Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells

Brian G Till, Michael C Jensen, Jinjuan Wang, Eric Y Chen, Brent L Wood, Harvey A Greisman, Xiaojun Qian, Scott E James, Andrew Raubitschek, Stephen J Forman, Ajay K Gopal, John M Pagel, Catherine G Lindgren, Philip D Greenberg, Stanley R Riddell, Oliver W Press, Brian G Till, Michael C Jensen, Jinjuan Wang, Eric Y Chen, Brent L Wood, Harvey A Greisman, Xiaojun Qian, Scott E James, Andrew Raubitschek, Stephen J Forman, Ajay K Gopal, John M Pagel, Catherine G Lindgren, Philip D Greenberg, Stanley R Riddell, Oliver W Press

Abstract

Adoptive immunotherapy with T cells expressing a tumor-specific chimeric T-cell receptor is a promising approach to cancer therapy that has not previously been explored for the treatment of lymphoma in human subjects. We report the results of a proof-of-concept clinical trial in which patients with relapsed or refractory indolent B-cell lymphoma or mantle cell lymphoma were treated with autologous T cells genetically modified by electroporation with a vector plasmid encoding a CD20-specific chimeric T-cell receptor and neomycin resistance gene. Transfected cells were immunophenotypically similar to CD8(+) effector cells and showed CD20-specific cytotoxicity in vitro. Seven patients received a total of 20 T-cell infusions, with minimal toxicities. Modified T cells persisted in vivo 1 to 3 weeks in the first 3 patients, who received T cells produced by limiting dilution methods, but persisted 5 to 9 weeks in the next 4 patients who received T cells produced in bulk cultures followed by 14 days of low-dose subcutaneous interleukin-2 (IL-2) injections. Of the 7 treated patients, 2 maintained a previous complete response, 1 achieved a partial response, and 4 had stable disease. These results show the safety, feasibility, and potential antitumor activity of adoptive T-cell therapy using this approach. This trial was registered at www.clinicaltrials.gov as #NCT00012207.

Figures

Figure 1
Figure 1
Schema of clinical protocol.
Figure 2
Figure 2
Expression of the CD20-specific cTCR. (A) Schematic diagram of the CD20-specific scFvFc:ζ chimeric T-cell receptor cDNA plasmid. (B) A representative Western blot analysis of cTCR expression performed using whole-cell lysates of preinfusion T cells from patient B, probed with mouse anti–human CD3ζ monoclonal Ab. Negative control was parental PBMCs, and positive control was transfected Jurkat cell line. A 21-kDa band corresponding to the endogenous CD3ζ chain and a 66-kDa band representing the expected cTCR protein were detected. The intermediate bands indicate degradation products or truncated forms of the cTCR.
Figure 3
Figure 3
Clonality of T cells produced by limiting dilution and in bulk culture. T-cell clonality was determined by flow cytometric T-cell receptor (TCR) Vβ spectratyping (top) and by PCR amplification of clonal V-J rearrangements at the TCRγ locus (bottom). Representative results for T cells produced by limiting dilution (A) and in bulk culture (B) are shown. (A) T cells produced by limiting dilution (patient B), showing clonal expression of Vβ17 in 98% of CD8+ T cells by Vβ spectratyping (top; ■) and showing 2 predominant TCRγ rearrangements (bottom). Because each T-cell clone can rearrange one or both of its TCRγ alleles, the 2 PCR products could represent either 1 T-cell clone with biallelic TCRγ rearrangements or 2 singly rearranged clones, although the single predominant Vβ17 clone identified by spectratyping would favor a single doubly rearranged clone. (B) T cells produced in bulk culture (patient G) showing oligoclonal Vβ expression in CD8+ T cells (16% Vβ16; 9% Vβ7.1; 3% each Vβ3, Vβ13.2 and Vβ17; 2% each Vβ1 and Vβ13.1; and 1% each Vβ5.1, Vβ13.6, Vβ21.3, and Vβ23) and 7 distinct TCRγ rearrangements by PCR (bottom; ■) that could correspond to between 4 and 7 different T-cell clones, depending on the number of singly and doubly rearranged clones (see Table S1). The □ in both top panels represent the average expression levels for each Vβ chain in normal polyclonal T-cell populations.
Figure 4
Figure 4
Growth curves of genetically modified T cells. Patient PBMCs were transfected with the scFvFc:ζ plasmid by electroporation after stimulation with OKT3. For patients A, B, and D, populations of G418-resistant T cells were generated by limiting dilution, and T-cell cultures exhibiting the most favorable cytotoxicity and growth profiles were selected for expansion to therapeutic numbers. For patients F through I, G418-resistant cells were grown as bulk cultures.
Figure 5
Figure 5
Immunophenotypes of infused T cells. The phenotypes shown were determined using multicolor flow cytometry and are expressed in terms of percentage of the population of infused cells. (A) CD8+ versus CD4+ cells, (B) cells with a CD8+ effector T-cell phenotype (CD8+/CD62L−/CCR7−/CD45RA−/CD127−), (C) cells with CD8+ central memory (CD62L+/CCR7+/CD45RA−/CD127+) versus effector memory (CD62L−/CCR7−/CD45RA−/CD127+) T-cell phenotypes, (D) cells with CD4+ central memory versus effector memory T-cell phenotypes, and (E) cells with a regulatory T-cell (Treg) phenotype (CD4+/CD25+/FoxP3+) are shown.
Figure 6
Figure 6
Cytotoxicity of modified T cells. Standard chromium release assays were performed using preinfusion-modified T cells at 8 to 12 days after restimulation, using the following MHC-mismatched target cells: EL4-CD20, a murine T-cell lymphoma line transfected to express the human CD20 molecule (----), the parental EL4 nontransfected CD20− line (…), and the Daudi CD20+ Burkitt lymphoma cell line (—), at the E:T ratios shown. The calculated specific cytolysis values are displayed as percentages. (A) The CD20-specific cytotoxicity of the reinfused T cells. Data shown represent the mean combined data from all treated patients (± 1 SD). Triplicate assays were performed for each patient. (B) CD20 expression of the target cell lines of EL4, EL4-CD20, and Daudi, as determined by flow cytometry using PE-labeled mouse anti–human CD20 Ab.
Figure 7
Figure 7
In vivo persistence of modified T cells. Genomic DNA was isolated from patient PBMCs collected at serial time points after T-cell infusions and used for quantitative real-time PCR using one primer within the human CD3ζ gene and the other from the adjacent CD4 transmembrane region in the scFvFc:ζ plasmid. The copy number of scFvFc:ζ-specific DNA based on quantitative reverse transcription-PCR results for all treated patients is shown. Arrows denote T-cell infusions, and horizontal black bars indicate the period of subcutaneous IL-2 injections for patients F, G, H, and I. Modified T cells were detectable for 12, 5, 21, 63, 63, 35, and 65 days, respectively, in the 7 patients.

