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
Clinical Trial
. 2024 Jan 23;8(2):453-467.
doi: 10.1182/bloodadvances.2023011287.

Timing of anti-PD-L1 antibody initiation affects efficacy/toxicity of CD19 CAR T-cell therapy for large B-cell lymphoma

Affiliations
Clinical Trial

Timing of anti-PD-L1 antibody initiation affects efficacy/toxicity of CD19 CAR T-cell therapy for large B-cell lymphoma

Alexandre V Hirayama et al. Blood Adv. .

Abstract

More than half of the patients treated with CD19-targeted chimeric antigen receptor (CAR) T-cell immunotherapy for large B-cell lymphoma (LBCL) do not achieve durable remission, which may be partly due to PD-1/PD-L1-associated CAR T-cell dysfunction. We report data from a phase 1 clinical trial (NCT02706405), in which adults with LBCL were treated with autologous CD19 CAR T cells (JCAR014) combined with escalating doses of the anti-PD-L1 monoclonal antibody, durvalumab, starting either before or after CAR T-cell infusion. The addition of durvalumab to JCAR014 was safe and not associated with increased autoimmune or immune effector cell-associated toxicities. Patients who started durvalumab before JCAR014 infusion had later onset and shorter duration of cytokine release syndrome and inferior efficacy, which was associated with slower accumulation of CAR T cells and lower concentrations of inflammatory cytokines in the blood. Initiation of durvalumab before JCAR014 infusion resulted in an early increase in soluble PD-L1 (sPD-L1) levels that coincided with the timing of maximal CAR T-cell accumulation in the blood. In vitro, sPD-L1 induced dose-dependent suppression of CAR T-cell effector function, which could contribute to inferior efficacy observed in patients who received durvalumab before JCAR014. Despite the lack of efficacy improvement and similar CAR T-cell kinetics early after infusion, ongoing durvalumab therapy after JCAR014 was associated with re-expansion of CAR T cells in the blood, late regression of CD19+ and CD19- tumors, and enhanced duration of response. Our results indicate that the timing of initiation of PD-L1 blockade is a key variable that affects outcomes after CD19 CAR T-cell immunotherapy for adults with LBCL.

