scholarly journals GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade

2016 ◽  
Vol 24 (6) ◽  
pp. 1135-1149 ◽  
Author(s):  
Tessa Gargett ◽  
Wenbo Yu ◽  
Gianpietro Dotti ◽  
Eric S Yvon ◽  
Susan N Christo ◽  
...  
Author(s):  
Gaia Zuccolotto ◽  
Alessandro Penna ◽  
Giulio Fracasso ◽  
Isabella Monia Montagner ◽  
Debora Carpanese ◽  
...  

Despite advances in the understanding of its molecular pathophysiology, prostate cancer remains largely incurable, highlighting the need for novel therapies. We developed a chimeric antigen receptor (CAR) specific for prostate specific membrane antigen (PSMA), a glycoprotein that is overexpressed in prostate cancer, which expression involves neovasculature of several tumor entities, thus envisaging an additional antiangiogenic effect. To optimize the CAR design, we compared two CARs with signaling domains containing one or two T cell costimulatory elements, in addition to CD3ζ. Conversely, what has been described for other CARs, a third-generation CAR (containing CD28 and 41BB co-signaling domains) induced a potent antitumor effect similar to a second-generation CAR (containing CD28 co-signaling domain), though we observed a detrimental effect of the additional costimulatory domain that was attributed to increased activation-induced cell death (AICD). This “super-stimulation” resulted in exhaustion of cells, higher frequencies of cell death and, more importantly, the impossibility of sufficiently expanding the CAR cells to obtain the minimum number of cells requested for in vivo therapies. While the superiority of 2nd and 3rd generation over 1st generation CAR T cells has been clearly shown in both preclinical and clinical studies, the optimal combination of costimulatory domains for 3rd generation CAR-T cells must still be defined and should be evaluated case-by-case in order to fine-tune immunotherapy approaches.


2021 ◽  
Vol 11 ◽  
Author(s):  
Gaia Zuccolotto ◽  
Alessandro Penna ◽  
Giulio Fracasso ◽  
Debora Carpanese ◽  
Isabella Monia Montagner ◽  
...  

Prostate cancer (PCa) is the second leading cause of malignancy-related mortality in males in the Western world. Although treatment like prostatectomy and radiotherapy for localized cancer have good results, similar positive outcomes are not achieved in metastatic PCa. Consequently, these aggressive and metastatic forms of PCa urgently need new methods of treatment. We already described an efficient and specific second-generation (2G) Chimeric Antigen Receptor (CAR) against Prostate Specific Membrane Antigen (PSMA), a glycoprotein overexpressed in prostate cancer and also present on neovasculature of several tumor entities. In an attempt to improve efficacy and in vivo survival of anti-PSMA 2G CAR-T cells, we developed a third generation (3G) CAR containing two costimulatory elements, namely CD28 and 4-1BB co-signaling domains, in addition to CD3ζ. Differently from what described for other 3G receptors, our third generation CAR disclosed an antitumor activity in vitro similar to the related 2G CAR that comprises the CD28 co-signaling domain only. Moreover, the additional costimulatory domain produced detrimental effects, which could be attributed to an increased activation-induced cell death (AICD). Indeed, such “superstimulation” resulted in an exhausted phenotype of CAR-T cells, after prolonged in vitro restimulation, a higher frequency of cell death, and an impairment in yielding sufficient numbers of transgenic T lymphocytes. Thus, the optimal combination of costimulatory domains for CAR development should be assessed cautiously and evaluated case-by-case.


Cancers ◽  
2021 ◽  
Vol 13 (6) ◽  
pp. 1401
Author(s):  
Shabirul Haque ◽  
Sarah R. Vaiselbuh

CAR-T cell therapy is not without some clinical adverse effects, namely cytokine storms, due to a massive release of cytokines when CAR-T cells multiply in the body. Our goal was to develop exosomes expressing CD19 CAR to treat CD19-positive B-cell malignancies, instead of using whole CD19 CAR-T cells, thereby reducing the clinical risk of uncontrolled cytokine storms. Exosomes are extracellular nanovesicles (30–150 nm), composed of lipids, proteins, and nucleic acids, that carry the fingerprint of their parent cells. Exosomes are a preferred delivery system in nano-immunotherapy. Here, HEK293T parent cells were transduced with CD19 CAR plasmids and cellular CD19 CAR expression was confirmed. Exosomes (Exo-CD19 CAR) were isolated from the conditioned medium of non-transduced (WT) and CD19 CAR plasmid transduced HEK293T cells. Consequently, CD19 B-lineage leukemia cell lines were co-cultured with Exo-CD19 CAR and cell death was measured. Our data show that Exo-CD19 CAR treatment induced cytotoxicity and elevated pro-apoptotic genes in CD19-positive leukemia B-cells without inducing cell death in CD19-negative cells. Overall, the novel CD19 CAR exosomes target the CD19 surface antigens of leukemic B-cells and can induce contact-dependent cytotoxicity.


BMC Medicine ◽  
2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Chunyi Shen ◽  
Zhen Zhang ◽  
Yonggui Tian ◽  
Feng Li ◽  
Lingxiao Zhou ◽  
...  

