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.

Several lymphoma subtypes are incurable with standard chemotherapy and radiation, but immune-based therapies have emerged as effective treatment and offer a potential for cure. Monoclonal antibodies (Abs) against the B-cell lymphoma marker CD20 have activity alone,1,2  in combination with chemotherapy,3-5  or conjugated with radiation-emitting nuclides.6-8  Adoptive cellular therapy with nonmyeloablative allogeneic stem cell transplantation (SCT) or donor lymphocyte infusion (DLI) can eradicate tumors, resulting in long-term survival, even in highly chemotherapy-refractory lymphomas.9-11  Both of these immunotherapy approaches have limitations, however, because antibodies fail to cure many types of lymphoma, and SCT and DLI, although potentially curative, cannot be used in many patients because of significant toxicity and transplantation-related mortality

Because the graft-versus-tumor effect of SCT and DLI appears to be mediated by alloreactive donor T lymphocytes,12,13  generating T cells specific for tumor antigens minimally expressed in normal tissues is an attractive strategy for harnessing this antitumor effector activity. One technique involves genetically modifying autologous T cells to express a chimeric T-cell receptor (cTCR) that targets a tumor antigen and induces antigen-specific T-cell activation, proliferation, and killing. Because this antigen-induced activation of the T cell occurs in an MHC-independent fashion, a single vector can be used universally to confer recognition of a selected target antigen. By introducing the cTCR into autologous T cells, the risk of graft-versus-host disease is eliminated. Such genetically modified T cells have been designed to target antigens associated with a variety of tumors, with success in animal models14-16  and some early evidence of clinical efficacy in human subjects.17 

Our group has developed a technique to manufacture CD20-specific T cells by transfecting peripheral blood mononuclear cells (PBMCs) with a linearized naked DNA plasmid encoding a cTCR derived from a murine anti–human CD20 Ab.18-20  The cell-surface antigen CD20 is an attractive target for immune-based therapies because it is present in more than 90% of B-cell lymphomas, is expressed at a high copy number, is stable on the cell surface, and does not internalize on binding Abs.21  These modified T cells secrete interleukin-2 (IL-2) in an antigen-dependent manner,19  selectively kill CD20+ target cells in vitro,20  and eradicate human xenograft tumors in mice.22  Application of this approach to the treatment of lymphoma in human subjects has not yet been described. We report here the results of a proof-of-concept clinical trial in which ex vivo–expanded, genetically modified autologous CD20-specific T cells were used as adoptive cellular therapy for patients with relapsed or refractory indolent B-cell non-Hodgkin lymphoma (NHL) and mantle cell lymphoma (MCL). We show that these T cells can be reproducibly generated and expanded to therapeutic numbers, exhibit in vitro antitumor cytotoxicity, persist in vivo for up to 9 weeks, and appear to be safe, well tolerated, and potentially capable of mediating in vivo antitumor activity.

Clinical protocol

This clinical protocol was approved by the Fred Hutchinson Cancer Research Center Institutional Review Board, the University of Washington Institutional Biosafety Committee, the US Food and Drug Administration, and the Recombinant DNA Advisory Committee of the National Institutes of Health. Informed consent was obtained in accordance with the Declaration of Helsinki. Patients were eligible if they had a pathologically confirmed diagnosis of CD20+ MCL or indolent B-cell lymphoma, had relapsed or refractory disease after at least one prior chemotherapy, were deemed not to be candidates for (or refused) stem cell transplantation, and had serologic evidence of prior Epstein-Barr virus (EBV) exposure (because the TM-LCL cell line used in T-cell culture is EBV-transformed). Patients were excluded if they received fludarabine or cladribine within 2 years before apheresis (but could receive these drugs as cytoreductive therapy after apheresis), anti-CD20 Ab within 4 months of T-cell infusions, or chemotherapy within 4 weeks of T-cell infusions; had lymph nodes more than 5 cm or more than 5000 circulating lymphoma cells in the peripheral blood at the time of T-cell infusions, a previous allogeneic stem cell transplantation, or human anti–mouse Ab (HAMA) seropositivity; required corticosteroids during the study period; had pulmonary or central nervous system involvement with lymphoma; were HIV-seropositive; or were pregnant.

Patients underwent leukapheresis after signing informed consent, and then they were allowed to receive cytoreductive chemotherapy for disease control or debulking during the 2- to 4-month period of T-cell generation, at the discretion of their referring physician. For patients A to E, PBMCs were activated, transfected, and plated at limiting dilution with the intention of isolating and subsequently expanding T-cell clones. This approach proved to be laborious and inefficient, however, and the protocol was modified for patients F to I to allow expansion of modified cells in bulk culture. Patients subsequently received 3 infusions of autologous CD20-specific T cells 2 to 5 days apart in escalating doses (108 cells/m2, 109 cells/m2, and 3.3 × 109 cells/m2) followed by 14 days of subcutaneous low-dose (500 000 IU/m2) interleukin-2 (IL-2) injections twice daily (patients F-I only). Patients then underwent clinical follow-up to evaluate toxicities related to therapy, which were assessed according to National Institutes of Health Common Terminology Criteria for Adverse Events, version 3.0 (http://ctep.cancer.gov/). A Data and Safety Monitoring Board was assembled that performed reviews of the safety data every 6 months. Clinical responses were assessed according to International Working Group criteria.23 

T-cell transfection, selection, and expansion

All cell culture for therapeutic use was performed in the Cell and Gene Therapy Core Laboratory at the University of Washington General Clinical Research Center, under current good manufacturing practice standards. PBMCs collected by apheresis were diluted 1:2 with PBS containing 200 mg/L EDTA, isolated by density gradient centrifugation over Ficoll-Paque (GE Healthcare, Little Chalfont, United Kingdom), washed, and resuspended in RPMI 1640 medium containing 2 mmol of l-glutamine, 25 mmol HEPES, and 10% fetal calf serum. Cells were activated with 30 ng/mL OKT3, and after overnight incubation recombinant human IL-2 was added (50 U/mL).

Electroporation and selection.

