throbber
Antitransgene Rejection Responses Contribute to
`Attenuated Persistence of Adoptively Transferred
`CD20/CD19-Specific Chimeric Antigen Receptor
`Redirected T Cells in Humans
`
`Michael C. Jensen,1,2 Leslie Popplewell,2 Laurence J. Cooper,2 David DiGiusto,2
`Michael Kalos,1 Julie R. Ostberg,1 Stephen J. Forman1,2
`
`Immunotherapeutic ablation of lymphoma is a conceptually attractive treatment strategy that is the subject of
`intense translational research. Cytotoxic T lymphocytes (CTLs) that are genetically modified to express
`CD19- or CD20-specific, single-chain antibody–derived chimeric antigen receptors (CARs) display HLA-
`independent antigen-specific recognition/killing of lymphoma targets. Here, we describe our initial experi-
`ence in applying CAR-redirected autologous CTL adoptive therapy to patients with recurrent lymphoma.
`Using plasmid vector electrotransfer/drug selection systems, cloned and polyclonal CAR1 CTLs were gen-
`erated from autologous peripheral blood mononuclear cells and expanded in vitro to cell numbers sufficient
`for clinical use. In 2 FDA-authorized trials, patients with recurrent diffuse large cell lymphoma were treated
`with cloned CD81 CTLs expressing a CD20-specific CAR (along with NeoR) after autologous hematopoi-
`etic stem cell transplantation, and patients with refractory follicular lymphoma were treated with polyclonal
`T cell preparations expressing a CD19-specific CAR (along with HyTK, a fusion of hygromycin resistance and
`HSV-1 thymidine kinase suicide genes) and low-dose s.c. recombinant human interleukin-2. A total of 15 in-
`fusions were administered (5 at 108cells/m2, 7 at 109cells/m2, and 3 at 2  109cells/m2) to 4 patients. Overt
`toxicities attributable to CTL administration were not observed; however, detection of transferred CTLs in
`the circulation, as measured by quantitative polymerase chain reaction, was short (24 hours to 7 days), and
`cellular antitransgene immune rejection responses were noted in 2 patients. These studies reveal the primary
`barrier to therapeutic efficacy is limited persistence, and provide the rationale to prospectively define T cell
`populations intrinsically programmed for survival after adoptive transfer and to modulate the immune status
`of recipients to prevent/delay antitransgene rejection responses.
`Biol Blood Marrow Transplant 16: 1245-1256 (2010) Ó 2010 American Society for Blood and Marrow Transplantation
`
`KEY WORDS: Cellular immunotherapy, Adoptive therapy, T lymphocyte, Clinical trial
`
`From the 1Department of Cancer Immunotherapeutics and Tumor
`Immunology; and 2Department of Hematology and Hemato-
`poietic Cell Transplant, Beckman Research Institute, City of
`Hope National Medical Center, Duarte, California.
`Laurence J. Cooper is currently at the Division of Pediatrics, M.D.
`Anderson Cancer Center, Houston, Texas. Michael Kalos is
`currently at the Abramson Family Cancer Research Institute,
`University of Pennsylvania School of Medicine, Philadelphia,
`Pennsylvania.
`Financial disclosure: See Acknowledgments, page 1256.
`Correspondence and reprint requests: Michael C. Jensen, MD,
`Department of Cancer Immunotherapeutics and Tumor Immu-
`nology, Beckman Research Institute, City of Hope National
`Medical Center, 1500 East Duarte Road, Duarte, CA 91010-
`3000 (e-mail: mjensen@coh.org).
`Received January 26, 2010; accepted March 11, 2010
`Ó 2010 American Society for Blood and Marrow Transplantation
`1083-8791/$36.00
`doi:10.1016/j.bbmt.2010.03.014
`
`INTRODUCTION
`
`Although conventional chemotherapy, radiation
`therapy, and antibody therapy can be efficacious in
`treating lymphoma, relapse and progressive disease
`are the major sources of patient morbidity and mortal-
`ity [1,2]. Experimental evidence that
`the cellular
`immune system can eradicate lymphoma provides
`a basis for the development of therapies aimed at
`amplifying antitumor immune responses [3,4]. The
`adoptive transfer of lymphoma-specific T cells is one
`strategy to augment antilymphoma immunity. A sig-
`nificant challenge to executing this strategy is the iso-
`lation of T cells specifically reactive to lymphoma.
`Alternately, the ex vivo derivation of tumor-specific
`T lymphocytes by genetic modification to express
`tumor-targeting chimeric antigen receptors (CARs)
`is a rapidly evolving focus of translational cancer
`
`1245
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`1246
`
`M. C. Jensen et al.
`
`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`immunotherapy [5,6]. Antibody-based CARs are
`HLA-unrestricted and thus can be used in patient pop-
`ulations with target-antigen–positive tumors.
