Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Apr 7;29(4):1529-1540.
doi: 10.1016/j.ymthe.2020.12.033. Epub 2021 Jan 1.

Leukemic extracellular vesicles induce chimeric antigen receptor T cell dysfunction in chronic lymphocytic leukemia

Affiliations

Leukemic extracellular vesicles induce chimeric antigen receptor T cell dysfunction in chronic lymphocytic leukemia

Michelle J Cox et al. Mol Ther. .

Abstract

Chimeric antigen receptor (CAR) T cell therapy has yielded unprecedented outcomes in some patients with hematological malignancies; however, inhibition by the tumor microenvironment has prevented the broader success of CART cell therapy. We used chronic lymphocytic leukemia (CLL) as a model to investigate the interactions between the tumor microenvironment and CART cells. CLL is characterized by an immunosuppressive microenvironment, an abundance of systemic extracellular vesicles (EVs), and a relatively lower durable response rate to CART cell therapy. In this study, we characterized plasma EVs from untreated CLL patients and identified their leukemic cell origin. CLL-derived EVs were able to induce a state of CART cell dysfunction characterized by phenotypical, functional, and transcriptional changes of exhaustion. We demonstrate that, specifically, PD-L1+ CLL-derived EVs induce CART cell exhaustion. In conclusion, we identify an important mechanism of CART cell exhaustion induced by EVs from CLL patients.

Keywords: CART cell exhaustion; chimeric antigen receptor T cells; chronic lymphocytic leukemia; extracellular vesicles; microenvironment.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests S.S.K. is an inventor on patents in the field of CAR immunotherapy that are licensed to Novartis (through an agreement between the Mayo Clinic, the University of Pennsylvania, and Novartis). M.J.C., R.S., and S.S.K. are inventors on patents in the field of CAR immunotherapy that are licensed to Humanigen (through the Mayo Clinic). S.S.K. is an inventor on patents in the field of CAR immunotherapy that are licensed to Mettaforge (through the Mayo Clinic). S.S.K. receives research funding from Kite, Gilead, Juno, Celgene, Novartis, Humanigen, MorphoSys, Tolero, Sunesis, Leahlabs, and Lentigen. M.J.C., F.L., N.E.K., and S.S.K. are inventors on patents related to this work. N.E.K. receives research funding from Acerta Pharma, BMS, Pharmacyclics, MEI Pharma, and Sunesis. N.E.K. has participated in Advisory Board meetings of Cytomx Therapy, Janssen, Juno Therapeutics, AstraZeneca, and Oncotracker, and on the DSMC for Agios and Cytomx Therapeutics. S.A.P. receives research funding from Pharmacyclics, MorphoSys, Janssen, AstraZeneca, TG Therapeutics, Bristol Myers Squibb, AbbVie, and Ascentage Pharma. S.A.P. has participated in Advisory Board meetings of Pharmacyclics, AstraZeneca, Genentech, Gilead, GlaxoSmithKline, Verastem Oncology, and AbbVie (he was not personally compensated for his participation).