On day 4 of culture, cells were harvested and resuspended in chilled hypo-osmolar electroporation buffer (Eppendorf North America, New York, NY) at 20 × 106 cells/mL. Cell suspensions were mixed with linearized plasmids (25 μg/mL) encoding a CD20-specific scFvFc:ζ cTCR,18-20,24  and divided into aliquots into chilled 0.2-cm electroporation cuvettes. Cells were electroporated with an Eppendorf Multiporator at 250 V for 40 microseconds (μsec) as previously described.20  Approximately 3 days after electroporation, G418 was added to flasks (at 0.8 mg/mL). Cells were selected by G418 for 8 days before generating cells by limiting dilution.

Generation and expansion of genetically modified T cells.

Transfected cells from patients A through E were selected in G418, and attempts were made to generate T-cell clones by limiting dilution as previously described.24,25  Although the intention was to isolate clonal populations derived from a single progenitor cell, the plating density required to yield reliable growth of T cells resulted in the presence of 1 to 3 clones per well, as subsequently determined by Vβ TCR spectratyping. For patients F through I, G418-resistant transfected cells were grown in bulk cultures as previously described.25  As cell numbers increased, T cells were transferred to 1-L or 3-L tissue culture bags (Lifecell, Branchburg, NJ). During the expansion, 5 to 8 stimulation cycles were performed. Fresh T cells were infused in patients A and B. For logistic reasons, T cells from patients D, F, G, H, and I were cryopreserved between days 70 and 132 after apheresis in Plasmalyte-A containing 5% HSA and 10% DMSO and thawed 3 to 4 hours before infusion (48 hours before infusion for patient D). Release criteria included detectable cTCR expression by flow cytometry, negative bacterial, fungal, and Mycoplasma cultures, endotoxin level no more than 5 EU/kg per hour, Gram stain–negative on day of infusion, greater than 80% cell viability, TCRα/β+ and CD3+ phenotype by flow cytometry, IL-2 growth dependence, and CD20-specific cytotoxicity.

T-cell clonality assays

T-cell clonality was determined by polymerase chain reaction (PCR) amplification of rearrangements at the T-cell receptor gamma (TCRγ) locus as previously described,26  except that VγI-Jγ1/2, VγII-Jγ1/2, VγI-JγP1/2, and VγII-JγP1/2 rearrangements were amplified in a single multiplex PCR reaction and analyzed by capillary electrophoresis on an Applied Biosystems Model 3130 (Foster City, CA). See Document S1 (available on the Blood website; see the Supplemental Materials link at the top of the online article) for detailed methods.

Vβ spectratyping was also performed by flow cytometry. Cells were labeled with monoclonal antibodies CD8 ECD and IOTest Beta Mark Kit (Beckman Coulter, Fullerton, CA). The expression of each of the 24 T-cell receptor isoforms present in the Beta Mark Kit (approximately 70% coverage of the normal human TCR Vβ repertoire) were determined independently on the CD8+ T-cell populations, and a threshold of 85% positivity for a single isoform or an absence of expression of all 24 isoforms outside the reference range was considered to represent a clonal expansion. Samples showing 2 or more isoforms outside the reference range were considered oligoclonal.

Western blot assay

Whole cell lysates of modified T cells were probed with a mouse anti–human CD3ζ monoclonal Ab (BD PharMingen, San Diego, CA) as previously described.20 

Cytotoxicity assays

T-cell cytotoxicity was analyzed 2 to 7 weeks before planned T-cell infusions to permit selection of optimal “clones” of T cells for expansion. CD20-specific cytotoxicity was assessed with the use of standard chromium-release assays with the following target cell lines: EL4-CD20 (a murine T-cell lymphoma line transfected to express the human CD20 molecule), the parental CD20–nontransfected EL4 cell line, or the Daudi Burkitt lymphoma cell line, as previously described.25  Cytotoxicity assays were repeated in some patients just before T-cell infusions and showed levels of cytotoxicity comparable to assays performed 2 to 7 weeks before infusion.

Flow cytometry for immunophenotypic characterization of T cells and lymphocyte subset analysis

Flow cytometry was performed with the use of standard methods. Briefly, cells cryopreserved within 1 day of the first T-cell infusion were thawed, washed, and labeled with the indicated monoclonal Ab for 15 minutes at room temperature in the dark. The samples were then washed once, resuspended in a dilute DNA binding dye (DAPI), incubated for 10 minutes, and approximately 20 000 events acquired on an LSRII flow cytometer (Becton Dickinson, Franklin Lakes, NJ). Data were analyzed using software developed in our laboratory (WoodList). Positivity for DAPI was used to exclude nonviable cells, and thresholds for positivity were determined with unstained cells and isotype control Ab, as appropriate. Antibodies were used at the manufacturer's recommended concentrations. A complete list of Abs used is included in Document S1. Flow cytometry to detect cTCR expression was performed using a FITC-labeled polyclonal goat anti–mouse IgG Fab-specific Ab (Sigma-Aldrich, St Louis, MO) as previously described.25 

Detection of modified T cells in vivo

PBMCs collected serially after T-cell infusions were isolated by Ficoll density-gradient centrifugation, and genomic DNA was extracted using a QIAamp DNA Blood Mini Kit (Qiagen, Valencia, CA). The standard consisted of 10-fold serial dilutions of purified scFvFc:ζ plasmid DNA starting at 106 copies/μL, with each sample containing 1 μg of preinfusion PBMC DNA to control for background signal. The negative control was preinfusion PBMC genomic DNA. A 72-bp (base pair) fragment containing portions of the CD3ζ chain and adjacent CD4 transmembrane domain sequences was amplified using forward primer 5′-TCGCCGGCCTCCTGCTTT-3′ and reverse primer 5′-CGTCTGCGCTCCTGCTGA-3′. The probe used was 5′-FAM-TGGGCTAGGCATCTTCTTCAGAGTGAA-TAMRA-3′. Primers that amplify a fragment of the β-actin gene (TaqMan B-actin Detection Reagent Kit; Applied Biosystems) were used as an internal control and for normalization of DNA quantities. Quantitative real-time PCR was performed in triplicate with 1 μg DNA in each reaction, using TaqMan Universal PCR Master Mix in a 7900HT Sequence Detection System (all Applied Biosystems).