References

    1. McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol. 1998;16:2825–2833.
    1. Foran JM, Rohatiner AZ, Cunningham D, et al. European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma. J Clin Oncol. 2000;18:317–324.
    1. Czuczman MS, Grillo-Lopez AJ, White CA, et al. Treatment of patients with low-grade B-cell lymphoma with the combination of chimeric anti-CD20 monoclonal antibody and CHOP chemotherapy. J Clin Oncol. 1999;17:268–276.
    1. Forstpointner R, Dreyling M, Repp R, et al. The addition of rituximab to a combination of fludarabine, cyclophosphamide, mitoxantrone (FCM) significantly increases the response rate and prolongs survival as compared with FCM alone in patients with relapsed and refractory follicular and mantle cell lymphomas: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood. 2004;104:3064–3071.
    1. Howard OM, Gribben JG, Neuberg DS, et al. Rituximab and CHOP induction therapy for newly diagnosed mantle-cell lymphoma: molecular complete responses are not predictive of progression-free survival. J Clin Oncol. 2002;20:1288–1294.
    1. Kaminski MS, Zasadny KR, Francis IR, et al. Iodine-131-anti-B1 radioimmunotherapy for B-cell lymphoma. J Clin Oncol. 1996;14:1974–1981.
    1. Gopal AK, Rajendran JG, Petersdorf SH, et al. High-dose chemo-radioimmunotherapy with autologous stem cell support for relapsed mantle cell lymphoma. Blood. 2002;99:3158–3162.
    1. Witzig TE, White CA, Wiseman GA, et al. Phase I/II trial of IDEC-Y2B8 radioimmunotherapy for treatment of relapsed or refractory CD20(+) B-cell non-Hodgkin's lymphoma. J Clin Oncol. 1999;17:3793–3803.
    1. Rezvani AR, Storer B, Maris M, et al. Nonmyeloablative allogeneic hematopoietic cell transplantation in relapsed, refractory, and transformed indolent non-Hodgkin's lymphoma. J Clin Oncol. 2008;26:211–7.
    1. Khouri IF, McLaughlin P, Saliba RM, et al. Eight-year experience with allogeneic stem cell transplantation for relapsed follicular lymphoma after nonmyeloablative conditioning with fludarabine, cyclophosphamide, and rituximab. Blood. 2008;111:5530–5536.
    1. Khouri IF, Lee MS, Saliba RM, et al. Nonablative allogeneic stem-cell transplantation for advanced/recurrent mantle-cell lymphoma. J Clin Oncol. 2003;21:4407–4412.
    1. Porter DL, Antin JH. Donor leukocyte infusions in myeloid malignancies: new strategies. Best Pract Res Clin Haematol. 2006;19:737–755.
    1. Bortin MM, Truitt RL, Rimm AA, Bach FH. Graft-versus-leukaemia reactivity induced by alloimmunisation without augmentation of graft-versus-host reactivity. Nature. 1979;281:490–491.
    1. Brentjens RJ, Latouche JB, Santos E, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med. 2003;9:279–286.
    1. Haynes NM, Trapani JA, Teng MWL, et al. Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation. J Immunol. 2002;169:5780–5786.
    1. Kershaw MH, Jackson JT, Haynes NM, et al. Gene-engineered T cells as a superior adjuvant therapy for metastatic cancer. J Immunol. 2004;173:2143–2150.
    1. Park JR, Digiusto DL, Slovak M, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–833.
    1. Jensen MC, Cooper LJ, Wu AM, Forman SJ, Raubitschek A. Engineered CD20-specific primary human cytotoxic T lymphocytes for targeting B-cell malignancy. Cytotherapy. 2003;5:131–138.
    1. Jensen M, Tan G, Forman S, Wu AM, Raubitschek A. CD20 is a molecular target for scFvFc:zeta receptor redirected T cells: implications for cellular immunotherapy of CD20+ malignancy. Biol Blood Marrow Transplant. 1998;4:75–83.
    1. Wang J, Press OW, Lindgren CG, et al. Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes. Mol Ther. 2004;9:577–586.
    1. Reff ME, Carner K, Chambers KS, et al. Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood. 1994;83:435–445.
    1. Chen EY, Press OW, Jensen MC. CD8 immunomagnetic selection and interleukin-15 (IL-15) facilitate the isolation of human CD8+ cytotoxic T-lymphocytes (CTL) genetically engineered to express an anti-CD20 chimeric T-cell receptor (cTCR). J Clin Oncol. 2004;22(suppl 14):2542.
    1. Cheson BD, Horning SJ, Coiffier B, et al. Report of an international workshop to standardize response criteria for non-Hodgkin's lymphomas. NCI Sponsored International Working Group. J Clin Oncol. 1999;17:2454–2460.
    1. Jensen MC, Clarke P, Tan G, et al. Human T lymphocyte genetic modification with naked DNA. Mol Ther. 2000;1:49–55.
    1. Wang J, Jensen M, Lin Y, et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther. 2007;18:712–725.
    1. Sprouse JT, Werling R, Hanke D, et al. T-cell clonality determination using polymerase chain reaction (PCR) amplification of the T-cell receptor gamma-chain gene and capillary electrophoresis of fluorescently labeled PCR products. Am J Clin Pathol. 2000;113:838–850.
    1. Riddell SR, Elliott M, Lewinsohn DA, et al. T-cell mediated rejection of gene-modified HIV-specific cytotoxic T lymphocytes in HIV-infected patients. Nat Med. 1996;2:216–223.
    1. Klebanoff CA, Gattinoni L, Restifo NP. CD8+ T-cell memory in tumor immunology and immunotherapy. Immunol Rev. 2006;211:214–224.
    1. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712.
    1. Berger C, Flowers ME, Warren EH, Riddell SR. Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation. Blood. 2006;107:2294–2302.
    1. Igarashi T, Ohtsu T, Fujii H, et al. Re-treatment of relapsed indolent B-cell lymphoma with rituximab. Int J Hematol. 2001;73:213–221.
    1. Maris MB, Sandmaier BM, Storer BE, et al. Allogeneic hematopoietic cell transplantation after fludarabine and 2 Gy total body irradiation for relapsed and refractory mantle cell lymphoma. Blood. 2004;104:3535–3542.
    1. Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A. 2002;99:16168–16173.
    1. Tan JT, Ernst B, Kieper WC, LeRoy E, Sprent J, Surh CD. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation of memory phenotype CD8+ cells but are not required for memory phenotype CD4+ cells. J Exp Med. 2002;195:1523–1532.
    1. Surh CD, Boyman O, Purton JF, Sprent J. Homeostasis of memory T cells. Immunol Rev. 2006;211:154–163.
    1. Antony PA, Piccirillo CA, Akpinarli A, et al. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. J Immunol. 2005;174:2591–2601.
    1. Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23:2346–2357.
    1. Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–129.
    1. Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305.
    1. McLaughlin P, Cabanillas F, Grillo-Lopez A, et al. IDEC-C2B8 anti-CD20 antibody: final report on a phase III pivotal trial in patients with relapsed low-grade or follicular lymphoma [abstract]. Blood. 1996;88:90a. Abstract 349.

Source: PubMed

3
Abonnieren