`We have constructed 2 CARs specific for the B cell
`lineage antigens CD20 and CD19 for the purpose of
`targeting lymphomas and leukemias [7,8]. When
`expressed in cytotoxic T lymphocytes (CTLs), these
`CARs redirect effector cells to lyse B-lineage lymph-
`oma targets [7,8]. Here we report our initial clinical
`experience in manufacturing and infusing autologous
`T cells expressing CD20R or CD19R in patients with
`relapsed B cell lymphoma under City of Hope–held
`FDA-authorized trials BB-IND-8513/IRB 98142 and
`BB-IND-11411/IRB 01160, respectively.
`
`MATERIALS AND METHODS
`
`Patients
`
`City of Hope Internal Review Board (IRB) proto-
`cols 98142 and 01160 were activated for patient accrual
`following IRB and Institutional Biological Safety
`Committee approval, FDA authorization (BB-IND-
`8513 and BB-IND-11411, respectively), and National
`Institutes of Health Office of Biotechnology Activities
`registration (9907-330 and 0207-543, respectively). In
`brief, for IRB 98142, patients were eligible if they had
`immunohistopathologically documented CD201 dif-
`fuse large cell lymphoma (DLCL) with a history of
`recurrent or refractory disease and did not have central
`nervous system metastases. After leukapheresis, pa-
`tients began salvage/mobilization chemotherapy,
`then underwent hematopoietic stem cell transplanta-
`tion (HSCT). The first of 3 escalating-dose T cell infu-
`sions was given at 28 days post-HSCT. For IRB
`protocol 01160, patients were eligible if they had path-
`ologically documented follicular lymphoma (FL) with
`evidence of progression after previous rituximab ther-
`apy and did not have central nervous system metastases
`or a history of allogeneic HSCT. These patients were
`enrolled no sooner than 3 weeks after their most recent
`cytotoxic chemotherapy.
`
`Plasmid Vectors
`
`The plasmid expression vectors encoding the
`CD20R chimeric immunoreceptor and the neomycin
`phosphotransferase cDNAs and the CD19R chimeric
`immunoreceptor and the selection-suicide HyTK (a
`fusion of hygromycin resistance and HSV-1 thymidine
`kinase suicide genes) cDNAs have been described
`previously [7,8] (Figure S1A). In brief, the chimeric
`construct consists of VH and VL gene segments of
`the CD20-specific Leu-16 or CD19-specific FMC63
`monoclonal antibodies (mAbs), an IgG hinge-CH2-
`CH3 region, a CD4 transmembrane region, and the
`cytoplasmic domain of the CD3z chain (Figure S1B).
`
`Isolation, Transfection, Selection, Cloning, and
`Expansion of T Cells
`
`The methods for OKT3 stimulation of peripheral
`blood mononuclear cells (PBMCs), and for PBMC
`electroporation, selection, cloning (IRB 98142 only),
`and subsequent growth using the rapid expansion
`method (REM), consisting of recursive 14-day cycles
`of activation with OKT3, recombinant human inter-
`leukin (rHuIL)-2, and PBMC/lymphoblastoid cell
`line (LCL)-irradiated feeders, have been described
`previously [9]. The overall T cell product manufactur-
`ing schemas for each trial are depicted in Figure S1C.
`
`Cell Product Quality Control Tests
`
`The cell product quality control tests (QCTs) per-
`formed and the requisite test results for product release
`are summarized in Table S1.
`
`Confirmation of Plasmid Vector Integration (IRB
`98142 Only)
`
`A single site of plasmid vector chromosomal inte-
`gration was confirmed by Southern blot analysis of
`XbaI/HindIII-digested T cell genomic DNA using
`a 420-bp NeoR-specific probe generated using the
`pcDNA3.1(-) plasmid as a template [9]. The pass crite-
`rion of this test was defined as detection of a single
`band.
`
`Confirmation of CAR Expression
`
`Western blot analysis for CAR expression has been
`described previously [10]. In brief, reduced whole-cell
`lysates are subjected to Western blot analysis with an
`anti-human CD3-z (cytoplasmic tail)-specific mAb
`8D3 (BD Pharmingen, San Diego, CA). This probe
`detects both the 16-kDa endogenous z and the 66-
`kDa CAR z. Pass criteria were defined as visualization
`of both the 16-kDa and 66-kDa bands. Flow cytometry
`analysis for surface CAR expression was determined
`using a fluorescein isothiocyanate (FITC)-conjugated
`Fc-specific
`antibody
`(Jackson ImmunoResearch,
`West Grove, PA). Pass criteria were defined as unim-
`odal positive staining for Fc compared with the
`FITC-conjugated isotype control (BD Biosciences,
`San Jose, CA).
`
`Surface Phenotype Determination
`
`T cell products were evaluated for cell-surface
`phenotype using standard staining and flow cytometric
`procedures with FITC-conjugated mAbs (BD Biosci-
`ences), followed by analysis on a FACScaliber analyzer
`(BD Biosciences). The pass criterion was $90% posi-
`tive staining for TCR-ab and CD8 (IRB 98142) or
`CD3 (IRB 01160) compared with the isotype control.