PubMed Disclaimer

Conflict of interest statement

Conflict-of-interest disclosure: A.V.H. has received research funding from Juno Therapeutics, a Bristol Myers Squibb company, and Nektar Therapeutics, and honoraria from Bristol Myers Squibb and Novartis. E.L.K. has received research funding from Juno Therapeutics, a Bristol Myers Squibb company. S.F. reports grants from Bristol Myers Squibb and other support in the form of pending equity from Link Immunotherapeutics outside of the submitted work; has issued patents for PCT/US2021/025255 and PCT/US2021/025248d; and a patent for PCT/US2021/025260 issued, licensed, and with royalties paid from Bristol Myers Squibb. J.G. has received research funding from Sobi, Juno Therapeutics, a Bristol Myers Squibb company, Celgene, and Angiocrine Bioscience, and has received honoraria/consulting fees from Sobi, Legend Biotech, Janssen, Kite Pharma, a Gilead company, and MorphoSys. C.C.S.Y. has received research funding from OBI Pharma, Lonza, Sensei, Signal One, and Pfizer, and serves on scientific advisory boards for Twinstrand Biosciences, AbbVie, Eli Lilly, and Loxo. R.D.C. has received research funding from Amgen, Incyte, Kite Pharma, a Gilead company, Pfizer, Servier, and Vanda Pharmaceuticals; has received honoraria/consulting fees from Amgen, Jazz, Kite/Gilead, and Pfizer; serves on a data and safety monitoring board for Pepromene Bio and on an independent response review committee for Autolus; and his spouse has been employed by and owned stock in Seagen. A.G.C. has received research funding from Juno Therapeutics, a Bristol Myers Squibb company. D.J.G. has received research funding from, has served as an adviser for, and has received royalties from Juno Therapeutics, a Bristol Myers Squibb company; has served as an adviser and received research funding from Seattle Genetics; has served as an adviser to GlaxoSmithKline, Celgene, Janssen Biotech, and Legend Biotech; and has received research funding from SpringWorks Therapeutics, Sanofi, and Cellectar Biosciences. M.S. has received research funding from Mustang Bio, Bristol Myers Squibb, Pharmacyclics, Genentech, AbbVie, TG Therapeutics, BeiGene, AstraZeneca, Genmab, MorphoSys/Incyte, and Vincerx; has served as a consultant for AbbVie, Genentech, AstraZeneca, Pharmacyclics, Beigene, Bristol Myers Squibb, MorphoSys/Incyte, Kite, Eli Lilly, Genmab, Mustang Bio, Regeneron, ADC Therapeutics, Janssen, Fate Therapeutics, and MEI Pharma; and his spouse is an employee of Bristol Myers Squibb. B.G.T. has received research funding from Mustang Bio and Juno Therapeutics, a Bristol Myers Squibb company; serves on scientific advisory boards for Mustang Bio and Proteios Technology; and has the right to receive royalties from Fred Hutch as an inventor on licensed patents. S.R.R. is a cofounder and adviser to Lyell Immunopharma and has received research funding from and intellectual property licensed to Lyell Immunopharma; was a cofounder of Juno Therapeutics, a Bristol Myers Squibb company; is an inventor of patents licensed to Juno Therapeutics; and served as an adviser to Juno Therapeutics and Adaptive Biotechnologies. D.G.M. has received research funding from Juno Therapeutics, a Bristol Myers Squibb company, Celgene, and Kite Pharma, a Gilead company; has served on ad hoc advisory board meetings for Amgen, Bristol Myers Squibb, Genentech, Gilead, Incyte, Janssen, Legend Biotech, Mustang Bio, MorphoSys, Novartis, Pharmacyclics, and Umoja; has rights to receive royalties from Fred Hutch for patents licensed to Juno Therapeutics; and serves on scientific advisory board with stock options and compensations for A2 Biotherapeutics and Navan Technologies. C.J.T. has received research funding from Juno Therapeutics, a Bristol Myers Squibb company, NanoString Technologies, and Nektar Therapeutics; serves on scientific advisory boards for Caribou Biosciences, T-CURX, Myeloid Therapeutics, ArsenalBio, and Cargo Therapeutics; serves on a data and safety monitoring board for Kyverna; has served on ad hoc advisory board meetings (last 12 months) for Legend Biotech, Nektar Therapeutics, and Syncopation Life Sciences; performs consulting for Century Therapeutics, Orna Therapetuics, and IGM Biosciences; has stock options in Eureka Therapeutics, Caribou Biosciences, Myeloid Therapeutics, Cargo Therapeutics, and ArsenalBio; and has the right to receive payments from Fred Hutch as an inventor on licensed patents. The remaining authors have declared no competing financial interests.

