Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma

Frederick L Locke, Sattva S Neelapu, Nancy L Bartlett, Tanya Siddiqi, Julio C Chavez, Chitra M Hosing, Armin Ghobadi, Lihua E Budde, Adrian Bot, John M Rossi, Yizhou Jiang, Allen X Xue, Meg Elias, Jeff Aycock, Jeff Wiezorek, William Y Go, Frederick L Locke, Sattva S Neelapu, Nancy L Bartlett, Tanya Siddiqi, Julio C Chavez, Chitra M Hosing, Armin Ghobadi, Lihua E Budde, Adrian Bot, John M Rossi, Yizhou Jiang, Allen X Xue, Meg Elias, Jeff Aycock, Jeff Wiezorek, William Y Go

Abstract

Outcomes for patients with refractory diffuse large B cell lymphoma (DLBCL) are poor. In the multicenter ZUMA-1 phase 1 study, we evaluated KTE-C19, an autologous CD3ζ/CD28-based chimeric antigen receptor (CAR) T cell therapy, in patients with refractory DLBCL. Patients received low-dose conditioning chemotherapy with concurrent cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) for 3 days followed by KTE-C19 at a target dose of 2 × 106 CAR T cells/kg. The incidence of dose-limiting toxicity (DLT) was the primary endpoint. Seven patients were treated with KTE-C19 and one patient experienced a DLT of grade 4 cytokine release syndrome (CRS) and neurotoxicity. Grade ≥3 CRS and neurotoxicity were observed in 14% (n = 1/7) and 57% (n = 4/7) of patients, respectively. All other KTE-C19-related grade ≥3 events resolved within 1 month. The overall response rate was 71% (n = 5/7) and complete response (CR) rate was 57% (n = 4/7). Three patients have ongoing CR (all at 12+ months). CAR T cells demonstrated peak expansion within 2 weeks and continued to be detectable at 12+ months in patients with ongoing CR. This regimen of KTE-C19 was safe for further study in phase 2 and induced durable remissions in patients with refractory DLBCL.

Keywords: CAR T; CD19; KTE-C19; diffuse large B cell lymphoma; refractory NHL.

Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Clinical Efficacy after KTE-C19 Infusion (A) Duration of response and survival post-infusion with KTE-C19. (B) CR at 30 days post KTE-C19 infusion in patient 5. Representative PET-CT scans at baseline and 30 days post KTE-C19 infusion in a patient with DLBCL relapsing after prior therapy with R-CHOP, R-ICE, and ASCT with Rituximab-gemcitabine-busulfan-melphalan+azacitidine-vorinostat.
Figure 2
Figure 2
Apheresis and Product Phenotype as Determined by Flow Cytometry Using CD45RA and CCR7 Cell Surface Markers N, naive; CM, central memory; EM, effector memory; Eff, effector. The bars and boxes show the minimum, maximum, median, and interquartile range.
Figure 3
Figure 3
Kinetics of Peripheral Blood CAR T Cells and Serum Biomarkers (A) PCR data demonstrates exponential expansion and persistence of CD19 CAR T cells in blood. Expansion occurs rapidly with peak levels achieved within the first 7–14 days post-KTE-C19 infusion (note: patient 7 was not tested). Persisting CD19 CAR T cells were detectable in six of six (100%) patients at week 4 and in four of five (80%) patients with samples available for testing at month 3. Three patients with ongoing CR had detectable CAR T cells at 12 months. Limit of detection of the qPCR assay is 0.001% or 1 × 10−5. (B) Analysis of patient serum reveals a biomarker profile composed of specific cytokines, chemokines, and effector proteins associated with KTE-C19 treatment. The expansion of CD19 CAR T cells (Figure 3A) was mirrored by induction and elevation of a range of cytokines that regulate proliferation, activation, and effector function. Induction of IL-15 occurs during conditioning chemotherapy and levels continue to rise post-infusion, promoting anti-CD19 CAR T cell expansion. CRP levels parallel CRS and generally resolve within the first 28 days. Granzyme B levels peak 3–7 days post-infusion, during peak anti-CD19 CAR T cell expansion, and provide evidence of effector function and tumor killing. (C) Heat map of serum biomarkers demonstrates sequential induction and gradual resolution within the first 2 weeks after KTE-C19 infusion of key cytokines, chemokines, and effector proteins. Patients 1–6 demonstrated similar baseline levels and post-infusion kinetics for induction of IL-15 (T cell proliferation), CRP (marker of inflammation), granzyme B (evidence of effector function), and IP-10 (chemokine that promotes CAR T cell homing). Early induction of IL-15, CRP, and IP-10 was observed 1–3 days post-infusion and effector function occurred around days 3–7 for these patients. In contrast, patient 7 demonstrated a dysregulated profile relative to the other six patients both at baseline (IL-15, CRP, and IP-10) and after KTE-C19 administration (all markers), indicative of an inflammatory state prior to KTE-C19.

References

    1. Chaganti S., Illidge T., Barrington S., Mckay P., Linton K., Cwynarski K., McMillan A., Davies A., Stern S., Peggs K., British Committee for Standards in Haematology Guidelines for the management of diffuse large B-cell lymphoma. Br. J. Haematol. 2016;174:43–56.
    1. Morton L.M., Wang S.S., Devesa S.S., Hartge P., Weisenburger D.D., Linet M.S. Lymphoma incidence patterns by WHO subtype in the United States, 1992-2001. Blood. 2006;107:265–276.
    1. Sehn L.H., Gascoyne R.D. Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity. Blood. 2015;125:22–32.
    1. Crump M., Kuruvilla J., Couban S., MacDonald D.A., Kukreti V., Kouroukis C.T., Rubinger M., Buckstein R., Imrie K.R., Federico M. Randomized comparison of gemcitabine, dexamethasone, and cisplatin versus dexamethasone, cytarabine, and cisplatin chemotherapy before autologous stem-cell transplantation for relapsed and refractory aggressive lymphomas: NCIC-CTG LY.12. J. Clin. Oncol. 2014;32:3490–3496.
    1. Matasar M.J., Czuczman M.S., Rodriguez M.A., Fennessy M., Shea T.C., Spitzer G., Lossos I.S., Kharfan-Dabaja M.A., Joyce R., Fayad L. Ofatumumab in combination with ICE or DHAP chemotherapy in relapsed or refractory intermediate grade B-cell lymphoma. Blood. 2013;122:499–506.
    1. Gisselbrecht C., Glass B., Mounier N., Singh Gill D., Linch D.C., Trneny M., Bosly A., Ketterer N., Shpilberg O., Hagberg H. Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era. J. Clin. Oncol. 2010;28:4184–4190.
    1. Nagle S.J., Woo K., Schuster S.J., Nasta S.D., Stadtmauer E., Mick R., Svoboda J. Outcomes of patients with relapsed/refractory diffuse large B-cell lymphoma with progression of lymphoma after autologous stem cell transplantation in the rituximab era. Am. J. Hematol. 2013;88:890–894.
    1. Seshadri T., Stakiw J., Pintilie M., Keating A., Crump M., Kuruvilla J. Utility of subsequent conventional dose chemotherapy in relapsed/refractory transplant-eligible patients with diffuse large B-cell lymphoma failing platinum-based salvage chemotherapy. Hematology. 2008;13:261–266.
    1. Telio D., Fernandes K., Ma C., Tsang R., Keating A., Crump M., Kuruvilla J. Salvage chemotherapy and autologous stem cell transplant in primary refractory diffuse large B-cell lymphoma: outcomes and prognostic factors. Leuk. Lymphoma. 2012;53:836–841.
    1. Crump M., Neelapu S.S., Farooq U., Van Den Neste E., Kuruvilla J., Ahmed M.A., Link B.K., Hay A.E., Cerhan J.R., Zhu L. Outcomes in refractory aggressive diffuse large B-cell lymphoma (DLBCL): results from the international SCHOLAR-1 study. J. Clin. Oncol. 2016;34 abstr 7516.
    1. Kochenderfer J., Somerville R., Lu T., Shi V., Yang J.C., Sherry R., Klebanoff C., Kammula U.S., Goff S.L., Bot A. Anti-CD19 chimeric antigen receptor T cells preceded by low-dose chemotherapy to induce remissions of advanced lymphoma. J. Clin. Oncol. 2016;34 abstr LBA3010.
    1. Hans C.P., Weisenburger D.D., Greiner T.C., Gascoyne R.D., Delabie J., Ott G., Müller-Hermelink H.K., Campo E., Braziel R.M., Jaffe E.S. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004;103:275–282.
    1. Lee D.W., Gardner R., Porter D.L., Louis C.U., Ahmed N., Jensen M., Grupp S.A., Mackall C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124:188–195.
    1. Kochenderfer J.N., Dudley M.E., Kassim S.H., Somerville R.P., Carpenter R.O., Stetler-Stevenson M., Yang J.C., Phan G.Q., Hughes M.S., Sherry R.M. 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;33:540–549.