`Independent of the QCT guidelines, other correlative
`surface markers included CD4 for IRB 98142 and both
`CD4 and CD8 for IRB 01160.
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`Adoptive Therapy with Redirected T Cells for Lymphoma
`
`1247
`
`Assay for Antilymphoma Cytolytic Activity
`Cytolytic activity of CAR1 CTLs against 51Cr-
`labeled human lymphoma Daudi cells was assessed as
`described previously using a 4-hour chromium release
`assay [8]. The pass criterion was $50% specific lysis at
`an effector-to-target ratio of 25:1.
`
`Viability
`
`Viability was determined by standard trypan blue
`dye exclusion. The pass criterion was .90% viability.
`
`Sensitivity to Ganciclovir Ablation (IRB 01160
`Only)
`
`To test for acquired cytocidal sensitivity to ganci-
`clovir (GCV), aliquots of cells were harvested from
`5-day REM cultures, then maintained for 14 days in
`37.5 U/mL rHuIL-2 with or without 1 mM GCV.
`Then the cells were harvested and subjected to viability
`testing. The pass criterion was #25% viability in the
`GCV-treated cultures.
`
`Assay for Antigen/IL-2–Independent Growth
`First, 5  106 cells were washed and plated in anti-
`gen- and IL-2–free culture media at the end of a 14-day
`REM cycle. Parallel cultures of Jurkat T cells (Ameri-
`can Type Culture Collection, Manassas, VA) (IRB
`98142) or T cells cultured in the presence of 37.5 U/
`mL rHuIL-2 (IRB 01160) served as controls for expan-
`sion and viability. For IRB 98142, following an 11-day
`incubation, cultures were harvested, counted using try-
`pan blue, plated into 96-well plates at 6000 viable cells
`per well, and pulsed with 1 mCi of 3H-TdR. DNA was
`harvested following a 4-hour incubation at 37C. For
`IRB 01160, viable cell numbers of 14-day cultures
`were determined by flow cytometry as described
`previously [11]. Release criteria specified that cells
`must exhibit \10% of the Jurkat cpm (IRB 98142) or
`\10% of the IL-21 control cell number (IRB 01160).
`
`Sterility
`
`Sterility tests were performed according to an FDA
`Center for Biologics and Evaluation of Research–man-
`dated schedule. Aliquots of media from the T cell cul-
`tures were plated onto bacterial and fungal growth
`media. Mycoplasma detection was conducted on me-
`dia aliquots using the Gen-Probe Mycoplasma Tissue
`Culture-NI Rapid Detection System (Gen-Probe, San
`Diego, CA), and endotoxin levels were determined by
`enzyme-linked immunosorbent assay. Pass criteria
`were negative bacterial,
`fungal, and mycoplasma
`results, along with an endotoxin level\5 EU/kg recip-
`ient weight.
`
`numbers before being resuspended in 0.9% NaCl
`with 2% human serum albumin in a clinical reinfusion
`bag. T cells were reinfused i.v. over 30 minutes
`through either a central line or an age-appropriate
`sized i.v. catheter inserted into a peripheral vein. The
`infusion bag was mixed gently every 5 minutes during
`the infusion. The intrapatient dose escalation plan is
`shown schematically in Figure S2. In IRB 01160, flu-
`darabine (Flu) was administered after the first T cell
`infusion as a potential nonmyeloablative (NMA)
`immunosuppressive regimen for attenuating possible
`rejection responses against the transferred T cells.
`The guidelines provided in the National Cancer Insti-
`tute’s (NCI) Common Toxicity Criteria, version 2.0
`(https://ctep.ifo.nih.gov/l) were followed for the mon-
`itoring of toxicity and adverse event reporting. Rules
`for dose escalation, de-escalation, and cancellation
`were strictly enforced and resulted in 3 of the 4 treated
`patients deviating from the planned infusion cell dose
`escalation at least once.
`
`In Vivo Persistence of Transferred T Cells
`
`For IRB 98142, PBMCs from heparinized periph-
`eral blood samples were isolated and analyzed for per-
`centage of transfected cells by quantitative polymerase
`chain reaction (qPCR) as described previously [12]
`using primers and probes to quantify CD20R copy
`number (details available on request).
`For IRB 01160, samples were received, processed,
`stored, and analyzed in accordance with current good
`laboratory practice guidelines. A validated qPCR-
`based assay to quantify CD19R plasmid vector DNA
`in samples was developed and performed using an
`MJ Research DNA engine with a Chromo 4 continu-
`ous fluorescence detector qPCR module (Bio-Rad,
`Hercules, CA). Real-time qPCR was performed in
`a 20-mL reaction mixture volume containing 50 ng
`DNA, 10 mL of IQ SYBR Green Supermix (Bio-Rad;
`catalog no. 170-8880), and 0.5 pmol of each primer.
`Quantification of the CD19R transgene sequence in
`DNA isolated from patient PBMCs was evaluated us-
`ing qPCR to amplify a 182-nucleotide fragment that
`spanned the CD4 transmembrane–zeta junction
`within the transgene coding sequence, and a standard
`curve derived by dilution of DNA isolated from a clone
`with a single integration of the CD19R transgene
`(primers, probes, and amplification conditions avail-
`able on request). The qualification studies for this am-
`plification reaction demonstrated no amplification
`from healthy donor-derived PBMCs (n 5 5), whereas
`the transgene sequence could be quantified in
`a PBMC sample if the transgene containing DNA
`composed as little as 0.1% of the total DNA sample.
`
`Adoptive Transfer of T Cells
`
`Analysis of Antitransgene Rejection Responses
`
`Processed and cryopreserved cell banks were
`thawed and expanded in culture to the desired cell
`
`Pretreatment and post-treatment PBMCs (on days
`175, 177, and 150 from UPN006, UPN009, and
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`1248
`
`M. C. Jensen et al.
`
`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`UPN035, respectively) were first stimulated with irra-
`diated therapeutic T cells (3000 rads) or LCLs with
`and without pcDNA3.1(-) plasmid (8000 rads) plus ir-
`radiated pretreatment PBMCs as feeder cells at a 10:1
`responder-to-stimulator ratio in RPMI 1640 (Irvine
`Scientific, Santa Ana, CA) supplemented with 2 mM
`L-glutamine (Irvine Scientific), 25 mM Hepes (Irvine
`Scientific), 100 U/mL penicillin, 0.1 mg/mL strepto-
`mycin (Irvine Scientific), and 10% heat-inactivated
`human serum. One week later, the same irradiated
`stimulator, as well as irradiated pretreatment PBMC
`feeders (3500 rads), were added at a 1:1:1 responder-
`to-stimulator-to-feeder ratio. This stimulation schema
`was repeated up to 2 more times (once weekly) until suf-
`ficient numbers were obtained for chromium-release
`assays. Cytolytic activity of these stimulated PBMC
`against 51Cr-labeled targets was analyzed as described
`previously using a 4-hour chromium-release assay [8].
`For IRB 01160, cellular antitransgene immune re-
`sponses were evaluated directly ex vivo using a combina-
`tion of T cell receptor (TCR) Vb spectratyping and
`CD107 degranulation assays. For the TCR Vb spectra-
`typing analysis, RNA was
`isolated from PBMC
`collected before
`and after
`infusion using the
`RNAqueous-4 PCR Kit for Isolation of DNA-free
`RNA (Applied Biosystems/Ambion, Austin, TX), and
`cDNA was then synthesized using the iScript cDNA
`Synthesis Kit (Bio-Rad). TCR Vb spectratyping analy-
`sis was performed on cDNA samples essentially as de-
`scribed previously [13] using pools of Vb-specific
`primers. A parallel series of amplifications using
`cDNA generated from pooled healthy donor PBMCs
`was performed as a quality control for amplification of
`each Vb family. Aliquots of the amplification mixes
`were run on sequencing gels, followed by analyses using
`Genemapper v3.7 software (Applied Biosystems). The
`CD107 degranulation/mobilization assay was per-
`formed essentially as described previously [14], using
`patient PBMCs collected before and after infusion as ef-
`fectors and infused T cell product or OKT3-expanded
`preinfusion PBMCs (i.e., autologous T cells) as targets.
`To detect spontaneous degranulation, a control sample
`without target cells was included in every experiment.
`FITC-conjugated anti-CD107a
`and anti-CD107b
`(BD Biosciences) were added directly to the tubes be-
`fore incubation. After 5 hours of coincubation, cells
`were washed twice
`and stained with PE-Cy5-
`conjugated anti-CD8b and PE-conjugated anti-TCR
`Vb23 (Beckman Coulter, Fullerton, CA) for 30 minutes
`at room temperature in the dark, rewashed, and ana-
`lyzed on a FC500 flow cytometer using FCS Express
`v3.0 software (Beckman Coulter), with gating on
`CD8b1 and Vb231 lymphocytes.
`
`Serologic Anti-CAR Immune Response Analysis
`
`Serum was isolated from patient blood samples
`collected in red-top (no additive) tubes using an
`
`established laboratory standard operating procedure
`and a qualified assay to detect CAR-specific serologic
`responses in samples. Samples were allowed to clot
`for 2-1/2 hours at room temperature, then centrifuged
`at 1000  g for 15 minutes at 4C. Serum was col-
`lected, aliquoted, and frozen immediately at 280C.
`Flow cytometry detection of potential serum antibody
`responses against
`the anti-CD19R transgene was
`performed using parental versus CD19R-expressing
`Jurkat cell lines as indicator cell lines. The presence
`of antibodies in patient serum that specifically bound
`to CD19R1 Jurkat cells was evaluated by a subsequent
`incubation with FITC-conjugated AffiniPure F(ab’)2
`fragment goat anti-human IgG (Fcg; Jackson Immu-
`noResearch). The cutoff for a negative response was
`established by defining the 95% one-sided prediction
`interval using a pool of non–CAR-reactive serum
`samples from healthy volunteers.
`
`RESULTS
`
`Patient Characteristics
`
`The patients in these studies either had DLCL
`(BB-IND 8513/IRB 98142) or
`follicular non-
`Hodgkin lymphoma (NHL; BB-IND 11411/IRB
`01160) (Table S2). One of the 4 research participants
`had bulky disease, including sites at the neck, chest,
`lymph nodes, and pelvis, at the time of enrollment.
`Three of the 4 participants had received rituximab
`(chimeric mAb specific for CD20) therapy before the
`first infusion of therapeutic T cells. The average dura-
`tion of time from leukapheresis to first infusion was
`106 days, and was affected by the time required to
`manufacture the T cell product and/or the timing of
`the patient’s recovery from salvage therapy.
`
`Generation of Genetically Modified T Cells
`
`Cell products meeting all quality control release
`tests (Table S1) were successfully generated for 2 of
`the 5 patients enrolled on IRB 98142, and for both pa-
`tients enrolled on IRB 01160. The failure to release
`products for 3 of the patients enrolled on IRB 98142
`stemmed from an inability to isolate T cell clones
`that expressed CD8, expressed endogenous TCR, or
`expanded adequately in vitro. The results of Southern
`blot analysis indicating the desired single-site inser-
`tions of the CD20R transgene within the released
`clones of IRB 98142 are depicted in Figure 1. Western
`blot and cell-surface expression profiles of T cell
`products for both trials are also depicted, confirming
`expression of the CAR protein. These cells were fur-
`ther subjected to flow cytometry analysis for confirma-
`tion of the T cell subset markers CD4, CD8, and either
`TCR-ab or CD3. All of the cell products used in ther-
`apy also exhibited redirected killing of CD191 and
`CD201 human Daudi lymphoma targets in 4-hour
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`Adoptive Therapy with Redirected T Cells for Lymphoma
`
`1249
`
`51
`Cr-Release
`
`GCV Sensitivity IL-2 Dependence
`10 6
`
`10 5
`
`10 4
`
`10 3
`
`CPM
`
`10 2
`Jurkat
`
`1A11
`
`Jurkat
`
`6D10
`
`10 5
`
`10 4
`
`10 3
`
`10 2
`
`CPM
`
`10 8
`
`10 7
`10 6
`
`IL2
`
`(-)
`
`IL2
`
`(-)
`
`10 5
`
`10 4
`
`10 9
`10 8
`10 7
`10 6
`10 5
`10 4
`
`Viable Cell #
`
`N.D.
`
`N.D.
`
`10 8
`
`10 7
`
`10 6
`
`IL2
`
`IL2
`+GCV
`
`Viable Cell #
`
`10 5
`
`10 9
`
`10 8
`
`10 7
`
`10 6
`
`IL2
`
`IL2
`+GCV
`
`50:1 25:1 5:1 1:1
`
`50:1 25:1 5:1 1:1
`
`50:1 25:1 5:1 1:1
`
`50:1 25:1 5:1 1:1
`Effector:Target
`
`100
`80
`60
`40
`20
`
`0
`
`100
`80
`60
`40
`20
`0
`
`100
`80
`60
`40
`20
`0
`
`100
`80
`60
`40
`20
`0
`
`% Specific Lysis
`
`Southern Western
`
`Flow Cytometry
`
`66kd
`
`UPN006
`Clone 1A11
`
`UPN009
`Clone 6D10
`
`UPN035
`
`N.D.
`
`UPN037
`
`N.D.
`
`16 kd
`
`anti-Fc
`
`anti-TCR
`
`anti-CD8
`
`anti-CD4
`
`66kd
`
`16 kd
`
`66kd
`
`16 kd
`
`66kd
`
`anti-Fc
`
`anti-TCR
`
`anti-CD8
`
`anti-CD4
`
`anti-Fc
`
`anti-CD3
`
`anti-CD8
`
`anti-CD4
`
`16 kd
`
`anti-Fc
`
`anti-CD3
`
`anti-CD8
`
`anti-CD4
`
`Figure 1. T cell products meet release requirements. Depicted from left to right: Southern blots of T cell genomic DNA using an HyTK-specific probe
`showing existence of single bands as indicated by arrows; Western blots revealing both the 16-kDA endogenous CD3z and the 66-kDA CE7R chimeric z
`bands detected with anti-human CD3z cytoplasmic tail specific antibody; flow cytometry analysis for surface expression of the chimeric receptor using
`anti-Fc antibody, or for the T cell markers CD8, CD4, and TCR or CD3, where isotype control staining is indicated with the open histogram; ability of
`CTL clones to lyse CD191 CD201 Daudi targets was determined in a 4-hour 51Cr release assay; ganciclovir (GCV) sensitivity using a flow cytometry–
`based assay for viable cell numbers after 14 days of culture with either rHuIL-2 or rHuIL-2 1 GCV; assays for IL-2 dependence were performed using
`3H-thymidine incorporation measurements (cpm) of Jurkat T cells versus the indicated T cell clones after 11 days of culture in the absence of rHuIL-2
`(UPN006, UPN009), or using a flow cytometry–based assay for viable cell numbers after the T cell products were cultured in the presence versus the
`absence of rHuIL-2 for 14 days (UPN035 and UPN037). N.D., not done.
`
`chromium release assays. Furthermore, all of the cell
`lines
`retained their dependence on exogenous
`rHuIL-2 for survival and proliferation, and the
`HyTK-expressing lines of IRB 01160 tested positive
`for sensitivity to GCV-mediated ablation.
`
`Treatment Experience
`
`As depicted in Figure 2, the intrapatient dose esca-
`lations were carried out as planned (compare with
`Figure S2), with the exception that the 1010/m2 cell
`dose was never given in IRB 98142 because of protocol
`toxicity dose modification rules
`(UPN006 and
`UPN009). Indeed, because of grade 2 hepatic toxic-
`ities that were noticed with the first infusion dose of
`108/m2 in UPN006, the second infusion was repeated
`at 108/m2, followed by an escalation to 109/m2 for the
`third infusion. Patient UPN009 exhibited a drop in he-
`moglobin after the second infusion that, although clin-
`ically insignificant (from 10.6 to 8.6), represented
`a Common Toxicity Criteria grade change from grade
`1 to grade 2 anemia status, requiring repetition of the
`
`109/m2 dose for the third infusion based on the proto-
`col’s defined rules for dose escalation. In UPN006, the
`second infusion was cancelled and rescheduled because
`of a puncture in the bag, which compromised the in-
`tegrity of the T cell product. In UPN037, the last in-
`fusion was cancelled because of the detection of
`contaminated T cell product. Neither myeloablation
`(MA) followed by HSCT nor Flu resulted in a drop
`in absolute lymphocyte count (ALC) below the normal
`range (Table S3).
`There were no grade 3 or higher adverse events
`with a possible correlation to administration of 108
`T cells per m2. However, examination of the adverse
`events at 109 T cells per m2 revealed one case of
`grade 3 self-limited lymphopenia in both IRB
`98142 and IRB 01160, possibly attributed to cell
`administration (Table 1). At 2  109 T cells per
`m2, grade 3 lymphopenia and grade 3 eosinophilia
`each occurred once in IRB 01160; both resolved
`spontaneously with no adverse sequelae to the
`patients. Overall, the safety profile of this adoptive
`transfer therapy was acceptable.
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`1250
`
`M. C. Jensen et al.
`
`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`UPN006
`
`e m C e l l s
`
`S t
`
`BCNU
`
`Cytoxan
`
`VP-16
`
`2
`
`/ m
`
`8
`
`0
`
`s i o
`
`I n f u
`
`n C a
`
`d
`
`2
`
`e l l e
`
`c
`
`n
`
`e #
`
`s
`
`D o
`
`2
`
`/ m
`
`8
`
`0
`
`: 1
`
`s
`
`D o
`
`: 1
`
`3
`
`e #
`
`2
`
`/ m
`
`9
`
`0
`
`: 1
`
`1
`
`e #
`
`s
`
`D o
`
`d -37 d0 d14 d28 d42
`
`e m C e l l s
`
`S t
`
`2
`
`/ m
`
`8
`
`0
`
`s
`
`D o
`
`: 1
`
`2
`
`e #
`
`2
`
`/ m
`
`9
`
`0
`
`2
`
`/ m
`
`9
`
`0
`
`s
`
`D o
`
`: 1
`
`3
`
`e #
`
`: 1
`
`1
`
`e #
`
`s
`
`D o
`
`UPN009
`
`TBI
`
`Cytoxan
`
`VP-16
`
`d -28 d0 d14 d28
`
`UPN035
`
`2
`
`/ m
`
`8
`
`2
`
`9
`
`2
`
`9
`
`2
`
`/ m
`
`9
`
`0
`
`2
`
`/ m
`
`9
`
`0
`
`0
`
`: 1
`
`1
`
`e #
`
`s
`
`D o
`
`0
`
`: 1
`
`2
`
`0
`
`: 1
`
`3
`
`/ m
`
`e #
`
`s
`
`D o
`
`/ m
`
`e #
`
`s
`
`D o
`
`1
`
`x
`
`: 2
`
`4
`
`e #
`
`s
`
`D o
`
`1
`
`x
`
`: 2
`
`5
`
`e #
`
`s
`
`D o
`
`d0 d4 d8 d14 d21 d26 d28 d29 d31 d35
`
`Fludarabine
`
`IL-2
`
`IL-2
`
`2
`
`/ m
`
`8
`
`0
`
`UPN037
`
`: 1
`
`1
`
`e #
`
`s
`
`D o
`
`2
`
`/ m
`
`9
`
`0
`
`2
`
`/ m
`
`9
`
`0
`
`s
`
`D o
`
`: 1
`
`3
`
`e #
`
