throbber
Downloaded from http://ashpublications.org/blood/article-pdf/112/6/2261/1482883/zh801808002261.pdf by guest on 31 May 2022
`
`CLINICAL TRIALS AND OBSERVATIONS
`
`Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell
`lymphoma using genetically modified autologous CD20-specific T cells
`Brian G. Till,1,2 Michael C. Jensen3, Jinjuan Wang,1 Eric Y. Chen,1 Brent L. Wood,4 Harvey A. Greisman,4 Xiaojun Qian,1
`Scott E. James,1 Andrew Raubitschek,5 Stephen J. Forman,6 Ajay K. Gopal,1,2 John M. Pagel,1,2 Catherine G. Lindgren,2
`Philip D. Greenberg,1,2 Stanley R. Riddell,1,2 and Oliver W. Press1,2
`
`1Clinical Research Division of the Fred Hutchinson Cancer Research Center, Seattle, WA; 2Department of Medicine, University of Washington, Seattle;
`3Department of Pediatric Hematology-Oncology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA; 4Department of Laboratory
`Medicine, University of Washington, Seattle; and 5Department of Radioimmunotherapy and 6Division of Hematology and HCT, City of Hope National Medical
`Center and Beckman Research Institute, Duarte, CA
`
`Adoptive immunotherapy with T cells ex-
`pressing a tumor-specific chimeric T-cell
`receptor is a promising approach to can-
`cer 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 indo-
`lent B-cell lymphoma or mantle cell lym-
`phoma were treated with autologous
`T cells genetically modified by electropo-
`ration with a vector plasmid encoding a
`Introduction
`
`CD20-specific chimeric T-cell receptor and
`neomycin resistance gene. Transfected
`cells were immunophenotypically similar
`to CD8ⴙ effector cells and showed CD20-
`specific cytotoxicity in vitro. Seven pa-
`tients received a total of 20 T-cell infu-
`sions, 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 re-
`sponse, 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.
`(Blood. 2008;112:
`2261-2271)
`
`Several lymphoma subtypes are incurable with standard chemo-
`therapy 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 conju-
`gated 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 activa-
`tion, proliferation, and killing. Because this antigen-induced activa-
`tion 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.
`
`Submitted December 14, 2007; accepted May 3, 2008. Prepublished online as
`Blood First Edition paper, May 28, 2008; DOI 10.1182/ blood-2007-12-128843.
`
`An Inside Blood analysis of this article appears at the front of this issue.
`
`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 online version of this article contains a data supplement.
`
`© 2008 by The American Society of Hematology
`
`BLOOD, 15 SEPTEMBER 2008 䡠 VOLUME 112, NUMBER 6
`
`2261
`
`UPenn Ex. 2011
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/112/6/2261/1482883/zh801808002261.pdf by guest on 31 May 2022
`
`2262
`
`TILL et al
`
`Methods
`
`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 Declara-
`tion 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
`
`BLOOD, 15 SEPTEMBER 2008 䡠 VOLUME 112, NUMBER 6
`
`Generation and expansion of genetically modified T cells. Trans-
`fected 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 V␤ 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, TCR␣/␤⫹ 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 (TCR␥) locus
`as previously described,26 except that V␥I-J␥1/2, V␥II-J␥1/2, V␥I-J␥P1/2,
`and V␥II-J␥P1/2 rearrangements were amplified in a single multiplex PCR
`reaction and analyzed by capillary electrophoresis on an Applied Biosys-
`tems 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.
`V␤ 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 V␤ 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
`
`lysates of modified T cells were probed with a mouse
`Whole cell
`anti–human CD3␨ monoclonal Ab (BD PharMingen, San Diego, CA) as
`previously described.20
`
`T-cell transfection, selection, and expansion
`
`Cytotoxicity assays
`
`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 ␮g/mL) encoding a
`CD20-specific scFvFc:␨ cTCR,18-20,24 and divided into aliquots into chilled
`0.2-cm electroporation cuvettes. Cells were electroporated with an Eppen-
`dorf Multiporator at 250 V for 40 microseconds (␮sec) 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.