Figures

None
Graphical abstract
Figure 1.
Figure 1.
Best response rate at 3 months. (A) Best response at 3 months on 26 of the 29 patients included in the efficacy-evaluable set according to disease histology (DLBCL NOS; tDLBCL; and HGBL with MYC and BCL2 and/or BCL6 rearrangements) and in the whole cohort. (B) Subgroup analysis of the ORR for key baseline and clinical covariates. ∗Indicates double expressor excluding double- or triple-hit lymphoma (total of 23 patients). † indicates cell of origin by the Hans algorithm. ‡ indicates the sum of the product of the perpendicular diameters of up to 6 target measurable nodes and extranodal sites. § indicates the maximum standardized uptake value in target lesion. The 95% CI was calculated with the use of the Wilson/Brown method. ECOG PS, Eastern Cooperative Oncology Group performance score; HGBL, high-grade B-cell lymphoma; IPI, International Prognostic Index; LDH, lactate dehydrogenase; NOS, not otherwise specified; SPD, sum of the product of the perpendicular diameters of up to 6 target measurable nodes and extranodal sites; tDLBCL, DLBCL transformed from indolent histology.
Figure 2.
Figure 2.
CAR T-cell kinetics in patients treated with JCAR014 with and without durvalumab. (A) CAR T-cell counts in the blood by qPCR in groups 1 and 2 (NCT02706405) and JCAR014 alone (NCT01865617) cohorts. Each thin line represents a single patient; bold lines represent the averaged data using local polynomial regression (LOESS) curve fitting approximation with the standard error in gray. (B) Time to CAR T-cell peak counts in the blood by qPCR in groups 1 and 2 (NCT02706405) and the JCAR014 alone (NCT01865617) cohorts. Mann-Whitney tests were used to compare differences between the groups.
Figure 3.
Figure 3.
Serum biomarkers in patients treated with JCAR014 with and without durvalumab. AUC0-28/D0 after CAR T-cell infusion of serum IFN-γ (A), macrophage inflammatory protein-1β (MIP-1β) (B), and sPD-L1 (C) according to the treatment group. (D) Serum sPD-L1 peak concentration after CAR T-cell infusion according to treatment group. (E) Serum sPD-L1 in the JCAR014 in combination with durvalumab (groups 1 and 2; NCT02706405) and JCAR014 alone cohorts (NCT01865617). (F) Serum sPD-L1 and CAR T-cell counts in the blood by qPCR in the JCAR014 in combination with durvalumab (groups 1 and 2; NCT02706405) and JCAR014 alone cohorts (NCT01865617). (E-F) Each thin line represents a single patient, and each dot represents a sample; bold lines represent the averaged data using LOESS curve fitting approximation. Mann-Whitney tests were used to compare differences between groups.
Figure 4.
Figure 4.
sPD-L1 inhibits CAR T-cell cytokine production in vitro in a dose-dependent manner. Human CD19 CAR T cells were generated from healthy donors (n = 3) and assayed on days 14 to 16 after the start of manufacturing. CD4+ CAR T cells were cocultured with CD19-expressing K562 cells at a 1:1 effector-to-target ratio and the indicated sPD-L1 concentrations or media alone for ∼24 hours. (A) The percentages of CD4+ CAR T cells expressing the indicated numbers of cytokines were measured by intracellular flow cytometry. IFN-γ (B) and IL-2 (C) individual production. Data are representative of at least 2 independent experiments. Figures show mean ± standard error of the mean. One-way analysis of variance tests with Tukey correction for multiple comparisons were used to compare differences between groups. IL-2, interleukin-2; ns, not significant.
Figure 5.
Figure 5.
DOR in patients treated with JCAR014 with and without durvalumab. Kaplan-Meier estimates of the DOR according to treatment cohort. P value per log-rank test. The numbers of patients at risk at 10-month intervals are indicated.
Figure 6.
Figure 6.
CAR T-cell progenitor exhausted phenotype and in vivo re-expansion in the blood with repeated dosing of durvalumab after JCAR014. (A) Aliquots of the infusion products (n = 26) and blood collected after CAR T-cell infusion (expansion, n = 26; contraction, n = 21; day 28, n = 12) were analyzed by flow cytometry. Percentage of PD-1+, TCF-1+, and PD-1+TCF-1+ CD3+ CAR T cells at each timepoint are shown. Figures show mean ± standard error of the mean. (B) CAR T-cell counts in the blood by qPCR in patients treated with JCAR014 in combination with durvalumab who experienced in vivo re-expansion of CAR T cells. Arrowheads on the x-axis indicate the time of durvalumab doses. Arrows show re-expansion events. Horizontal dashed lines show the qPCR assay limit of detection.
Figure 7.
Figure 7.
Tumor burden stabilization/regression and late conversion to CR with repeated dosing of durvalumab after JCAR014. (A) Spider plot of the change in the sum of the product of the perpendicular diameters of ≤6 target measurable nodes and extranodal sites (SPD) over time compared with the baseline prelymphodepletion according to best response at 3 months. ∗ represents patients with extranodal disease and no measurable target lesion. ‡ represents patients who converted to CR at 1 year. PET/CT (B) and immunohistochemistry (C) images at different timepoints in a patient with late conversion to CR with continued durvalumab despite evidence of CD19 escape. Images were taken at original magnification ×40.

Comment in

References

    1. Hirayama AV, Gauthier J, Hay KA, et al. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood. 2019;133(17):1876–1887. - PMC - PubMed
    1. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531–2544. - PMC - PubMed
    1. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377(26):2545–2554. - PMC - PubMed
    1. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839–852. - PubMed
    1. Deng Q, Han G, Puebla-Osorio N, et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat Med. 2020;26(12):1878–1887. - PMC - PubMed

Publication types

MeSH terms

Associated data