Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia

Hanren Dai, Zhiqiang Wu, Hejin Jia, Chuan Tong, Yelei Guo, Dongdong Ti, Xiao Han, Yang Liu, Wenying Zhang, Chunmeng Wang, Yajing Zhang, Meixia Chen, Qingming Yang, Yao Wang, Weidong Han, Hanren Dai, Zhiqiang Wu, Hejin Jia, Chuan Tong, Yelei Guo, Dongdong Ti, Xiao Han, Yang Liu, Wenying Zhang, Chunmeng Wang, Yajing Zhang, Meixia Chen, Qingming Yang, Yao Wang, Weidong Han

Abstract

Background: Despite the impressive complete remission (CR) induced by CD19 CAR-T cell therapy in B-ALL, the high rate of complete responses is sometimes limited by the emergence of CD19-negative leukemia. Bispecific CAR-modified T cells targeting both CD19 and CD22 may overcome the limitation of CD19-negative relapse.

Methods: We here report the design of a bispecific CAR simultaneous targeting of CD19 and CD22. We performed a phase 1 trial of bispecific CAR T cell therapy in patients with relapsed/refractory precursor B-ALL at a dose that ranged from 1.7 × 106 to 3 × 106 CAR T cells per kilogram of body weight.

Results: We demonstrate bispecific CD19/CD22 CAR T cells could trigger robust cytolytic activity against target cells. MRD-negative CR was achieved in 6 out of 6 enrolled patients. Autologous CD19/CD22 CAR T cells proliferated in vivo and were detected in the blood, bone marrow, and cerebrospinal fluid. No neurotoxicity occurred in any of the 6 patients treated. Of note, one patient had a relapse with blast cells that no longer expressed CD19 and exhibited diminished CD22 site density approximately 5 months after treatment.

Conclusion: In brief, autologous CD19/CD22 CAR T cell therapy is feasible and safe and mediates potent anti-leukemic activity in patients with relapsed/refractory B-ALL. Furthermore, the emergence of target antigen loss and expression downregulation highlights the critical need to anticipate antigen escape. Our study demonstrates the reliability of bispecific CD19/CD22 CAR T cell therapy in inducing remission in adult patients with relapsed/refractory B-ALL.

Trial registration: ClinicalTrials.gov identifier: NCT03185494.