`
`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
`
`UPenn Ex. 2011
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 15 SEPTEMBER 2008 䡠 VOLUME 112, NUMBER 6
`
`ANTI-CD20 T-CELL THERAPY FOR NHL
`
`2263
`
`Downloaded from http://ashpublications.org/blood/article-pdf/112/6/2261/1482883/zh801808002261.pdf by guest on 31 May 2022
`
`Figure 1. Schema of clinical protocol.
`
`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
`incremental doses (108 cells/m2, 109 cells/m2, and
`apart, at
`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.
`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).
`
`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 CD3␨ intracellular signaling do-
`main, 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).
`
`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:␨ plasmid DNA starting at 106 copies/␮L, with each
`sample containing 1 ␮g 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 CD3␨ chain and adjacent CD4
`transmembrane domain sequences was amplified using forward primer 5⬘-
`TCGCCGGCCTCCTGCTTT-3⬘ and reverse primer 5⬘-CGTCTGCGCTCCT-
`GCTGA-3⬘. The probe used was 5⬘-FAM-TGGGCTAGGCATCTTCTTCA-
`GAGTGAA-TAMRA-3⬘. Primers that amplify a fragment of the ␤-actin gene
`(TaqMan 〉-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 ␮g 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 ␮g 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 ␮g/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 ␮L/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
`the PBMCs were tested in 51Cr release
`stimulation 1 week apart,
`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.
`
`Results
`
`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
`
`UPenn Ex. 2011
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/112/6/2261/1482883/zh801808002261.pdf by guest on 31 May 2022
`
`2264
`
`TILL et al
`
`Table 1. Patient characteristics
`
`BLOOD, 15 SEPTEMBER 2008 䡠 VOLUME 112, NUMBER 6
`
`Patient
`
`Age, y
`
`Sex
`
`Diagnosis
`
`Stage
`
`Prior therapies
`
`Cytoreductive therapy before
`T-cell infusions
`
`A
`B
`C
`D
`E
`F
`G
`H
`I
`
`44
`70
`47
`60
`63
`46
`43
`46
`77
`
`F
`M
`M
`M
`M
`M
`M
`M
`M
`
`FL
`FL
`FL
`FL
`MCL
`FL
`FL
`FL
`FL
`
`IV-B
`II-A
`IV-B
`IV-A
`IV-A
`IV-A
`IV-A
`IV-B
`III-A
`
`R-CHOP
`CHOP, rituximab, 131I-tositumomab
`ProMACE/MOPP, ASCT, fludarabine (10 cycles)
`Rituximab
`R-HyperCVAD, GCD-R
`R-CVP
`CHOP, IFN, CY ⫹ VP16, R-CY, CY ⫹ DEX, GCD-R, ASCT
`R-CHOP, fenretinide
`R-CVP, R-CHOP, GCD-R
`
`CVP
`CVP
`CVP
`CVP
`None
`FND
`None
`FND
`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-␣; CY, cyclophosphamide; VP16, etoposide; and DEX, dexamethasone.
`
`Modified T cells were generated for the first 5 patients by
`limiting dilution and selected for CD20 cytotoxicity by chro-
`mium 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 V␤ TCR
`spectratyping and TCR␥ 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).
`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). V␤
`TCR spectratyping and TCR␥ 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.
`
`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 infu-
`sions 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 effec-
`tor memory (CD62L⫺/CCR7⫺/CD45RA⫺/CD127⫹) phenotypes.
`
`B
`
`Patient B
`
`Negative Positive
`
`Chimeric Zeta Chain
`
`Ribosome
`Binding
`Sequence
`
`Leu 16
`scFv
`
`Transmembrane
`domain
`
`SV40
`promoter
`
`NeoR
`
`CD3ζ
`
`CH2-CH3
`Hinge
`
`CD4
`TM
`
`VH
`
`Linker
`
`VL
`
`Murine κ
`Signal
`Peptide
`
`Human
`IgG1 Fc
`
`Endogenous Zeta Chain
`
`A
`
`I/Epromoter
`
`CMV
`
`Figure 2. Expression of the CD20-specific cTCR. (A) Schematic diagram of the CD20-specific scFvFc:␨ 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 CD3␨ monoclonal Ab. Negative
`control was parental PBMCs, and positive control was transfected Jurkat cell line. A 21-kDa band corresponding to the endogenous CD3␨ 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.