Immune response assays

Two assays were performed to test for humoral immune responses to the cTCR. In the first assay, 96-well enzyme-linked immunoabsorbent assay (ELISA) plates were coated with 0.5 μg Leu-16 murine anti–human CD20 Ab (BD Biosciences, San Diego, CA) in pH 9.6 carbonate buffer and blocked with 5% milk before adding samples of goat anti–mouse IgG Fab-specific Ab (standard curve; Jackson ImmunoResearch Laboratories, West Grove, PA), serially diluted 2% BSA/PBS (negative control), baseline patient serum (negative control), HAMA+ patient serum (positive control), or study subject serum from serial postinfusion time points. Biotinylated Leu-16 murine anti–human CD20 Ab (BD Biosciences; 10 μg/mL) was added to each well as the primary Ab, followed by 1:1000 horseradish peroxidase-Avidin D (BD Biosciences). Samples were incubated for 30 minutes at room temperature and washed 3 times with 0.01 M PBS/0.3% Tween between each step. Color reagent (2,2,-azino-bis[3-ethylbenzothiazoline-6-sulfonic acid] diammonium salt; Sigma-Aldrich) at 0.42 mg/mL in citrate buffer (citrate 10.5 mg/mL, pH 4.0) plus hydrogen peroxide (100 μL/12 mL buffer) was added to each well; absorbency was read with a Bio-Tek XS ELISA reader (Bio-Tek Instruments, Winooski, VT). Optical density measurements were converted to concentration values as calculated from the standard curve. In the second assay, flow cytometry was used to assess the presence of anti-cTCR Ab in posttreatment patient serum samples (see Document S1 for detailed methods).

Cellular immune response assays were performed by coincubating patient-derived PBMCs (106 cells/mL) serially collected after T-cell infusions with irradiated anti-CD20 cTCR-expressing T cells (106 cells/mL) from infused batches, at a 2:1 ratio. After 2 rounds of stimulation 1 week apart, the PBMCs were tested in 51Cr release cytotoxicity assays using either autologous T cells transfected with the cTCR-encoding plasmid or nontransfected autologous PBMC as target cells at a 25:1 E/T ratio. In the first 2 patients treated we also assessed the responsiveness of recovered T cells to histocompatibility locus antigen–disparate cells as a positive control.

Study design and patient characteristics

The primary objective of this study was to assess the feasibility, safety, and toxicity of adoptive therapy using patient-derived T cells bearing a CD20-specific cTCR to treat indolent and mantle cell lymphomas. Autologous PBMCs were collected by apheresis, genetically modified, and expanded ex vivo, a process that typically required 2 to 4 months. During this interval patients underwent cytoreductive chemotherapy if necessary for tumor debulking or to maintain disease control. Subjects were then treated with 3 infusions of modified CD20-specific T cells, 2 to 5 days apart, at incremental doses (108 cells/m2, 109 cells/m2, and 3.3 × 109 cells/m2) similar to those used in previous adoptive T-cell therapy trials,27  but with a shorter interval between infusions to limit the potential for development of an immune response against the transfected cells. The last 4 patients received low-dose subcutaneous injections of IL-2 twice daily for 14 days after the final T-cell infusion to enhance in vivo T-cell survival and proliferation. Patients then underwent follow-up for clinical and research end points, and long-term monitoring for adverse events for 2 years. The study design is outlined in Figure 1.

Figure 1

Schema of clinical protocol.

Figure 1

Schema of clinical protocol.

Close modal

Nine patients with relapsed or refractory indolent B-cell NHL or MCL were enrolled: 8 men and 1 woman between the ages of 43 and 77 years; 8 had relapsed follicular lymphoma, and 1 had relapsed MCL. Patients had been treated with a median of 2 prior therapies (range, 1-7 therapies; Table 1).

Table 1

Patient characteristics

PatientAge, ySexDiagnosisStagePrior therapiesCytoreductive therapy before T-cell infusions
44 FL IV-B R-CHOP CVP 
70 FL II-A CHOP, rituximab, 131I-tositumomab CVP 
47 FL IV-B ProMACE/MOPP, ASCT, fludarabine (10 cycles) CVP 
60 FL IV-A Rituximab CVP 
63 MCL IV-A R-HyperCVAD, GCD-R None 
46 FL IV-A R-CVP FND 
43 FL IV-A CHOP, IFN, CY + VP16, R-CY, CY + DEX, GCD-R, ASCT None 
46 FL IV-B R-CHOP, fenretinide FND 
77 FL III-A R-CVP, R-CHOP, GCD-R 131I-tositumomab 
PatientAge, ySexDiagnosisStagePrior therapiesCytoreductive therapy before T-cell infusions
44 FL IV-B R-CHOP CVP 
70 FL II-A CHOP, rituximab, 131I-tositumomab CVP 
47 FL IV-B ProMACE/MOPP, ASCT, fludarabine (10 cycles) CVP 
60 FL IV-A Rituximab CVP 
63 MCL IV-A R-HyperCVAD, GCD-R None 
46 FL IV-A R-CVP FND 
43 FL IV-A CHOP, IFN, CY + VP16, R-CY, CY + DEX, GCD-R, ASCT None 
46 FL IV-B R-CHOP, fenretinide FND 
77 FL III-A R-CVP, R-CHOP, GCD-R 131I-tositumomab 

F indicates female; M, male; FL, follicular lymphoma; MCL, mantle cell lymphoma; CHOP, cyclophosphamide, doxorubicin, vincristine, and prednisone; R, rituximab; CVP, cyclophosphamide, vincristine, and prednisone; ProMACE/MOPP, procarbazine, methotrexate with leucovorin, doxorubicin, cyclophosphamide, etoposide, mechlorethamine, vincristine, and prednisone; ASCT, high-dose therapy followed by autologous stem cell transplantation; HyperCVAD, cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with cycles of high-dose cytarabine and methotrexate; GCD, gemcitabine, carboplatin, and dexamethasone; FND, fludarabine, mitoxantrone, and dexamethasone; IFN, interferon-α; CY, cyclophosphamide; VP16, etoposide; and DEX, dexamethasone.

Generation and expansion of autologous CD20-specific T cells

PBMCs collected by apheresis were stimulated with anti-CD3 Ab (OKT3) and IL-2 and transfected by electroporation with a naked DNA plasmid encoding a cTCR consisting of a murine kappa leader sequence, CD20-specific scFv derived from the Leu16 murine Ab, human IgG1 CH2CH3 hinge, human CD4 transmembrane, and human CD3ζ intracellular signaling domain, as well as a neomycin resistance gene (neoR) under a separate promoter (Figure 2A).20,25  Anti-CD20 cTCR surface expression was confirmed by Western blot (Figure 2B) and flow cytometry (Figure S3).

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 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.

Close modal

Modified T cells were generated for the first 5 patients by limiting dilution and selected for CD20 cytotoxicity by chromium release assay and cTCR expression by flow cytometry. At the plating density required to reproducibly generate modified T cells, the resulting T-cell populations consisted of cells derived from 1 to 3 clones of T cells as assessed by Vβ TCR spectratyping and TCRγ clonality testing by PCR (Figure 3A; Table S1). This expansion and selection process proved to be laborious and inefficient, requiring approximately 4 months to achieve the target cell dose. Moreover, T cells generated by limiting dilution could not be expanded adequately for infusions in 2 of these initial 5 patients, and in 2 of the other 3 patients the target cell doses could not be reached (Table 2).

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 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.

Close modal
Table 2

T-cell infusions

PatientInfusion 1, cells/m2*Infusion 2, cells/m2Infusion 3, cells/m2Fresh versus thawed cellsTime from apheresis to target cell number, dNo. of stimulation cycles§
108 109 3.3 × 109 Fresh 130 
108 109 2 × 109 Fresh 129+ 
    Expansion failed 
108 4 × 108  Thawed 159+ 
    Expansion failed 
108 109 3.3 × 109 Thawed 96 
108 109 3.3 × 109 Thawed 90 
108 109 3.3 × 109 Thawed 81 
108 109 2 × 109 Thawed 104 
PatientInfusion 1, cells/m2*Infusion 2, cells/m2Infusion 3, cells/m2Fresh versus thawed cellsTime from apheresis to target cell number, dNo. of stimulation cycles§
108 109 3.3 × 109 Fresh 130 
108 109 2 × 109 Fresh 129+ 
    Expansion failed 
108 4 × 108  Thawed 159+ 
    Expansion failed 
108 109 3.3 × 109 Thawed 96 
108 109 3.3 × 109 Thawed 90 
108 109 3.3 × 109 Thawed 81 
108 109 2 × 109 Thawed 104 

For patients A through E, T cells were selected and expanded by limiting dilution. For patients F through I, T cells were expanded in bulk culture. Patients C and E did not receive T-cell infusions. Patients B and D received infusions but did not reach the target cell dose.

*

Target dose was 108 cells/m2.

Target dose was 109 cells/m2.

Target dose was 3.3 × 109 cells/m2.

§

Defined as the number of times cells were stimulated with OKT3 in the presence of irradiated feeder PBMCs and LCL (repeated every 14 days, first cycle at day 14).

The target number of cells was generated for this patient, but nearly half of the last cell dose was lost during a quality control assay before infusion.

We subsequently elected to modify the protocol to include expansion of T-cell transfectants in bulk culture to circumvent the difficulties of expanding T cells after limiting dilution. Successful expansion of modified T cells was achieved for the subsequent 4 patients using this approach, and the time required to reach the target cell dose was reduced by approximately 50% (Figure 4). Vβ TCR spectratyping and TCRγ clonality testing by PCR showed more heterogeneous T-cell populations in these bulk cultures, although several of the cultures contained prominent T-cell clones (Figure 3B; Table S1). Three of these 4 patients received all planned doses of T cells. The target cell number was reached for the fourth patient as well, but the third infusion consisted of only 2 × 109 cells/m2 because of a loss of cells during a quality control assay.

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 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.

Close modal

Immunophenotype of modified T cells

The phenotype of ex vivo–expanded cTCR-bearing T cells has not been well described. We analyzed the immunophenotype of the infused T cells using multicolor flow cytometry and found it to be similar to that of activated effector T cells,28,29  expressing CD3, CD8, and CD45RO and lacking CD62L, CCR7, and CD127 (Figure 5). As expected, patients treated with CD8+ T cells derived by limiting dilution received negligible numbers of CD4-expressing cells (0.67%-4.5%), whereas patients receiving infusions of T cells grown in bulk culture received 3.4% to 38.6% CD4+ cells. Infused T cells also expressed the activation marker CD95, but relatively few cells (1.3%-6.2%) expressed CD134 (OX40; Table 3). We found negligible numbers of cells expressing central memory (CD62L+/CCR7+/CD45RA/CD127+) and effector memory (CD62L/CCR7/CD45RA/CD127+) phenotypes.T cells resulting from bulk culture and from limiting dilution were phenotypically similar, although the former generally contained higher proportions of CD4+ cells (3.4%-38.6% compared with 0.67%-4.5%) and cells with a regulatory T (Treg)–like phenotype (CD4+/CD25+/FoxP3+) (0.54%-1.9% compared with 0.61%-21.6%). Treg functionality studies were not performed, however. All T cells exhibited low expression of costimulatory markers CD28 (0.92%-5.4%) and CD137 (0.47%-4.4%). High proportions of cells from all patients expressed adhesion molecules such as CD11a (98.7%-100%), CD44 (99.8%-100%), and CD49d (85.8%-99.6%).

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 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.

Close modal
Table 3

Flow cytometric immunophenotypes of infused T cells

MarkerPatient APatient BPatient DPatient FPatient GPatient HPatient I
CD2 100 100 99 100 99.9 100 99.8 
CD3 99 99.8 99 99.9 99.9 99.9 99.8 
CD4 0.67 3.2 4.5 38.6 13.6 3.4 7.8 
CD5 5.4 25.3 93 85.8 97.3 93.8 92.3 
CD8 99.8 99.7 98.5 98.5 97.7 99.4 99.6 
CD11a 100 100 98.7 99.9 99.8 99.9 99.8 
CD25 2.4 45.9 22.1 23.2 52.5 52.7 16.6 
CD28 0.92 3.2 4.4 2.6 5.4 1.9 2.7 
CD44 99.8 100 99.5 99.9 100 99.9 99.9 
CD45RA 61.5 14.1 2.3 3.1 9.5 2.2 23.0 
CD45RO 96.8 99.3 99.1 99.4 99.4 99.2 99.8 
CD49d 85.8 98.7 91.2 95.2 99.6 98.1 97.3 
CD56 99.9 98.4 98.7 95.7 91.7 95.8 99.3 
CD62L 1.3 6.8 4.6 2.1 6.2 2.7 2.5 
CD94 98.8 67.3 38.7 32.8 7.1 17.6 33.6 
CD95 99.4 99.5 99.8 98.1 98.4 99.6 98.6 
CD134 1.3 2.9 6.2 4.1 5.3 3.2 4.5 
CD137 (4-1BB) 0.47 1.9 4.4 1.2 3.1 0.69 3.7 
CD154 (CD40L) 12.5 3.3 3.2 4.4 2.6 0.64 24.4 
CD314 (NKG2D) 99.5 97.7 92.6 93.7 99.2 95.3 97.4 
CCR5 (CD195) 58.7 43.1 58.1 27.9 85.9 6.0 96.8 
CCR6 5.5 31.1 10.8 3.4 10.7 12.3 9.9 
CCR7 0.21 2.3 4.2 2.1 4.6 0.75 5.4 
CXCR3 97 89.9 91.9 95.2 97.5 93.1 98.1 
CXCR4 6.5 9.6 6.1 3.9 21.1 9.7 2.6 
CXCR5 1.1 3.1 3.8 2.7 6.1 2.0 3.5 
TCR α/β 92.8 99.2 98.7 99 96 99.7 99.7 
MarkerPatient APatient BPatient DPatient FPatient GPatient HPatient I
CD2 100 100 99 100 99.9 100 99.8 
CD3 99 99.8 99 99.9 99.9 99.9 99.8 
CD4 0.67 3.2 4.5 38.6 13.6 3.4 7.8 
CD5 5.4 25.3 93 85.8 97.3 93.8 92.3 
CD8 99.8 99.7 98.5 98.5 97.7 99.4 99.6 
CD11a 100 100 98.7 99.9 99.8 99.9 99.8 
CD25 2.4 45.9 22.1 23.2 52.5 52.7 16.6 
CD28 0.92 3.2 4.4 2.6 5.4 1.9 2.7 
CD44 99.8 100 99.5 99.9 100 99.9 99.9 
CD45RA 61.5 14.1 2.3 3.1 9.5 2.2 23.0 
CD45RO 96.8 99.3 99.1 99.4 99.4 99.2 99.8 
CD49d 85.8 98.7 91.2 95.2 99.6 98.1 97.3 
CD56 99.9 98.4 98.7 95.7 91.7 95.8 99.3 
CD62L 1.3 6.8 4.6 2.1 6.2 2.7 2.5 
CD94 98.8 67.3 38.7 32.8 7.1 17.6 33.6 
CD95 99.4 99.5 99.8 98.1 98.4 99.6 98.6 
CD134 1.3 2.9 6.2 4.1 5.3 3.2 4.5 
CD137 (4-1BB) 0.47 1.9 4.4 1.2 3.1 0.69 3.7 
CD154 (CD40L) 12.5 3.3 3.2 4.4 2.6 0.64 24.4 
CD314 (NKG2D) 99.5 97.7 92.6 93.7 99.2 95.3 97.4 
CCR5 (CD195) 58.7 43.1 58.1 27.9 85.9 6.0 96.8 
CCR6 5.5 31.1 10.8 3.4 10.7 12.3 9.9 
CCR7 0.21 2.3 4.2 2.1 4.6 0.75 5.4 
CXCR3 97 89.9 91.9 95.2 97.5 93.1 98.1 
CXCR4 6.5 9.6 6.1 3.9 21.1 9.7 2.6 
CXCR5 1.1 3.1 3.8 2.7 6.1 2.0 3.5 
TCR α/β 92.8 99.2 98.7 99 96 99.7 99.7 

Values represent percentages of total cell population.

Cytotoxicity of modified T cells in vitro

Our group showed in preclinical studies that T-cell clones bearing CD20-specific cTCRs exhibit antigen-specific cytotoxicity.20  We assessed the cytotoxicity of T cells used in this trial by coincubation with 51Cr-labeled CD20+ target cells (Daudi lymphoma cells and EL4 mouse lymphoma cells transfected to express human CD20), and the expanded T cells used for all 7 patients killed CD20+ lymphoma cells in an antigen-specific manner (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 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.

Close modal

Adoptive therapy with CD20-specific T cells is safe and well tolerated

Seven patients received a total of 20 T-cell infusions. No grade 3 or 4 toxicities were seen, and no adverse events attributable to the T-cell infusions themselves were observed. Grade 2 toxicities associated with subcutaneous IL-2 injections included a flulike syndrome (1 of 4 patients), fever (1 of 4 patients), and skin reactions at injection site (2 of 4 patients) and resolved after the cessation of IL-2. Grade 1 toxicities attributable to IL-2 included chills, myalgias, dyspnea, dysgeusia, malaise or fatigue, diaphoresis, and injection site reactions (Table 4).

Table 4

Adverse events possibly or probably related to the treatment regimen

Patient*Grade 1Grade 2
None None 
None None 
None None 
Chills, myalgias, and shortness of breath (all during IL-2 treatment) Injection site skin reaction (during IL-2 treatment) 
None Injection site skin reaction (during IL-2 treatment) 
Dysgeusia, fatigue, diaphoresis, and injection site skin reactions (all during IL-2 treatment) Flulike syndrome (during IL-2 treatment) 
Malaise/fatigue, chills, injection site reactions, and dyspnea (all during IL-2 treatment) Fever (during IL-2 treatment) 
Patient*Grade 1Grade 2
None None 
None None 
None None 
Chills, myalgias, and shortness of breath (all during IL-2 treatment) Injection site skin reaction (during IL-2 treatment) 
None Injection site skin reaction (during IL-2 treatment) 
Dysgeusia, fatigue, diaphoresis, and injection site skin reactions (all during IL-2 treatment) Flulike syndrome (during IL-2 treatment) 
Malaise/fatigue, chills, injection site reactions, and dyspnea (all during IL-2 treatment) Fever (during IL-2 treatment) 

None of the patients experienced any grade 3 or grade 4 events.

*

Patients C and E are omitted because they did not receive T-cell infusions.

In vivo persistence of modified T cells

We measured the in vivo persistence of modified T cells using quantitative real-time PCR of DNA from patient PBMCs collected at serial time points after T-cell infusion. Modified T cells were detectable by PCR for 5 to 21 days in the first 3 patients receiving T cells without adjuvant IL-2. In contrast, modified T cells were detectable for 5 to 9 weeks in the last 4 patients, who received T cells produced in bulk culture and 14 days of subcutaneous IL-2 (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.

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.

Close modal

Modified T cells were also detectable by PCR in the bone marrow 24 hours after the final T-cell infusion in patients A, D, F, and G (lymphoma cells were detectable in the marrow by flow cytometry only in patients G and H at this time point). Of the 7 treated patients, only patients B and G had accessible lymph nodes for biopsy after T-cell infusions. The lymph node from patient B showed only fibroadipose tissue, and patient G's lymph node showed tumor, but no modified T cells were detectable.

Modified T cells were not immunogenic

The introduction of foreign transgenes in therapeutic vectors has resulted in immune responses against modified T cells in previous gene therapy clinical trials.27,30  Given the significant immunosuppression present in patients with lymphoma, we hypothesized that the transgenic cells in this trial might elicit lower immune response rates, and we used 3 assays to test this hypothesis. To detect cellular immune responses, 51Cr-release assays were performed with serially collected patient PBMCs that had been coincubated with irradiated T cells expressing the scFvFc:ζ and neoR gene products (Figure S1A), using transfected and nontransfected T cells as target cells. In 2 patients the antigen responsiveness of recovered T cells was confirmed using allogeneic LCL as target cells; in both cases a cytotoxic response was elicited at all time points tested (Figure S1B). For humoral immune responses, patient serum collected at various time points after T-cell infusion was assessed in an ELISA assay in which the Leu-16 murine anti–human CD20 Ab served as an internal control (Figure S1C). Although it is theoretically possible that immune responses against other cTCR epitopes (eg, junctional regions) could be missed with this assay, the Leu-16 murine domain is expected to be the dominant immunogenic cTCR epitope because all the other domains are of human derivation. The absence of anti-cTCR Ab in patient sera at serial time points was confirmed by flow cytometry (Figure S2). No humoral or cellular immune responses were observed with these assays in any of the 7 patients who received infusions of genetically modified T cells. Two patients, however, were subsequently found to be seropositive for HAMA, 3 and 12 months after T-cell infusions, respectively. One of those patients had been previously treated with a radioiodinated murine monoclonal Ab (tositumomab).

Effect of adoptive T-cell therapy on native lymphocyte subsets

Anti-CD20 Ab therapy substantially depletes normal CD20+ B cells,31  and adoptive anti-CD20 T-cell therapy was therefore also expected to reduce circulating B-cell numbers. We found, however, that, although B-cell counts as measured by flow cytometry fluctuated during the period of T-cell infusions, the number of CD20+ B cells remained stable or slightly increased in the months after treatment in all patients (Table S2). Other lymphocyte subsets (CD3+, CD4+, CD8+, CD56+, and FoxP3+ cells) showed similar vacillations before and after T-cell infusions, but no consistent trends were noted (Table S3).

Responses to T-cell therapy

Two of the 7 patients achieved a complete response to cytoreductive chemotherapy administered before the T-cell infusions and remained disease-free 3 months and 13 months after T-cell infusions (Table 5). Another patient attained an objective partial response lasting 3 months after treatment with T-cell infusions plus IL-2. Four patients exhibited stable disease for 3, 5, 6, and 12 months. One of these patients showed decreased uptake of fluorodeoxyglucose, a marker for viable tumor metabolism, on positron emission tomographic scanning 3 months after T-cell infusions.

Table 5

Clinical responses

Patient*Response to cytoreductive chemotherapyResponse to T-cell InfusionsDuration of response after T-cell infusions, mo
CR NED 13 
PR SD 
SD SD 12 
CR NED 
Did not receive chemotherapy SD (with FDG response on PET scan) 
PR PR§ 
PR SD 
Patient*Response to cytoreductive chemotherapyResponse to T-cell InfusionsDuration of response after T-cell infusions, mo
CR NED 13 
PR SD 
SD SD 12 
CR NED 
Did not receive chemotherapy SD (with FDG response on PET scan) 
PR PR§ 
PR SD 

CR indicates complete response; NED, no evidence of disease; PR, partial response; SD, stable disease; FDG, fluorodeoxyglucose; and PET, positron emission tomography.

*

Patients C and E are omitted because they did not receive T-cell infusions.

Response assessment was performed compared with the restaging imaging after cytoreductive chemotherapy.

All patients progressed after the times listed.

§

Patient H achieved an additional objective PR after T-cell infusions.

MCL and indolent NHL almost invariably relapse after standard therapy despite complete clinical remissions, suggesting the existence of a minimal residual disease state in which a small number of cells resistant to chemotherapy, radiation, or Ab therapy persist and eventually lead to recurrent lymphoma. These cells appear to be susceptible to cellular immune responses, however, because patients undergoing nonmyeloablative allogeneic SCT or DLI can achieve long-term disease-free remissions, suggesting cure.11  However, the high morbidity and mortality rates associated with these treatments,9,32  primarily resulting from graft-versus-host disease, indicate a need for less-toxic forms of cellular therapy. One such strategy is infusion of ex vivo–expanded autologous T cells modified to recognize lymphoma-associated surface antigens.

We initiated a proof-of-concept clinical trial to test the safety and feasibility of this approach and found that autologous PBMCs from patients with lymphoma could be reliably modified to express a CD20-specific cTCR, expanded ex vivo, and reinfused with minimal toxicities. The process of generating therapeutic numbers of genetically modified anti-CD20 T cells by limiting dilution proved laborious, however, and the target cell dose was reached in only 1 of 5 patients. In contrast, expansion of transfected cells produced in bulk cultures was reliable, more efficient, and may also facilitate longer in vivo persistence in patients receiving such T cells requiring a shorter ex vivo expansion period. All patients tolerated the T-cell infusions well, with no adverse events attributable to T cells, and only grade 1 to 2 toxicities associated with IL-2 injections.

Adoptively transferred T cells presumably must survive in vivo for many weeks to effect a complete clinical response, and establishing memory cell responses may be required to produce durable remissions. We found that the administration of low-dose IL-2 after T-cell infusions prolonged in vivo persistence of modified T cells, consistent with previous observations.33  Immunophenotypic differences between T cells produced by limiting dilution and those produced in bulk cultures were minimal, suggesting this was probably not the dominant reason for longer survival of the T cells from bulk cultures, although the shorter culture time may have contributed by decreasing replicative senescence. These questions cannot be directly assessed from our data because the change in culture methods was coincident with initiation of adjuvant IL-2 treatment. The administration of lymphodepleting chemotherapy before adoptive T-cell transfer is another factor potentially affecting in vivo persistence by enhancement of homeostatic proliferation by IL-7 and IL-15 up-regulation,34,35  depletion of Treg cells,36  and suppression of host immune responses to modified cells. Recent T-cell trials for melanoma incorporating aggressive lymphodepletion regimens have shown long periods of T-cell survival in some patients.37,38  In the present trial, few compelling conclusions can be drawn as to the potential contribution of lymphodepletion to T-cell persistence because of the limited number of patients, the variability in chemotherapy regimens used, and the required 4-week interval between chemotherapy and T-cell infusions.

Recent data by Berger et al39  indicate that CD8+ T cells derived from central memory cells (TCM) persist much longer in vivo than those derived from effector memory cells, raising the question of whether the longer persistence of T cells produced in bulk cultures in this study could be due to the presence of TCM-derived cells among the infused T cells. Although this is theoretically possible, the low transfection efficiency by electroporation (0.002%-0.004%)24  and the small number of clones infused make it unlikely that any infused cells were TCM derived. It is much more likely that the difference in persistence in our study was due primarily to the IL-2 injections. Nevertheless, it may be important in future clinical trials to select, modify, and expand CD62L+ cells to establish a population of persisting TCM.

The expansion of genetically modified T cells produced by limiting dilution confers the theoretical advantage of permitting selection of cells with an in vitro cytotoxicity that exceeds the mean of a population of unselected cells in bulk culture. We observed, however, that the highest lytic activity and best proliferative capacity usually did not coincide in the same clonal population, and, consequently, expansion of the most cytolytic T cells was problematic. We believe, therefore, that the more favorable growth kinetics and apparently longer in vivo persistence of T cells produced in bulk cultures outweighs any minor differences in cytotoxicity. T cells from all patients showed CD20-specific cytotoxicity despite the low levels of cTCR they expressed.

Efficacy was not a primary end point of this proof-of-concept study, but clearly that is the long-term goal. Objective clinical responses were modest, with one unequivocal partial remission among the 5 patients with evaluable disease at the time of T-cell infusions. There are many possible explanations for the limited therapeutic antitumor activity in this trial, including insufficient numbers of surviving modified T cells, CD20 antigen competition from native B cells, ineffective localization of T cells to tumor sites, inadequate cTCR surface expression leading to poor killing, and lack of costimulatory signaling from the cTCR construct. The suboptimal in vitro cytotoxicity of these cells suggests that the latter 2 factors were probably at least partially responsible. We plan to address some of these pitfalls in a subsequent trial by using T cells engineered with a “second-generation” anti-CD20 cTCR that includes CD28 and CD137 costimulatory domains and an SP163 translational enhancer. T cells transfected with this second-generation cTCR have shown superior proliferation, surface expression, and cytotoxicity in preclinical studies.25 

Another possible explanation for the modest responses is competitive inhibition of cTCR-antigen binding by residual anti-CD20 Ab present in the serum of patients previously treated with rituximab, ibritumomab, or tositumomab. However, none of the treated patients (with the exception of patient I, who received 131I-tositumomab 4 months before T-cell infusions) had been exposed to anti-CD20 Ab within 1 year of T-cell infusions. Therefore, with the exception of this patient, it is unlikely that significant circulating levels of anti-CD20 Ab were present at the time of adoptive T-cell transfer. It is somewhat surprising that significant changes in circulating B-cell levels were not seen in this study, in view of the well-documented B-cell depletion associated with rituximab treatment.31,40  We do not know whether this reflects a resistance of normal resting B cells in vivo to the cytolytic activity of the cTCR+ effector cells or a demonstration that adoptive T-cell immunotherapy with the “first-generation” plasmid as implemented in this trial has insufficient in vivo activity. This issue may be more clearly resolved after testing T cells expressing cTCR with the augmented second-generation plasmid described earlier.

In summary, adoptive immunotherapy with anti-CD20 cTCR-bearing T cells is a safe, feasible, and well-tolerated treatment for patients with relapsed or refractory indolent NHL, although clinical responses were modest. Expansion of transfected T cells produced in bulk cultures proved to be more efficient for achieving therapeutic numbers than generating T cells by limiting dilution. Furthermore, in vivo persistence of T cells was longer in patients receiving T cells produced by bulk cultures and low-dose IL-2 than in patients receiving T cells produced by limiting dilution and administered without subcutaneous IL-2. These results are encouraging but identify areas for improvement to be addressed in future adoptive T-cell therapy trials.

An Inside Blood analysis of this article appears at the front of this issue.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The plasmid used in these studies was produced by the National Gene Vector Laboratory at the City of Hope, Duarte, CA.

This work was supported by National Institutes of Health (NIH) grants R01 CA92302 and R21 CA-117131-01A (O.W.P.), Lymphoma Research Foundation grant MCLI-07-012 (O.W.P.) and Career Development Award (B.G.T.), American Society of Clinical Oncology Young Investigator Award (B.G.T.), and by gifts from David and Patricia Giuliani, Geary and Mary Britton-Simmons, the Hext Family Foundation, and the Edson Foundation. A portion of this work was conducted through the Clinical Research Center Facility at the University of Washington and supported by NIH grant M01-RR-00 037.