Figures

None
Graphical abstract
Figure 1
Figure 1
Identification of CLL-derived extracellular vesicles (EVs) in patients with CLL (A–E) Dot plots showing total particle number (A) and EV levels (B–E) measured by nanoscale flow cytometry in platelet-poor plasma isolated from normal individuals (n = 10) and CLL patients (n = 50). (B–E) A panel of fluorescent antibodies was used to enumerate levels of EVs for (B) CD45+, (C) CD19+, (D) CD5+CD19+, and (E) PD-L1+. Values represent number of EVs per microliter transformed in a logarithmic scale (Mann-Whitney test; error bars, SD). (F) Correlation analysis of levels of CLL-derived CD5+CD19+ EVs and PD-L1+ EVs in CLL patients. Pearson correlation coefficient was calculated with a two-tailed p value. (G) Western blot showing expression of three EV-enriched markers (TSG101, CD9, CD81) and PD-L1 in a panel of six EV lysates obtained from platelet-poor plasma of CLL patients. A second band at higher molecular weight was detected for PD-L1 that corresponds to a glycosylated form of the protein. (H) Relative intensity of gel bands for total PD-L1 (left panel) and glycosylated PD-L1 (right panel). Levels of PD-L1 were increased by 1.4-fold (minimum [min]-maximum [max], 1.17–1.77). The glycosylated form of PD-L1 was markedly increased in PD-L1high patients with a 3.0-fold increase (min-max, 1.78–4.37) compared to PD-L1low patients. (I) Correlation analysis of levels of total PD-L1 (left panel) and glycosylated PD-L1 (right panel) between western blot and nanoscale flow cytometric quantification methods. Pearson correlation coefficient was calculated with a two-tailed p value.
Figure 2
Figure 2
CLL-derived EVs induce a state of CART cell dysfunction (A and B) Inhibitory receptor expression on activated CART cells is upregulated by CLL-derived EVs. CART19 cells were co-cultured for 24 h with JeKo-1 cells with different concentrations of EVs (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001, one-way ANOVA; error bars, SEM; three biological and two technical replicates, two experiments). (C and D) CART19 cell antigen-specific proliferation and killing of CD19+ JeKo-1 cells were decreased in the presence of CLL-derived EVs (blue triangles) compared to controls (orange squares). EVs/CART19 cells at a 100:1 ratio were co-cultured for 6 h and plated at a 5:1 effector-to-target ratio (E:T ratio) with JeKo-1 cells (∗∗∗∗p < 0.0001, one-way ANOVA; error bars, SEM; three biological and two technical replicates, three experiments). (E) CART19 cell antigen-specific proliferation was further decreased in the presence of CLL-derived EVs at 1,000:1 and 10,000:1 compared to 100:1. EVs/CART cells were co-cultured for 6 h and plated at a 5:1 E:T ratio with JeKo-1 cells (∗p < 0.05, ∗∗∗∗p < 0.0001, one-way ANOVA; error bars, SEM; three biological and two technical replicates, one experiment). (F) Treatment of JeKo-1 cell xenografts with CART19 cells alone (red squares) improved survival compared to CART19 cells co-cultured with CLL-derived EVs (purple triangles) or untransduced (UTD) T cells (blue circles). NOD-SCID-γ−/− mice were engrafted with the CD19+ luciferase+ cell line JeKo-1 (1 × 106 cells intravenous [i.v.] via tail vein injection), and engraftment was confirmed through bioluminescence imaging (total flux, photons [p]/s). Mice were randomized to treatment with (1) UTD T cells, (2) CART19 cells, and (3) CART19 cells co-cultured ex vivo with CLL-derived EVs for 6 h prior to injection. All T cells were washed prior to injection. A single low dose of CART19 cells (2.5 × 105) was injected to induce relapse (∗p = 0.0198, log-rank test; five mice per group).
Figure 3
Figure 3
EVs from CLL patients induce phenotypical, functional, and transcriptomic changes of exhaustion in T cells (A) CLL-derived EVs do not express E-cadherin. E-cadherin was measured on EVs derived from normal donor (ND) and CLL patients by nanoscale flow cytometry compared to measurements of CD19 on CLL-derived EVs (∗p < 0.05, one-way ANOVA; error bars, SEM; three to five biological replicates, two technical replicates, one experiment). (B) CLL-derived EVs decrease E-cadherin CART cell antigen-specific proliferation. EVs/CART cells at a ratio of 100:1 were co-cultured for 6 h and plated at an E:T ratio of 5:1 with the E-cadherin+ breast cancer cell line MCF-7 (∗∗∗∗p < 0.0001, one-way ANOVA; error bars, SEM; three biological replicates, two technical replicates). The absolute number of live T cells significantly decreased when E-cadherin CART cells were co-cultured with MCF-7 cells in the presence of CLL-derived EVs (blue triangles) compared to E-cadherin CART cells co-cultured with MCF-7 alone (orange squares). The UTD negative control (pink circles) shows background proliferation. (C and D) CART19 cell transcriptome is modulated by CLL-derived EVs. CART19 cells were co-cultured with irradiated JeKo-1 cells for 24 h at a ratio of 10:1, 1:1, or 0:1 EVs/CART19 cells and then isolated by magnetic sorting (three biological replicates, adjusted p value < 0.05). Gene expression with 10:1 EVs/CART19 cells (green columns) and 1:1 EVs/CART19 cells (blue columns) compared to CART19 cells alone (salmon column). EVs increase the expression of AP-1 (FOS-JUN) and YY1. (E) Principal component analysis of CART19 cell RNA-sequencing samples. Similar gene expression patterns were noted between both 1:1 EV/CART19 cell (blue circles) and 10:1 EVs/CART19 cells (red circles). (F) Ingenuity Pathway Analysis predicts increased activation of the AP-1 pathway (FOS-JUN, orange) in CART19 cells co-cultured with CLL-derived EVs. (G) Gene set enrichment analysis for significantly upregulated genes shows enrichment for pathways associated with CD4 (p = 0.037) and CD8 (p = 0.0033) T cell signaling as well as AP-1 transcription factors (p = 0.0445) (red bars, p < 0.05).
Figure 4
Figure 4
CART cell dysfunction is facilitated by PD-L1+ CLL-derived EVs (A and B) CART19 cells alone (red squares) control tumor burden better compared to CART19 cells co-cultured ex vivo with PD-L1high CLL-derived EVs (purple triangles) (∗∗p = 0.0088, two-way ANOVA; error bars, SEM; five mice per group). NOD-SCID-γ−/− mice engrafted with the CD19+luciferase+ cell line JeKo-1 Luc-ZsGreen (1 × 106 cells i.v. via tail vein injection) and engraftment confirmed through bioluminescent imaging (total flux, p/s). Mice were then randomized for treatment with (1) UTD T cells, (2) CART19 cells, (3) CART19 cells co-cultured ex vivo with PD-L1high CLL-derived EVs for 6 h prior to injection, or (4) CART19 cells co-cultured ex vivo with PD-L1low CLL-derived EVs for 6 h prior to injection. A single low dose of CART19 cells (2.5 × 105) was injected to induce relapse. Mice treated with UTD T cells (blue squares) had continued progression of disease. Mice treated with CART19 cells that were pre-cultured with PD-L1low CLL-EVs had a non-statistically significant impairment of anti-tumor activity (green triangles). Mice treated with CART19 cells that were pre-cultured with PD-L1high CLL-EVs had significant impairment of anti-tumor activity. (C) CLL-derived PD-L1high EV impairment of CART19 cells is not significantly reversed by PD-L1 blockade. CART19 cells were co-cultured for 6 h with and without PD-L1high CLL-derived EVs (100:1 EV/CART cell ratio) and with and without anti-PD-L1 antibody. CD19+ JeKo-1 cells were added at an E:T ratio of 5:1. CART19 cell antigen-specific proliferation was significantly impaired in the presence of PD-L1high CLL-derived EVs (p < 0.01, two-way ANOVA). This inhibited CART19 cell antigen-specific proliferation did not improve following a co-culture with anti-PD-L1 antibody (n = 11 biological replicates, two technical replicates, four experiments).

References

    1. Neelapu S.S., Locke F.L., Bartlett N.L., Lekakis L.J., Miklos D.B., Jacobson C.A., Braunschweig I., Oluwole O.O., Siddiqi T., Lin Y. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 2017;377:2531–2544. - PMC - PubMed
    1. Porter D.L., Levine B.L., Kalos M., Bagg A., June C.H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 2011;365:725–733. - PMC - PubMed
    1. Maude S.L., Laetsch T.W., Buechner J., Rives S., Boyer M., Bittencourt H., Bader P., Verneris M.R., Stefanski H.E., Myers G.D. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 2018;378:439–448. - PMC - PubMed
    1. Locke F.L., Ghobadi A., Jacobson C.A., Miklos D.B., Lekakis L.J., Oluwole O.O., Lin Y., Braunschweig I., Hill B.T., Timmerman J.M. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019;20:31–42. - PMC - PubMed
    1. Siddiqi T., Soumerai J.D., Dorritie K.A., Stephens D.M., Riedell P.A., Arnason J.E., Kipps J.K., Gillenwater H.H., Gong L., Dubovsky J.A. Rapid undetectable MRD (uMRD) responses in patients with relapsed/refractory (R/R) chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) treated with lisocabtagene maraleucel (liso-cel), a CD19-directed CAR T cell product: updated results from Transcend CLL 004, a phase 1/2 study including patients with high-risk disease previously treated with ibrutinib. Blood. 2019;134(Suppl 1):503.

Publication types

MeSH terms