Keywords: B-ALL; Bispecific; CAR; CD19; CD22; Immunotherapy; Tumor antigen escape.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Preclinical evaluation of bispecific CD19/CD22 CAR T cells. a Schematic of the bispecific CD19/CD22 CAR structure. b Cytotoxicity of CAR T cells against target cells carrying luciferase reporter gene evaluated by luminescent assay, after co-culturing with tumor cells for 4 h at the indicated E:T ratios, with CAR T cells. c Cytokine production by anti-CD19 CAR-, anti-CD22 CAR-, and bispecific CAR-expressing T cells co-incubated with K562, K562-CD19, K562-CD22, and K562-CD19CD22 cell lines. Bars represent mean + SD of replicate samples. Data are representative of three independent experiments performed with CAR T cells from three separate donors. A two-tailed, unpaired two-sample t test was used for statistical analysis
Fig. 2
Fig. 2
Expansion and persistence of bispecific CAR T cells and tumor response in patients treated with bispecific CAR T cells. a Treatment response of each patient after bispecific CAR T cell treatment and the duration of response. Ongoing remission is marked by a black arrow. Patient number is shown to the left. b The presence of CD19/CD22 CAR T cells in the peripheral blood as assessed by quantitative real-time polymerase chain reaction (PCR) assay. Genomic DNA was isolated from samples of whole blood samples collected at serial time points before and after cell infusion. of the y axis.. c The results of flow cytometry analysis showing in vivo expansion of bispecific CAR T cells in the peripheral blood and bone marrow of representative patient 5, who achieved a MRD negative. Both the x and y axes are log10 scales. MRD, minimal residual disease
Fig. 3
Fig. 3
Serum cytokines, inflammatory markers, and B cell markers before and after bispecific CD19/CD22 CAR T cell therapy. a Serum levels of cytokines and inflammatory markers measured at the indicated time points after cell infusion. Analytes with a fold change greater than or equal to three are indicated and plotted as relative changes from the baseline (determined on day 0 before infusion). Increases in the levels of cytokines, including interleukin-1β, GM-CSF, interleukin-2, interleukin-6, and interleukin-10, tumor necrosis factor α, and interferon gamma, were observed in the three patients. b Serum ferritin and C-reactive protein concentrations. c Circulating blasts and low levels of nonmalignant B cells in patient 5 peripheral blood at day 1, followed by CAR T cell expansion coincident with the clearance of leukemia and nonmalignant B cells by day 20; the CD19/CD22 CAR T cells disappeared and B cell recovery occurred by day 100. Green dots show leukemic blasts (CD19+CD34+); red dots represent normal B cells
Fig. 4
Fig. 4
Persistent fevers in patients with acute lymphoblastic leukemia after infusion with bispecific CAR T cells. Changes in body temperature after cell infusion, including the maximum temperature per 24-h period in the six patients. The green line marks the minimum temperature for a fever (38 °C)
Fig. 5
Fig. 5
Changes in CD19 and CD22 cell surface expression in relapsed patient baseline and at the time of relapse. Bone marrow samples were obtained from patients 1 and 2 before cell infusion and at the time of relapse. Mononuclear cells isolated from the bone marrow samples were stained for CD45, CD34, CD19, and CD22. After gating on live cells, the blast gate (CD45+side scatter [SSC] low) was subgated on CD34+ cells, and histograms were generated for CD19 and CD22 expression. a CD19 and CD22 expression of B-ALL tumor cells in patient 1 from the initial diagnostic BM sample and the relapsed sample. b CD19 and CD22 expression of B-ALL tumor cells in patient 4 from the initial diagnostic BM sample and the relapsed sample. c Non-transduced (NT) T cells from the leukapheresis product or CAR T cells from the end-of-product formulated cells were incubated with the relapsed B-ALL tumor cells from patients 1, 2, and 4. Effectors were incubated with tumor cells at a 50:1 effector/target ratio for 4 h. Supernatants were harvested and analyzed using the CytoTox 96 Non-Radioactive Cytotoxicity Assay kit (Promega). d Changes in the CD19 cell surface expression in patient 2 between the baseline time point and the time of relapse. e Direct Sanger sequencing performed with patient 2 cDNA. Mutations in exon 2 of CD19 were found in the relapse samples from patient 2 and were predicted to result in a truncated protein. f Emergence of leukemia cell populations with CD22 expression different from that of the cells harvested before treatment. g The change in CD22 cell surface expression in patients 1, 2, and 4

References

    1. 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(5):439–48.
    1. June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379(1):64–73. doi: 10.1056/NEJMra1706169.
    1. Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra25. doi: 10.1126/scitranslmed.3008226.
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–528. doi: 10.1016/S0140-6736(14)61403-3.
    1. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517. doi: 10.1056/NEJMoa1407222.
    1. Dai H, Wang Y, Lu X, Han W. Chimeric antigen receptors modified T-cells for cancer therapy. J Natl Cancer Inst. 2016;108(7).
    1. Dai H, Zhang W, Li X, et al. Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology. 2015;4(11):e1027469. doi: 10.1080/2162402X.2015.1027469.
    1. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015;5(12):1282–1295. doi: 10.1158/-15-1020.
    1. Orlando EJ, Han X, Tribouley C, et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat Med. 2018;24(10):1504–1506. doi: 10.1038/s41591-018-0146-z.
    1. Evans AG, Rothberg PG, Burack WR, et al. Evolution to plasmablastic lymphoma evades CD19-directed chimeric antigen receptor T cells. Br J Haematol. 2015;171(2):205–209. doi: 10.1111/bjh.13562.
    1. Fry TJ, Shah NN, Orentas RJ, et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med. 2018;24(1):20–28. doi: 10.1038/nm.4441.
    1. Grada Z, Hegde M, Byrd T, et al. TanCAR: a novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol Ther Nucleic Acids. 2013;2:e105. doi: 10.1038/mtna.2013.32.
    1. Hegde M, Corder A, Chow KK, et al. Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma. Mol Ther. 2013;21(11):2087–2101. doi: 10.1038/mt.2013.185.
    1. Bielamowicz K, Fousek K, Byrd TT, et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro Oncol. 2018;20(4):506–518. doi: 10.1093/neuonc/nox182.
    1. Hegde M, Mukherjee M, Grada Z, et al. Tandem CAR T cells targeting HER2 and IL13Ralpha2 mitigate tumor antigen escape. J Clin Invest. 2016;126(8):3036–3052. doi: 10.1172/JCI83416.
    1. Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005;202(7):907–912. doi: 10.1084/jem.20050732.
    1. Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat Rev Cancer. 2013;13(10):739–752. doi: 10.1038/nrc3581.
    1. Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–2138. doi: 10.1172/JCI85309.
    1. van Dongen JJ, van der Velden VH, Bruggemann M, Orfao A. Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and standardized technologies. Blood. 2015;125(26):3996–4009.
    1. Park JH, Riviere I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449–59.
    1. Fielding AK, Richards SM, Chopra R, et al. Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood. 2007;109(3):944–950. doi: 10.1182/blood-2006-05-018192.
    1. Gokbuget N, Stanze D, Beck J, et al. Outcome of relapsed adult lymphoblastic leukemia depends on response to salvage chemotherapy, prognostic factors, and performance of stem cell transplantation. Blood. 2012;120(10):2032–2041. doi: 10.1182/blood-2011-12-399287.
    1. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
    1. Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33(6):540–549. doi: 10.1200/JCO.2014.56.2025.
    1. Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–195. doi: 10.1182/blood-2014-05-552729.
    1. Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–638. doi: 10.1016/j.bbmt.2018.12.758.
    1. Forman SJ, Rowe JM. The myth of the second remission of acute leukemia in the adult. Blood. 2013;121(7):1077–1082. doi: 10.1182/blood-2012-08-234492.
    1. Majzner RG, Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 2018;8(10):1219–1226. doi: 10.1158/-18-0442.
    1. Qin H, Ramakrishna S, Nguyen S, et al. Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol Ther Oncolytics. 2018;11:127–137. doi: 10.1016/j.omto.2018.10.006.
    1. Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol Res. 2016;4(6):498–508. doi: 10.1158/2326-6066.CIR-15-0231.
    1. Turtle CJ, Hanafi LA, Berger C, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116. doi: 10.1126/scitranslmed.aaf8621.
    1. Haneen Shalabi NNS, Fry TJ, Yates B, Delbrook C, Yuan C, Stetler-Stevenson M, Ramakrishna S. Intensification of lymphodepletion optimizes CAR re-treatment efficacy. Blood. 2017;130(Suppl. 1):3889.
    1. Maus MV, Haas AR, Beatty GL, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1(1):26–31. doi: 10.1158/2326-6066.CIR-13-0006.
    1. Hamieh M, Dobrin A, Cabriolu A, et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature. 2019;568(7750):112–116. doi: 10.1038/s41586-019-1054-1.
    1. Shalabi H, Kraft IL, Wang HW, et al. Sequential loss of tumor surface antigens following chimeric antigen receptor T-cell therapies in diffuse large B-cell lymphoma. Haematologica. 2018;103(5):e215–e2e8. doi: 10.3324/haematol.2017.183459.
    1. Wang N, Hu X, Cao W, et al. Efficacy and safety of CAR19/22 T-cell cocktail therapy in patients with refractory/relapsed B-cell malignancies. Blood. 2020;135(1):17–27. doi: 10.1182/blood.2019000017.
    1. Pan J, Yang JF, Deng BP, et al. High efficacy and safety of low-dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lymphoblastic leukemia patients. Leukemia. 2017;31(12):2587–2593. doi: 10.1038/leu.2017.145.
    1. Teachey DT, Rheingold SR, Maude SL, et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood. 2013;121(26):5154–5157. doi: 10.1182/blood-2013-02-485623.
    1. Risma K, Jordan MB. Hemophagocytic lymphohistiocytosis: updates and evolving concepts. Curr Opin Pediatr. 2012;24(1):9–15. doi: 10.1097/MOP.0b013e32834ec9c1.
    1. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 2018;24(6):731–738. doi: 10.1038/s41591-018-0041-7.
    1. Norelli M, Camisa B, Barbiera G, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–748. doi: 10.1038/s41591-018-0036-4.
    1. Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell Therapy for acute lymphoblastic leukemia. Cancer Discov. 2016;6(6):664–679. doi: 10.1158/-16-0040.
    1. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016;127(26):3321–30.

Source: PubMed

3
구독하다