Abstract Background Chimeric antigen receptor T (CAR-T) cell therapy has limited effects in the treatment of solid tumors. Sulforaphane (SFN) is known to play an important role in inhibiting tumor growth, but its effect on CAR-T cells remains unclear. The goal of the current study was to determine whether combined CAR-T cells and SFN could provide antitumor efficacy against solid tumors. Methods The effect of combined SFN and CAR-T cells was determined in vitro using a co-culture system and in vivo using a xenograft mouse model. We further validated the effects of combination therapy in patients with cancer. Results In vitro, the combination of SFN and CAR-T cells resulted in enhanced cytotoxicity and increased lysis of tumor cells. We found that SFN suppressed programmed cell death 1 (PD-1) expression in CAR-T cells and potentiated antitumor functions in vitro and in vivo. As a ligand of PD-1, programmed cell death ligand 1 (PD-L1) expression was also decreased in tumor cells after SFN treatment. In addition, β-TrCP was increased by SFN, resulting in higher activation of ubiquitination-mediated proteolysis of PD-L1, which induced PD-L1 degradation. The combination of SFN and CAR-T cell therapy acted synergistically to promote better immune responses in vivo compared with monotherapy. In clinical treatments, PD-1 expression was lower, and proinflammatory cytokine levels were higher in patients with various cancers who received CAR-T cells and took SFN orally than that in the control group. Conclusion SFN improves the cytotoxicity of CAR-T cells by modulating the PD-1/PD-L1 pathway, which may provide a promising strategy for the combination of SFN with CAR-T cells for cancer immunotherapy.


Cells ◽  
2020 ◽  
Vol 9 (4) ◽  
pp. 873 ◽  
Author(s):  
Andreas A. Hombach ◽  
Ulf Geumann ◽  
Christine Günther ◽  
Felix G. Hermann ◽  
Hinrich Abken

Chimeric antigen receptor (CAR) redirected T cells are efficacious in the treatment of leukemia/lymphoma, however, showed less capacities in eliminating solid tumors which is thought to be partly due to the lack of cytokine support in the tumor lesion. In order to deliver supportive cytokines, we took advantage of the inherent ability of mesenchymal stem cells (MSCs) to actively migrate to tumor sites and engineered MSCs to release both IL7 and IL12 to promote homeostatic expansion and Th1 polarization. There is a mutual interaction between engineered MSCs and CAR T cells; in presence of CAR T cell released IFN-γ and TNF-α, chronic inflammatory Th2 MSCs shifted towards a Th17/Th1 pattern with IL2 and IL15 release that mutually activated CAR T cells with extended persistence, amplification, killing and protection from activation induced cell death. MSCs releasing IL7 and IL12 were superior over non-modified MSCs in supporting the CAR T cell response and improved the anti-tumor attack in a transplant tumor model. Data demonstrate the first use of genetically modified MSCs as vehicles to deliver immuno-modulatory proteins to the tumor tissue in order to improve the efficacy of CAR T cells in the treatment of solid malignancies.


Cytotherapy ◽  
2018 ◽  
Vol 20 (10) ◽  
pp. 1259-1266 ◽  
Author(s):  
Sergey N. Zolov ◽  
Skyler P. Rietberg ◽  
Challice L. Bonifant

2019 ◽  
Vol 30 (4) ◽  
pp. 446-458 ◽  
Author(s):  
Bian Hu ◽  
Yan Zou ◽  
Linlin Zhang ◽  
Jiaxing Tang ◽  
Gabriele Niedermann ◽  
...  

2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. A1029-A1030
Author(s):  
Gail Petuna Risbridger ◽  
Laura Helen Porter ◽  
Joe Zhu ◽  
David Byrne ◽  
Natalie Lister ◽  
...  