National Institutes of Health

Contribution: B.G.T. enrolled patients, collected and analyzed data, and wrote the paper; M.C.J. designed the plasmid used to modify the T cells and developed the treatment protocol; J.W. performed PCR, cytotoxicity, and immune response assays; E.Y.C. enrolled patients and collected and analyzed data; B.L.W. and H.A.G. performed flow cytometry and TCR spectratyping experiments and contributed to writing the paper; X.Q. performed flow cytometry and humoral immune response assays; S.E.J. analyzed data and contributed to the design of experiments; A.R. and S.J.F. contributed to trial design; A.K.G. and J.M.P. enrolled patients and contributed to the interpretation of data; C.G.L. supervised T-cell transfection and expansion; P.D.G. and S.R.R. contributed to the design of the trial and experiments, provided the TM-LCL cell line, and analyzed data; and O.W.P. contributed to the conception, design, analysis, and interpretation of the clinical trial and experiments, wrote the protocol, and revised the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Brian Till, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-190, Seattle, WA 98109; e-mail: tillb@fhcrc.org.

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
, vol. 
16
 (pg. 
2825
-
2833
)
2
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
, vol. 
18
 (pg. 
317
-
324
)
3
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
, vol. 
17
 (pg. 
268
-
276
)
4
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
, vol. 
104
 (pg. 
3064
-
3071
)
5
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
, vol. 
20
 (pg. 
1288
-
1294
)
6
Kaminski
 
MS
Zasadny
 
KR
Francis
 
IR
, et al. 
Iodine-131-anti-B1 radioimmunotherapy for B-cell lymphoma.
J Clin Oncol
1996
, vol. 
14
 (pg. 
1974
-
1981
)
7
Gopal
 
AK
Rajendran
 
JG
Petersdorf
 
SH
, et al. 
High-dose chemo-radioimmunotherapy with autologous stem cell support for relapsed mantle cell lymphoma.
Blood
2002
, vol. 
99
 (pg. 
3158
-
3162
)
8
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
, vol. 
17
 (pg. 
3793
-
3803
)
9
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
, vol. 
26
 (pg. 
211
-
7
)
10
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
, vol. 
111
 (pg. 
5530
-
5536
)
11
Khouri
 
IF
Lee
 
MS
Saliba
 
RM
, et al. 
Nonablative allogeneic stem-cell transplantation for advanced/recurrent mantle-cell lymphoma.
J Clin Oncol
2003
, vol. 
21
 (pg. 
4407
-
4412
)
12
Porter
 
DL
Antin
 
JH
Donor leukocyte infusions in myeloid malignancies: new strategies.
Best Pract Res Clin Haematol
2006
, vol. 
19
 (pg. 
737
-
755
)
13
Bortin
 
MM
Truitt
 
RL
Rimm
 
AA
Bach
 
FH
Graft-versus-leukaemia reactivity induced by alloimmunisation without augmentation of graft-versus-host reactivity.
Nature
1979
, vol. 
281
 (pg. 
490
-
491
)
14
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
, vol. 
9
 (pg. 
279
-
286
)
15
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
, vol. 
169
 (pg. 
5780
-
5786
)
16
Kershaw
 
MH
Jackson
 
JT
Haynes
 
NM
, et al. 
Gene-engineered T cells as a superior adjuvant therapy for metastatic cancer.
J Immunol
2004
, vol. 
173
 (pg. 
2143
-
2150
)
17
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
, vol. 
15
 (pg. 
825
-
833
)
18
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
, vol. 
5
 (pg. 
131
-
138
)
19
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
, vol. 
4
 (pg. 
75
-
83
)
20
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
, vol. 
9
 (pg. 
577
-
586
)
21
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
, vol. 
83
 (pg. 
435
-
445
)
22
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
, vol. 
22
 
suppl 14
pg. 
2542
 
23
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
, vol. 
17
 (pg. 
2454
-
2460
)
24
Jensen
 
MC
Clarke
 
P
Tan
 
G
, et al. 
Human T lymphocyte genetic modification with naked DNA.
Mol Ther
2000
, vol. 
1
 (pg. 
49
-
55
)
25
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
, vol. 
18
 (pg. 
712
-
725
)
26
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
, vol. 
113
 (pg. 
838
-
850
)
27
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
, vol. 
2
 (pg. 
216
-
223
)
28
Klebanoff
 
CA
Gattinoni
 
L
Restifo
 
NP
CD8+ T-cell memory in tumor immunology and immunotherapy.
Immunol Rev
2006
, vol. 
211
 (pg. 
214
-
224
)
29
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
, vol. 
401
 (pg. 
708
-
712
)
30
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
, vol. 
107
 (pg. 
2294
-
2302
)
31
Igarashi
 
T
Ohtsu
 
T
Fujii
 
H
, et al. 
Re-treatment of relapsed indolent B-cell lymphoma with rituximab.
Int J Hematol
2001
, vol. 
73
 (pg. 
213
-
221
)
32
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
, vol. 
104
 (pg. 
3535
-
3542
)
33
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
, vol. 
99
 (pg. 
16168
-
16173
)
34
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
, vol. 
195
 (pg. 
1523
-
1532
)
35
Surh
 
CD
Boyman
 
O
Purton
 
JF
Sprent
 
J
Homeostasis of memory T cells.
Immunol Rev
2006
, vol. 
211
 (pg. 
154
-
163
)
36
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
, vol. 
174
 (pg. 
2591
-
2601
)
37
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
, vol. 
23
 (pg. 
2346
-
2357
)
38
Morgan
 
RA
Dudley
 
ME
Wunderlich
 
JR
, et al. 
Cancer regression in patients after transfer of genetically engineered lymphocytes.
Science
2006
, vol. 
314
 (pg. 
126
-
129
)
39
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
, vol. 
118
 (pg. 
294
-
305
)
40
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
, vol. 
88
 pg. 
90a
  
Abstract 349
Sign in via your Institution