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
. 2024 Jan 4;143(1):57-63.
doi: 10.1182/blood.2023020554.

Ibrutinib-based therapy reinvigorates CD8+ T cells compared to chemoimmunotherapy: immune monitoring from the E1912 trial

Affiliations

Ibrutinib-based therapy reinvigorates CD8+ T cells compared to chemoimmunotherapy: immune monitoring from the E1912 trial

Despoina Papazoglou et al. Blood. .

Abstract

Bruton tyrosine kinase inhibitors (BTKis) that target B-cell receptor signaling have led to a paradigm shift in chronic lymphocytic leukemia (CLL) treatment. BTKis have been shown to reduce abnormally high CLL-associated T-cell counts and the expression of immune checkpoint receptors concomitantly with tumor reduction. However, the impact of BTKi therapy on T-cell function has not been fully characterized. Here, we performed longitudinal immunophenotypic and functional analysis of pretreatment and on-treatment (6 and 12 months) peripheral blood samples from patients in the phase 3 E1912 trial comparing ibrutinib-rituximab with fludarabine, cyclophosphamide, and rituximab (FCR). Intriguingly, we report that despite reduced overall T-cell counts; higher numbers of T cells, including effector CD8+ subsets at baseline and at the 6-month time point, associated with no infections; and favorable progression-free survival in the ibrutinib-rituximab arm. Assays demonstrated enhanced anti-CLL T-cell killing function during ibrutinib-rituximab treatment, including a switch from predominantly CD4+ T-cell:CLL immune synapses at baseline to increased CD8+ lytic synapses on-therapy. Conversely, in the FCR arm, higher T-cell numbers correlated with adverse clinical responses and showed no functional improvement. We further demonstrate the potential of exploiting rejuvenated T-cell cytotoxicity during ibrutinib-rituximab treatment, using the bispecific antibody glofitamab, supporting combination immunotherapy approaches.

PubMed Disclaimer

Conflict of interest statement

Conflict-of-interest disclosure: S.H., M.B., and C.K. are employees of and declare patents and stock ownership with Roche Glycart AG. A.G.R. has received research support from Roche Glycart AG to Institution as part of a research collaboration. The remaining authors declare no competing financial interests.

Figures

None
Graphical abstract
Figure 1.
Figure 1.
Higher CD8+T-cell numbers at baseline and early on-therapy associate with favorable PFS and no infections with ibrutinib-rituxumab. (A) Schematic representation of the E1912 trial and biobanked peripheral blood mononuclear cell samples collected at baseline (B/L) and 6- (6M) and 12- (12M) month time points for correlative T-cell analysis. PFS, infection, and MRD clinical outcome data were collected. (B) Absolute numbers of CD8+ TEM cell subsets (CD45RACCR7) for ibrutinib-rituximab (n = 86 patients) and FCR (n = 50) at the time points indicated. Patient data are presented as Box and whiskers (10-90 percentile; log scale) plots. (C) Percentage of PD-1+ CD8+ TEM subsets during ibrutinib-rituximab (n = 86) or FCR (n = 50) treatments. (B-C) Data are given as the mean ± standard error of the mean; statistical analysis between time points were assessed using the Wilcoxon signed-rank test. (D) Tabular schematic summary of the significant correlations (Cox model) between higher immune subset levels (flow cytometry, median values used as cut-off point) and PFS for patients on ibrutinib-rituximab (n = 88 patients with 13 experiencing disease progression). Green rows (correlations with hazard ratio [HR] values < 1) indicate higher immune subsets associated with longer PFS, whereas higher immune subsets associating with shorter PFS (HR > 1) are highlighted in blue rows. Confidence intervals (95%) and P values are shown. (E) Schematic summary of the significant correlations (Wilcoxon test) between immune subsets and infection (any infection) during ibrutinib-rituximab treatment (n = 88 patients). Negative t statistics (t.stat) indicate higher immune subset levels in patients who did not develop infection (green rows). In contrast, correlations with a positive t.stat indicate higher immune subset levels in patients who developed infection (blue rows). (F) Kaplan-Meier curves of immune subsets associated to good prognosis for the ibrutinib-rituximab arm. Higher levels of percentage PD-L1+ CD19+ cells (high: 12 progression events per 43 patients and low: 1 progression event per 43 patients), absolute number of PD-1+CD8+ TEM (high: 3 progression events per 43 patients and low: 10 progression events per 43 patients), and PD-1+CD4+ T cells (high: 3 progression events per 43 patients and low: 10 progression events per 43 patients) at baseline associate with longer PFS. Higher percentage of CD8+ TEM (high: 3 progression events per 42 patients and low: 10 progression events per 43 patients) at the 6-month time point associate with longer PFS. Absolute number data are referred to as “ab.” P values indicated. ∗P < .05; ∗∗P < .01; ∗∗∗∗P < .0001. n/s, not significant.
Figure 2.
Figure 2.
Ibrutinib-rituximab promotes CD8+ T-cell lytic synapse activity and supports immunotherapy-triggered anti-CLL killing function. (A) Illustration of the autologous cytotoxicity assay using anti-CD3/-CD28–activated T cells (cytolytic T lymphocytes [CTLs]) from B/L, 6M, and 12M time points mixed with target B/L CLL B cells (pulsed with superantigen as a model antigen) with flow-based quantification of T-cell killing function. (B) T-cell–mediated CLL cell death comparing T cells purified from B/L, 6M, and 12M time point samples (n = 30 patients per treatment arm). Data at 6M and 12M were normalized to B/L levels to generate fold change values for each patient. (C) The association between patient’s T-cell killing function (12M ibrutinib-rituximab time point, n = 30) and infection status during ibrutinib-rituximab therapy (no infections vs grade 2 or 3 infections) (Wilcoxon test, P = .01). (D-E) Representative confocal medial optical section and 3-dimensional (3D) volume–rendered images of T-cell:CLL conjugates formed between patient T cells (B/L, 6M, and 12M on-ibrutinib-rituximab [D] or FCR [E]) interacting with autologous B/L CLL B cells (blue, CMAC dyed). Bar charts: quantitative relative recruitment index (RRI) analysis of F-actin polarization (red, rhodamine phalloidin) in T-cell:CLL conjugates (n = 50 patients per treatment arm). (F) Box and violin plots (minimum-maximum) showing the percentage of CD4+ or CD8+ T-cell:CLL conjugates formed from the total T-cell:CLL conjugates in B/L, 6M, and 12M ibrutinib-rituximab time point samples (n = 15 patients). Representative confocal images of CD8+ (white) and CD4+ (green) T-cell conjugates with CLL B cells (blue) at B/L vs on ibrutinib-rituximab therapy. (G) Representative confocal 3D volume–rendered images of granzyme B (GrB; white) expression at CD8+ T-cell synapses, comparing ibrutinib-rituximab and FCR 12M time point samples. (H) Kaplan-Meier curve showing the association between the strength of polarized F-actin CD4+ T-cell:CLL immune synapse interactions in patient B/L samples and their PFS outcomes during ibrutinib-rituximab administration. Median F-actin RRI values were used as a cut-off point to determine weak (<median RRI) vs strong (>median RRI) CD4+ T-cell synapses (n = 52 patients). Patients’ showing strong CD4+ T-cell:CLL immune synapses at B/L showed significantly adverse PFS (9 progression events per 29 patients) compared with patients showing weak CD4+ T-cell:CLL interactions (1 progression event per 23 patients). (Cox model, P = .01; HR, 9.14; 95% confidence interval, 1.15-72.47). Bar chart: F-actin RRI analysis of CD4+ T-cell:CLL conjugates at B/L and representative 3D volume–rendered confocal images comparing patients who progressed (n = 7) with those who did not (progression free, n = 7) during ibrutinib-rituximab therapy. (I) Illustration of the cytotoxicity assay after ex vivo treatment of purified T cells (B/L, 6M, and 12M time points) and B/L CLL cells with anti–PD-1 (αPD-1) or anti–PD-L1 (αPD-L1) blocking antibodies (10 μg/mL) or isotype controls. (J-K) T-cell killing function against autologous B/L CLL cells examining T cells at B/L or at the 6-month ibrutinib-rituximab (orange) or FCR (blue) time points after ex vivo treatment with (J) αPD-1 or isotype control (indicated using “−”) (B/L: n = 6, ibrutinib-rituximab: n = 13, and FCR: n = 15) or (K) αPD-L1 or isotype control (−) (B/L: n = 6, ibrutinib-rituximab: n = 23, and FCR: n = 13). (L) Illustration of the autologous cytotoxicity assay incorporating the CD20 × CD3 glofitamab or a nonbinding antibody control. (M-N) T-cell–mediated CLL cell death using purified T cells from B/L, 6M, 12M, or 18M ibrutinib-rituximab (M) or FCR (N) time points against target B/L CLL B cells after ex vivo treatment with glofitamab (0.01 μg/mL) or nonbinding antibody control (indicated as “−”) (B/L, n = 13; ibrutinib-rituximab 6M and 12M,n = 6; ibrutinib-rituximab 18M, n = 5; FCR 6M and 12M, n = 7; and FCR 18M, n = 5 patient samples). Data for all cytotoxicity assay timepoints were normalized to isotype antibody control for panels J-K or nonbinding antibody control for panels M-N treated sample levels and presented as fold change data for each immunotherapy treated patient sample. Wilcoxon signed-rank test for panels B,D-E,J-K,M-N, multiple comparisons mixed effect analysis of variance for panel F and Mann-Whitney U test for panel H. Mann-Whitney U test was used to compare cell death among CD20-TCB–treated conditions at B/L, 6M, 12M, and 18M. Original magnification ×63; scale bars, 10 μm. Bar chart data are presented as mean ± standard error of the mean. ∗P < .05; ∗∗P < .01; ∗∗∗P < .001; ∗∗∗∗P < .0001. CMAC, CellTracker Blue (7-amino-4-chloromethylcoumarin); TCB, T-cell bispecific antibody.
Figure 2.
Figure 2.
Ibrutinib-rituximab promotes CD8+ T-cell lytic synapse activity and supports immunotherapy-triggered anti-CLL killing function. (A) Illustration of the autologous cytotoxicity assay using anti-CD3/-CD28–activated T cells (cytolytic T lymphocytes [CTLs]) from B/L, 6M, and 12M time points mixed with target B/L CLL B cells (pulsed with superantigen as a model antigen) with flow-based quantification of T-cell killing function. (B) T-cell–mediated CLL cell death comparing T cells purified from B/L, 6M, and 12M time point samples (n = 30 patients per treatment arm). Data at 6M and 12M were normalized to B/L levels to generate fold change values for each patient. (C) The association between patient’s T-cell killing function (12M ibrutinib-rituximab time point, n = 30) and infection status during ibrutinib-rituximab therapy (no infections vs grade 2 or 3 infections) (Wilcoxon test, P = .01). (D-E) Representative confocal medial optical section and 3-dimensional (3D) volume–rendered images of T-cell:CLL conjugates formed between patient T cells (B/L, 6M, and 12M on-ibrutinib-rituximab [D] or FCR [E]) interacting with autologous B/L CLL B cells (blue, CMAC dyed). Bar charts: quantitative relative recruitment index (RRI) analysis of F-actin polarization (red, rhodamine phalloidin) in T-cell:CLL conjugates (n = 50 patients per treatment arm). (F) Box and violin plots (minimum-maximum) showing the percentage of CD4+ or CD8+ T-cell:CLL conjugates formed from the total T-cell:CLL conjugates in B/L, 6M, and 12M ibrutinib-rituximab time point samples (n = 15 patients). Representative confocal images of CD8+ (white) and CD4+ (green) T-cell conjugates with CLL B cells (blue) at B/L vs on ibrutinib-rituximab therapy. (G) Representative confocal 3D volume–rendered images of granzyme B (GrB; white) expression at CD8+ T-cell synapses, comparing ibrutinib-rituximab and FCR 12M time point samples. (H) Kaplan-Meier curve showing the association between the strength of polarized F-actin CD4+ T-cell:CLL immune synapse interactions in patient B/L samples and their PFS outcomes during ibrutinib-rituximab administration. Median F-actin RRI values were used as a cut-off point to determine weak (<median RRI) vs strong (>median RRI) CD4+ T-cell synapses (n = 52 patients). Patients’ showing strong CD4+ T-cell:CLL immune synapses at B/L showed significantly adverse PFS (9 progression events per 29 patients) compared with patients showing weak CD4+ T-cell:CLL interactions (1 progression event per 23 patients). (Cox model, P = .01; HR, 9.14; 95% confidence interval, 1.15-72.47). Bar chart: F-actin RRI analysis of CD4+ T-cell:CLL conjugates at B/L and representative 3D volume–rendered confocal images comparing patients who progressed (n = 7) with those who did not (progression free, n = 7) during ibrutinib-rituximab therapy. (I) Illustration of the cytotoxicity assay after ex vivo treatment of purified T cells (B/L, 6M, and 12M time points) and B/L CLL cells with anti–PD-1 (αPD-1) or anti–PD-L1 (αPD-L1) blocking antibodies (10 μg/mL) or isotype controls. (J-K) T-cell killing function against autologous B/L CLL cells examining T cells at B/L or at the 6-month ibrutinib-rituximab (orange) or FCR (blue) time points after ex vivo treatment with (J) αPD-1 or isotype control (indicated using “−”) (B/L: n = 6, ibrutinib-rituximab: n = 13, and FCR: n = 15) or (K) αPD-L1 or isotype control (−) (B/L: n = 6, ibrutinib-rituximab: n = 23, and FCR: n = 13). (L) Illustration of the autologous cytotoxicity assay incorporating the CD20 × CD3 glofitamab or a nonbinding antibody control. (M-N) T-cell–mediated CLL cell death using purified T cells from B/L, 6M, 12M, or 18M ibrutinib-rituximab (M) or FCR (N) time points against target B/L CLL B cells after ex vivo treatment with glofitamab (0.01 μg/mL) or nonbinding antibody control (indicated as “−”) (B/L, n = 13; ibrutinib-rituximab 6M and 12M,n = 6; ibrutinib-rituximab 18M, n = 5; FCR 6M and 12M, n = 7; and FCR 18M, n = 5 patient samples). Data for all cytotoxicity assay timepoints were normalized to isotype antibody control for panels J-K or nonbinding antibody control for panels M-N treated sample levels and presented as fold change data for each immunotherapy treated patient sample. Wilcoxon signed-rank test for panels B,D-E,J-K,M-N, multiple comparisons mixed effect analysis of variance for panel F and Mann-Whitney U test for panel H. Mann-Whitney U test was used to compare cell death among CD20-TCB–treated conditions at B/L, 6M, 12M, and 18M. Original magnification ×63; scale bars, 10 μm. Bar chart data are presented as mean ± standard error of the mean. ∗P < .05; ∗∗P < .01; ∗∗∗P < .001; ∗∗∗∗P < .0001. CMAC, CellTracker Blue (7-amino-4-chloromethylcoumarin); TCB, T-cell bispecific antibody.

Comment in

References

    1. Shanafelt TD, Wang XV, Kay NE, et al. Ibrutinib-rituximab or chemoimmunotherapy for chronic lymphocytic leukemia. N Engl J Med. 2019;381(5):432–443. - PMC - PubMed
    1. Shanafelt TD, Wang XV, Hanson CA, et al. Long-term outcomes for ibrutinib-rituximab and chemoimmunotherapy in CLL: updated results of the E1912 Trial. Blood. 2022;140(2):112–120. - PMC - PubMed
    1. Skånland SS, Mato AR. Overcoming resistance to targeted therapies in chronic lymphocytic leukemia. Blood Adv. 2021;5(1):334–343. - PMC - PubMed
    1. Mhibik M, Wiestner A, Sun C. Harnessing the effects of BTKi on T cells for effective immunotherapy against CLL. Int J Mol Sci. 2019;21(1):68. - PMC - PubMed
    1. Ding W, LaPlant BR, Call TG, et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood. 2017;129(26):3419–3427. - PMC - PubMed