLNz T cells upregulated PD-L1 expression upon activation and were subsequently attacked by dPD1z T or CARPD-L1z T cells. Conclusions: In conclusion, we demonstrate CAR-T cells targeting PD-L1 were effective for suppressing the growth of multiple types of solid tumors in PDX models though their safety needs to be carefully examined.


2020 ◽  
Vol 8 (1) ◽  
Author(s):  
Qibin Liao ◽  
Huan He ◽  
Yunyu Mao ◽  
Xiangqing Ding ◽  
Xiaoyan Zhang ◽  
...  

Abstract Chimeric antigen receptor-modified T cells (CAR-T cells) have shown good effects in the treatment of hematologic cancers; however, they may cause on-target off-tumor toxicity because of minimal expression of tumor-associated antigens (TAAs) on normal tissues, particularly in the context of treating solid tumors. Hypoxia is a common hallmark of solid tumors because of the Warburg effect. To minimize side effects, we designed a hypoxia-inducible CAR (HiCAR), which is driven by a hypoxia response element (HRE), and consists of a conventional CAR and an oxygen-dependent degradation domain (ODD) that is actively degraded under normoxia but stabilized under hypoxia. HiCAR-T cells showed enhanced cytotoxicity against tumor cells under hypoxia compared to normoxia in vitro and antitumor efficacy comparable to that of conventional CAR-T cells in vivo. Overall, our study demonstrates the potential of the HiCAR for improving the safety of CAR-T cells to promote the clinical application of CAR-T immunotherapy.


Author(s):  
Zhixiong Wang ◽  
Qian Liu ◽  
Na Risu ◽  
Jiayu Fu ◽  
Yan Zou ◽  
...  

Chimeric antigen receptor (CAR) T cell therapy still faces the challenge of immunosuppression when treating solid tumors. TGF-β is one of the critical factors in the tumor microenvironment to help tumors escape surveillance by the immune system. Here we tried using the combination of a small molecule inhibitor of TGF-β receptor I, Galunisertib, and CAR T cells to explore whether Galunisertib could enhance CAR T cell function against solid tumor cells. In vitro experiments showed Galunisertib could significantly enhance the specific cytotoxicity of both CD133- and HER2-specific CAR T cells. However, Galunisertib had no direct killing effect on target cells. Galunisertib significantly increased the cytokine secretion of CAR T cells and T cells that do not express CAR (Nontransfected T cells). Galunisertib did not affect the proliferation of T cells, the antigen expression on target cells and CD69 on CAR T cells. We found that TGF-β was secreted by T cells themselves upon activation, and Galunisertib could reduce TGF-β signaling in CAR T cells. Our findings can provide the basis for further preclinical and clinical studies of the combination of Galunisertib and CAR T cells in the treatment of solid tumors.


2021 ◽  
Vol 13 (578) ◽  
pp. eaba7308
Author(s):  
Huihui Zhang ◽  
Fanlin Li ◽  
Jiang Cao ◽  
Xin Wang ◽  
Hai Cheng ◽  
...  

Although chimeric antigen receptor (CAR)–modified T cells have shown great success in the treatment of B cell malignancies, this approach has limited efficacy in patients with solid tumors. Various modifications in CAR structure have been explored to improve this efficacy, including the incorporation of two costimulatory domains. Because costimulatory signals are transduced together with T cell receptor signals during T cell activation, we engineered a type of CAR-T cells with a costimulatory signal that was activated independently from the tumor antigen to recapitulate physiological stimulation. We screened 12 costimulatory receptors to identify OX40 as the most effective CAR-T function enhancer. Our data indicated that these new CAR-T cells showed superior proliferation capability compared to current second-generation CAR-T cells. OX40 signaling reduced CAR-T cell apoptosis through up-regulation of genes encoding Bcl-2 family members and enhanced proliferation through increased activation of the NF-κB (nuclear factor κB), MAPK (mitogen-activated protein kinase), and PI3K-AKT (phosphoinositide 3-kinase to the kinase AKT) pathways. OX40 signaling not only enhanced the cytotoxicity of CAR-T cells but also reduced exhaustion markers, thereby maintaining their function in immunosuppressive tumor microenvironments. In mouse tumor models and in patients with metastatic lymphoma, these CAR-T cells exhibited robust amplification and antitumor activity. Our findings provide an alternative option for CAR-T optimization with the potential to overcome the challenge of treating solid tumors.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A134-A134
Author(s):  
Jessica Lake ◽  
Kevin Winkler ◽  
Alexander Harrant ◽  
Ashley Yingst ◽  
Kristin Schaller ◽  
...  

BackgroundThe 5-year disease-free survival for children and young adults with metastatic sarcoma at diagnosis or recurrent disease after front-line therapy is 20–30%.1 2 Cellular immunotherapy using chimeric antigen receptor (CAR) T cells has shown dramatic benefits in leukemia, but only limited success in solid tumors.3 4 One limitation of CAR T cell therapy has been poor trafficking into solid tumors.5–7 Chemokines are small, secreted, cytokine-like molecules that mediate lymphocyte homing and migration.8 In this study, we discovered that both osteosarcoma (OS) and rhabdomyosarcoma (RMS) cells significantly increase expression of the chemokine IL-8 after clinically achievable doses of radiation, but not at rest. Given that CAR T cells do not express the receptor for IL-8, we created a construct with an IL-8 receptor (CXCR2) and a B7H3 CAR in T cells to improve CAR T homing and to create an effective new immunotherapy for patients with sarcoma.MethodsMultiple OS and RMS cell lines were irradiated at 10 Gy and IL-8 was measured by ELISA. We created retroviral constructs, B7H3 CAR-T2a-CXCR2 and B7H3 CAR. Peripheral blood T lymphocytes were stimulated with IL-2 and anti-CD3/28 antibodies for 48 hours prior to transduction with the retroviral vectors. Surface expression of the scFv (by L protein) and CXCR2 (mAb) were assessed using flow cytometry. In vitro cytotoxicity assays using sarcoma tumor spheroids were conducted using Incucyte. INF-γ and IL-2 production were measured by ELISA. NSG mice injected orthotopically with an IL-8 overexpressing RMS cell line were treated 4–7 days later with the B7H3 CAR-CXCR2 T cells or B7H3 T cells (control) and followed weekly with bioluminescent imaging.ResultsIrradiated (10 Gy) sarcoma cells express 2-9x higher IL-8 than non-irradiated sarcoma. T cells were transduced with efficiencies of 60–90%. INF-γ production was equivalent between the B7H3 CAR-T2a-CXCR2 T cells and B7H3 CAR T cells, but IL-2 production was significantly higher in the dual expressing CAR T cells. In vitro cytotoxicity with sarcoma spheroids was measured by Incucyte and showed faster and greater killing by B7H3 CAR-T2a-CXCR2 T cells than B7H3 CAR T cells. Furthermore, when sarcoma tumor bearing mice were treated with B7H3 CAR-T2a-CXCR2 T cells, tumors resolved completely by 4–5 weeks and had long-lasting remission.ConclusionsChemokine receptor expressing CAR T cells showed superior cytokine production and T cell activation/cytotoxicity compared to a CAR T construct alone. These finding lead to better efficacy in animal models and suggest a promising approach for pediatric sarcoma.ReferencesLuetke A, Meyers PA, Lewis I, Juergens H. Osteosarcoma treatment - where do we stand? A state of the art review. Cancer Treat Rev 2014;40:523–32.Bleyer A, Barr R, Hayes-Lattin B, et al. The distinctive biology of cancer in adolescents and young adults. Nat Rev Cancer 2008;8:288–98.Buechner J, SA G, SL M, et al. Global Registration Trial of Efficacy and Safety of CTL019 in Pediatric and Young Adult Patients with Relapsed/Refractory (R/R) Acute Lymphoblastic Leukemia (ALL): Update to the interim analysis. 2017 European Hematology Association Annual Meeting: Madrid, Spain2017.Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med 2018;378:439–48.Gill S, Maus MV, Porter DL. Chimeric antigen receptor T cell therapy: 25 years in the making. Blood Rev 2016;30:157–67.Fousek K, Ahmed N. The Evolution of T-cell Therapies for Solid Malignancies. Clin Cancer Res 2015;21:3384–92.Newick K, Moon E, Albelda SM. Chimeric antigen receptor T-cell therapy for solid tumors. Mol Ther Oncolytics 2016;3:16006.Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol 2017;17:559–72.Ethics ApprovalThe animal experiments discussed in the abstract were approved by the University of Colorado IACUC, protocol #00251.


2020 ◽  
Author(s):  
Le Qin ◽  
Ruocong Zhao ◽  
Dongmei Chen ◽  
Xinru Wei ◽  
Qiting Wu ◽  
...  

Abstract Background: Chimeric antigen receptor T cells (CAR-T cells) therapy has been well recognized for treating B cell-derived malignancy. However, the efficacy of CAR-T cells against solid tumors remains dissatisfactory, partially due to the heterogeneity of solid tumors and T cell exhaustion in tumor microenvironment. PD-L1 is up-regulated in multiple solid tumors, resulting in T cell exhaustion upon binding to its receptor PD-1. Methods: Here, we designed a dominant-negative form of PD-1 , dPD1z, a vector containing the extracellular and transmembrane regions of human PD-1, and a CAR vector against PD-L1, CARPD-L1z, a vector employs a high-affinity single-chain variable fragment (scFv) against human PD-L1. These two vectors shared the same intracellular structure, including 4-1BB and TLR2 co-stimulatory domains, and the CD3ζ signaling domain. Results: dPD1z T and CARPD-L1z T cells efficiently lysed PD-L1 + tumor cells and had enhanced cytokine secretion in vitro and suppressed the growth of non-small cell lung cancer (NSCLC), gastric cancer and hepatoma carcinoma in patient-derived xenograft (PDX). However, the combination of anti-mesothelin CAR-T cells (CARMSLNz T) with dPD1z T or CARPD-L1z T cells did not repress tumor growth synergistically in PDX, as CARMSLNz T cells upregulated PD-L1 expression upon activation and were subsequently attacked by dPD1z T or CARPD-L1z T cells. Conclusions: In conclusion, we demonstrate CAR-T cells targeting PD-L1 were effective for suppressing the growth of multiple types of solid tumors in PDX models though their safety needs to be carefully examined.


Sign in / Sign up

Export Citation Format

Share Document