    1. Kochenderfer J.N., Dudley M.E., Feldman S.A., Wilson W.H., Spaner D.E., Maric I., Stetler-Stevenson M., Phan G.Q., Hughes M.S., Sherry R.M. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012;119:2709–2720.
    1. Brentjens R.J., Davila M.L., Riviere I., Park J., Wang X., Cowell L.G., Bartido S., Stefanski J., Taylor C., Olszewska M. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl. Med. 2013;5:177ra38.
    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.
    1. Turtle C.J., Hanafi L.A., Berger C., Gooley T.A., Cherian S., Hudecek M., Sommermeyer D., Melville K., Pender B., Budiarto T.M. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 2016;126:2123–2138.
    1. Davila M.L., Riviere I., Wang X., Bartido S., Park J., Curran K., Chung S.S., Stefanski J., Borquez-Ojeda O., Olszewska M. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 2014;6:224ra25.
    1. Kochenderfer J.N., Feldman S.A., Zhao Y., Xu H., Black M.A., Morgan R.A., Wilson W.H., Rosenberg S.A. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J. Immunother. 2009;32:689–702.
    1. Klebanoff C.A., Gattinoni L., Torabi-Parizi P., Kerstann K., Cardones A.R., Finkelstein S.E., Palmer D.C., Antony P.A., Hwang S.T., Rosenberg S.A. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc. Natl. Acad. Sci. USA. 2005;102:9571–9576.
    1. Sallusto F., Lenig D., Förster R., Lipp M., Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712.
    1. Sommermeyer D., Hudecek M., Kosasih P.L., Gogishvili T., Maloney D.G., Turtle C.J., Riddell S.R. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016;30:492–500.
    1. Steel J.C., Waldmann T.A., Morris J.C. Interleukin-15 biology and its therapeutic implications in cancer. Trends Pharmacol. Sci. 2012;33:35–41.
    1. Maude S.L., Frey N., Shaw P.A., Aplenc R., Barrett D.M., Bunin N.J., Chew A., Gonzalez V.E., Zheng Z., Lacey S.F. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014;371:1507–1517.
    1. Topp M.S., Gökbuget N., Stein A.S., Zugmaier G., O’Brien S., Bargou R.C., Dombret H., Fielding A.K., Heffner L., Larson R.A. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16:57–66.
    1. Topp M.S., Gökbuget N., Zugmaier G., Klappers P., Stelljes M., Neumann S., Viardot A., Marks R., Diedrich H., Faul C. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J. Clin. Oncol. 2014;32:4134–4140.
    1. Brudno J.N., Kochenderfer J.N. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016;127:3321–3330.
    1. Namuduri M., Brentjens R.J. Medical management of side effects related to CAR T cell therapy in hematologic malignancies. Expert Rev. Hematol. 2016;9:511–513.
    1. Cheson B.D., Pfistner B., Juweid M.E., Gascoyne R.D., Specht L., Horning S.J., Coiffier B., Fisher R.I., Hagenbeek A., Zucca E., International Harmonization Project on Lymphoma Revised response criteria for malignant lymphoma. J. Clin. Oncol. 2007;25:579–586.
    1. Better M., Pugach O., Lu L., Somerville R., Kassim S., Kochenderfer J., Rosenberg S.A., Marshall M.A., Bot A., Nolop K.B. Rapid cell expansion (RACE) technology for production of engineered autologous T cell therapy: path toward manageable multi-center clinical trials in aggressive NHL with anti-CD19 CAR. J. Clin. Oncol. 2014;32(Suppl) abstr 3079.
    1. Kochenderfer J.N., Dudley M.E., Carpenter R.O., Kassim S.H., Rose J.J., Telford W.G., Hakim F.T., Halverson D.C., Fowler D.H., Hardy N.M. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–4139.

Source: PubMed

3
Abonner