Abstract Chimeric antigen receptor T (CAR T) cell therapy is an adoptive immunotherapy that has led to new treatments for lymphoma, leukemia, and other blood cancers; however, its efficacy for prostate cancer remains unproven. Here we report pre-clinical evidence of the efficacy of CAR T cell therapy against the Lewis Y antigen (LeY) using patient-derived models of prostate cancer. To assess the expression of LeY on prostate tumours, we performed immunohistochemistry on a cohort of 41 patient-derived xenografts (PDXs). Cytoplasmic and membrane expression were separately assessed and quantified, for each patient. Overall, 61% (25/41) of PDXs were positive for membrane LeY expression, of which 18 PDXs had greater than 50% membrane-positive cells, and considered most suitable to detection and stable binding by anti-LeY CAR T’s. To determine the in vitro sensitivity to CAR T cytotoxicity, we selected 4 PDXs with high and 2 PDXs with low LeY expression using 3 androgen receptor (AR)-positive adenocarcinomas and 3 AR-negative tumors expressing neuroendocrine markers. Next we established organoids for in vitro co-culture assays where organoids were co-incubated with an equal number of anti-LeY+ CAR T cells or Empty vector control CAR T cells (Ev CAR T). Using time-lapse microscopy we reported destruction of organoids by LeY+ CAR T cells as indicated by their morphological collapse and uptake of propidium iodide from the culture medium; control Ev CAR T cells produced no cytotoxicity. Over the 48h assay, the level of target cell death of the LeY+ organoids was correlated to the intensity LeY surface expression. Target cell death mediated by the CAR T cells required perforin and granzyme B, as potent and highly specific small molecule inhibitors of perforin (SN34960) and granzyme B (C20) applied alone or in combination greatly decreased PI uptake, indicating organoid survival. Neither inhibitor adversely affected CAR T cell viability as measured by PI and Annexin V staining. This demonstrated canonical activation of granule exocytosis pathway by the CAR T cells, leading to organoid cell death. To assess CAR T cell efficacy in vivo, we selected one PDX with high LeY expression. Monotherapy with CAR T cells failed to decrease tumour volume compared to vehicle control. However, CAR T cells given after a single dose of the chemotherapeutic agent carboplatin greatly and durably reduced tumour burden, with residual tumour mass being less than 1% of their original size (0.56 ± 0.23% of tumour volume at the start of treatment). Overall, these data provide preclinical evidence that: i) high membrane expression of LeY correlates with in vitro and in vivo CAR T cell-induced tumour cell death via the canonical perforin/granzyme B mechanism; and, ii) membrane LeY can be used as a biomarker for patient selection.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3226-3226 ◽  
Author(s):  
Bailin He ◽  
Lei Wang ◽  
Brigitte Neuber ◽  
Anita Schmitt ◽  
Niclas Kneisel ◽  
...  

Introduction Despite the encouraging outcome of anti-CD19 chimeric antigen receptor T (CAR T) cell therapy in patients with B cell malignancies, CAR T cell persistence remains a major clinical challenge. Activation-induced cell death (AICD) is a programmed cell death caused by the interaction of CD95 and CD95L. Through specific blocking of the CD95-CD95L pathway, the CD95L inhibitor APG101 (Asunercept, Apogenix AG, Heidelberg) could prevent activated T cells from AICD. APG101 is a fully human fusion protein consisting of the extracellular domain of CD95 receptor and the Fc domain of an IgG antibody. Thus, we evaluated whether a blockade of the CD95L pathway through APG101 might improve CAR T cell persistence and enhance antitumor efficacy. Methods Human peripheral blood mononuclear cells (PBMCs) from healthy donors were stimulated by plate-bound CD3 and CD28 antibodies, and thereafter transduced with a 3rd generation CD19.CAR.CD28.CD137zeta retroviral vector. An in vitro co-culture stress test assay was employed to assess the functional status and viability of CD19.CAR T cells upon repetitive stimulation with CD19+ tumor cells, i.e. Daudi cells. CAR T cells (5.0 x 105 per well) were co-cultured with tumor cells at a 1:1 E:T ratio (round I) in the presence of APG101. Additional tumor cells were supplied to the co-culture every 24 hours. After 3 rounds (72 hr) of stimulation, tumor cells (CD3-CD19+) and CAR T cells (CD3+CD19-) were harvested for FACS analysis. To assess the antigen-induced CAR T cell proliferation, CAR T cells were preloaded with Cell Trace Violet cytosolic dye and cocultured with tumor cells for 72 hours. Results Activation-induced cell death of CAR T cells was observed after repeated antigenic stimulation, accompanied by increased CD95L expression. CD4+ CAR T cells were more susceptible to AICD compared to CD8+ CAR T cells. But, there was no difference in the expression of CD95L between CD4+ and CD8+ CAR T cells. Interestingly, addition of APG101 significantly inhibited CD95L expression and resulted in a lower level of CAR T cell death. Importantly, APG101 did not hamper the activation and proliferation of CAR T cells but was able to restore CAR T cell viability. The expression of PD1, TIM3 and LAG3 were also up-regulated after successive stimulation, however, their expression on CAR T cells were not influenced by APG101. After 3 days of co-culture, the number of CAR T cells increased in the presence of APG101 (7.9 x 105 vs6.0 x 105, P = 0.01) and residual tumor cells were diminished (1.7 x 105 vs2.7 x 105, P = 0.02). Of note, APG101 itself did not affect CAR T cells or tumor cells when cultured separately. Moreover, the central memory CAR T (TCM) cell subset showed higher CD95L expression after coculturing which could be inhibited by APG101. Therefore, the addition of APG101 to the coculture resulted in a significant accumulation of TCM subset after APG101 treatment. Conclusion Upregulation of CD95L after repeated antigen stimulation was reversed by APG101. CD95L blockade enhanced CAR T cell survival and promoted killing of tumor cells in vitro. Combining CAR T cell therapy with CD95L inhibitor might improve CAR T cell persistence in vivo and thus enhance the effect of CAR T cell therapy. Disclosures Schmitt: Therakos Mallinckrodt: Other: Financial Support . Kneisel:Apogenix AG: Employment. Hoeger:Apogenix AG: Employment, Membership on an entity's Board of Directors or advisory committees. Schmitt:MSD: Membership on an entity's Board of Directors or advisory committees, Other: Sponsoring of Symposia; Therakos Mallinckrodt: Other: Financial Support.


Sign in / Sign up

Export Citation Format

Share Document