`: 1
`
`2
`
`e #
`
`s
`
`D o
`
`2
`
`/ m
`
`9
`
`0
`
`1
`
`x
`
`s i o
`
`I n f u
`
`: 2
`
`4
`
`e #
`
`s
`
`D o
`
`d
`
`e l l e
`
`c
`
`n
`
`n C a
`
`d0 d4 d8 d14 d21 d22 d26 d28 d30 d35 d38
`
`Fludarabine
`
`IL-2
`
`IL-2
`
`Figure 2. Treatment regimens for each patient. First i.v. infusions of T cells were administered on day 0 for each patient. For UPN006 and UPN009,
`fractionated total body irradiation (TBI) and/or myeloablative chemotherapies administered to UPN006 and UPN009 are indicated just before admin-
`istration of CD341 autologous stem cells. BCNU, bis-chloronitrosourea; cytoxan, cyclophosphamide; VP-16, etoposide. For UPN035 and UPN037,
`administration of fludarabine (i.v. at 25 mg/m2) occurred between days 4 and 8 after the first T cell infusion, and rHuIL-2 administration (5  105
`IU/ m2 BID) was initiated after the third T cell infusion.
`
`Follow-Up Clinical Status of Patients
`
`Although this was a phase I clinical trial with a pri-
`mary purpose of determining safety, we also moni-
`tored the disease and survival status of each patient.
`For IRB 98142 (CD20R; DLCL), UPN006 (last infu-
`sion on February 24, 2000) relapsed in September
`2001, whereas UPN009 (last infusion on November
`22, 2000) continues to be in remission after autologous
`HSCT. At the time of the writing of this report, both
`UPN006 (after additional treatment) and UPN009 are
`alive and in remission. For IRB 01160 (CD19R; FL),
`UPN035 (last infusion on May 4, 2006) presented
`
`with a new diagnosis of CD191/CD201 DLCL in
`June 2006, and died in June 2007. UPN037 (last infu-
`sion on February 15, 2007) displayed progression on
`computed tomography scan in September 2007, and
`is currently alive and undergoing additional treatment.
`
`In Vivo Persistence of Transferred T Cells
`
`Quantitative PCR performed to detect CD20R
`and CD19R plasmid copy numbers in PBMCs as a sur-
`rogate marker of the presence of adoptively transferred
`T cells showed varying T cell persistence among pa-
`tients (Figure 3). Only 1 of the 4 patients (UPN006)
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`Adoptive Therapy with Redirected T Cells for Lymphoma
`
`1251
`
`A
`
`UPN
`006
`
`UPN
`009
`
`B
`
`UPN
`035
`
`CD19R+ cells in PBMC
`
`UPN
`037 %
`
`109/m2
`
`108/m2
`
`108/m2
`
`0.015
`
`0.010
`
`* *
`
`109/m2
`
`109/m2
`
`108/m2
`
`* * *
`
`56
`49
`43
`42
`35
`29
`28
`27
`25
`15
`14
`
`017
`
`Day
`
`0.005
`
`0
`0.025
`
`0.020
`
`0.015
`
`0.010
`
`0.005
`
`0
`
`%CD20R+ cells in PBMC
`
`*
`
`2x109/m2
`
`2x109/m2
`
`*
`
`2x109/m2
`
`109/m2
`
`109/m2
`
`108/m2
`
`0.100
`
`0.075
`
`109/m2
`
`109/m2
`
`108/m2
`
`0.050
`
`0.025
`
`0
`2.0
`
`1.5
`
`1.0
`
`0.5
`
`0
`
`* *
`
`42
`41
`36
`35
`29
`28
`26
`22
`21
`15
`14
`
`Day
`
`014
`
`Figure 3. Transferred T cells do not persist long term in vivo. Using
`real time quantitative PCR, the percent of cells in the PBMC that
`were positive for the CD20R (A) or CD19R (B) genes were determined
`as an indicator of the relative amount of chimeric receptor expressing T
`cells in the PBMC samples collected at the indicated days during the
`treatment schedule. Escalating infusion doses are indicated by arrows.
`*Cells not harvested.
`
`irradiated 6D10 cells were cloned in limiting dilution;
`all clones were similarly specific for NeoR (Figure 4C).
`For IRB 01160 (CD19R; FL), the development of
`both antibody and cellular immune responses against
`the infused T cell products was evaluated. For the an-
`tibody analyses, serum collected from UPN035 and
`UPN037 at enrollment and after T cell administration
`were both negative for antibody reactivity against the
`surface-expressed CD19R transgene using a flow
`cytometry–based assay (Figure 5A); however, evidence
`was found for cell-mediated immunoreactivity against
`the infused T cells. Examination of the TCR Vb gene
`
`Table 1. Adverse Event Summary
`
`IRB Trial
`
`98142
`
`01160
`
`T Cell
`Dose, Cells/m2
`
`109
`
`109
`2  109
`
`Event*
`
`Occurrences
`
`Lymphopenia
`
`Lymphopenia
`Lymphopenia
`Eosinophilia
`
`1
`
`1
`1
`1
`
`IRB indicates internal review board; NCI, National Cancer Institute.
`*Only events of grade 3 or higher, according to the NCI Common Toxicity
`Criteria, with possible attribution to T cell administration are reported.
`
`had a detectable level of transferred T cells at 1 week
`after the first infusion of 108 cells/m2. A detectable
`level of transferred T cells at 1 week after infusion of
`109 cells/m2 was found in only 2 of the 7 higher doses
`(UPN006 infusion 3 and UPN009 infusion 2), and at
`no time were transferred T cells detected at 1 week af-
`ter infusion of 2  109 cells/m2. Thus, adoptively
`transferred T cell persistence did not appear to corre-
`late with cell dose. Compared with the persistence af-
`ter the initial
`infusion, UPN009, UPN035, and
`UPN037 also displayed significantly reduced levels
`of transferred T cells 24 hours after each additional in-
`fusion, suggesting the possibility of an antitransgene
`immune response mounted against the administered
`T cells (Figure 3).
`
`Detection of Transgene-Specific Immune
`Responses
`
`For IRB 98142 (CD20R; DLCL), the develop-
`ment of cellular immune responses against the infused
`T cell products was evaluated. For these analyses,
`PBMCs were collected from UPN006 and UPN009
`before and after T cell administration, and, after in vi-
`tro stimulation, were compared for cytotoxic activity
`using chromium-release assays (Figure 4). The use of
`irradiated lymphoblastoid cells (xLCLs) to stimulate
`the PBMCs resulted in successful lysis of 51Cr-labeled
`LCLs, indicating that functional effector cells could be
`derived from each patient’s PBMC samples. Interest-
`ingly, when the irradiated autologous T cell clones
`were used to stimulate the PBMCs, cytotoxic re-
`sponses were seen against the 51Cr-labeled T cell clone
`only in the posttreatment sample collected from
`UPN009 (Figure 4A). This immunoreactivity against
`the T cell clone 6D10 used in therapy appeared to be
`specific for neomycin phosphotransferase but not the
`CAR, because cytotoxic responses could be observed
`against 51Cr-labeled LCLs that had been transduced
`with the pcDNA3.1(-) vector, which directs the ex-
`pression of neomycin phosphotransferase, but lacks
`the CD20R transgene (Figure 4B). To better analyze
`the specificity of the rejection response, UPN009’s
`posttreatment PBMCs that had been stimulated with
`
`UPenn Ex. 2032
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`1252
`
`M. C. Jensen et al.
`
`Biol Blood Marrow Transplant 16:1245-1256, 2010
`
`Pre-Trtmt
`
`Post-Trtmt
`
`50:1
`
`25:1
`
`5:1
`
`1:1
`50:1
`Effector:Target
`
`25:1
`
`5:1
`
`1:1
`
`Pre-Trtmt
`
`Post-Trtmt
`
`100
`80
`
`% Cr Release
`
`51
`
`60
`40
`20
`0
`
`50:1
`
`25:1
`
`5:1
`
`1:1
`50:1
`Effector:Target
`
`25:1
`
`5:1
`
`1:1
`
`A
`
`UPN
`006
`
`UPN
`009
`
`B
`
`UPN
`009
`
`100
`80
`60
`40
`20
`0
`
`100
`80
`
`60
`40
`20
`0
`
`% Cr Release
`
`51
`
`Stim/Target:
`
`xLCL/LCL
`xLCL/1A11
`x1A11/LCL
`x1A11/1A11
`
`Stim/Target:
`
`xLCL/LCL
`xLCL/6D10
`x6D10/LCL
`x6D10/6D10
`
`Stim/Target:
`
`xLCL/LCL
`xLCL/LCLpcDNA
`x6D10/LCL
`x6D10/LCLpcDNA
`
`Target:
`
`LCL
`6D10
`LCLpcDNA
`
`F2
`
`E2
`
`D7
`
`D4
`
`Post-Trtmt PBMC clones
`
`(stim w/ x6D10)
`
`C
`
`UPN
`009
`
`-25 0 25 50 75 100 125 150 175 200
`51
`% Cr Release (25:1)
`
`Figure 4. Transgene rejection response detected when T cells administered after HSCT. In the trial targeting CD201 diffuse large-cell lymphoma,
`PBMCs collected before treatment

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