`
`UPenn Ex. 2011
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/112/6/2261/1482883/zh801808002261.pdf by guest on 31 May 2022
`
`BLOOD, 15 SEPTEMBER 2008 䡠 VOLUME 112, NUMBER 6
`
`ANTI-CD20 T-CELL THERAPY FOR NHL
`
`2265
`
`VB2
`
`V B3
`
`V B5.1
`
`V B4
`
`V B7.1
`VB5.2V B5.3
`V B7.2
`
`V B8
`
`VB9
`
`V B11
`
`V B12
`VB13.6
`V B13.1
`VB13.2
`
`V B14
`
`V B16
`
`V B17
`
`VB18
`
`V B20
`V B21.3
`
`V B22
`
`VB23
`
`TCR VBETA FAMILY
`
`B
`
`100
`
`90
`
`80
`
`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`V B1
`
`% VBETA USAGE
`
`V B2
`
`V B3
`
`V B5.1
`
`V B4
`
`V B7.2
`V B7.1
`V B5.2V B5.3
`
`V B8
`
`V B9
`
`V B11
`
`V B12
`V B13.6
`V B13.2
`V B13.1
`
`V B14
`
`V B16
`
`V B17
`
`V B18
`
`V B20
`V B21.3
`
`V B22
`
`V B23
`
`TCR VBETA FAMILY
`
`A
`
`100
`
`90
`
`80
`
`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`V B1
`
`% VBETA USAGE
`
`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) V␤ spectratyping
`(top) and by PCR amplification of clonal V-J rearrangements at the TCR␥ 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 V␤17 in 98% of CD8⫹ T cells by V␤ spectratyping (top; f) and showing
`2 predominant TCR␥ rearrangements (bottom). Because each T-cell clone can rearrange one or both of its TCR␥ alleles, the 2 PCR products could represent either 1 T-cell
`clone with biallelic TCR␥ rearrangements or 2 singly rearranged clones, although the single predominant V␤17 clone identified by spectratyping would favor a single doubly
`rearranged clone. (B) T cells produced in bulk culture (patient G) showing oligoclonal V␤ expression in CD8⫹ T cells (16% V␤16; 9% V␤7.1; 3% each V␤3, V␤13.2 and V␤17;
`2% each V␤1 and V␤13.1; and 1% each V␤5.1, V␤13.6, V␤21.3, and V␤23) and 7 distinct TCR␥ rearrangements by PCR (bottom; f) 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 䡺 in both top panels represent the average expression levels for
`each V␤ chain in normal polyclonal T-cell populations.
`
`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 functional-
`ity 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%).
`
`Cytotoxicity of modified T cells in vitro
`
`Our group showed in preclinical studies that T-cell clones
`bearing CD20-specific cTCRs exhibit antigen-specific cytotox-
`icity.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).
`
`Table 2. T-cell infusions
`Infusion 1,
`cells/m2*
`
`Patient
`
`A
`B
`C
`D
`E
`F
`G
`H
`I
`
`108
`108
`
`108
`
`108
`108
`108
`108
`
`Infusion 2,
`cells/m2†
`
`109
`109
`
`4 ⫻ 108
`
`109
`109
`109
`109
`
`Infusion 3,
`cells/m2‡
`
`3.3 ⫻ 109
`2 ⫻ 109
`
`3.3 ⫻ 109
`3.3 ⫻ 109
`3.3 ⫻ 109
`2 ⫻ 109㛳
`
`Fresh versus
`thawed cells
`
`Time from apheresis
`to target cell number, d
`
`No. of stimulation
`cycles§
`
`Fresh
`Fresh
`
`Thawed
`
`Thawed
`Thawed
`Thawed
`Thawed
`
`130
`129⫹
`Expansion failed
`159⫹
`Expansion failed
`96
`90
`81
`104
`
`7
`7
`5
`7
`5
`6
`5
`5
`8
`
`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).
`㛳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.
`
`UPenn Ex. 2011
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/112/6/2261/1482883/zh801808002261.pdf by guest on 31 May 2022
`
`2266
`
`TILL et al
`
`BLOOD, 15 SEPTEMBER 2008 䡠 VOLUME 112, NUMBER 6
`
`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).
`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 immunosup-
`pression 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 seri-
`ally collected patient PBMCs that had been coincubated with
`irradiated T cells expressing the scFvFc:␨ 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
`
`Figure 4. Growth curves of gen

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket