throbber
CLINICAL TRIALS AND OBSERVATIONS
`
`Safety and persistence of adoptively transferred autologous CD19-targeted T cells
`in patients with relapsed or chemotherapy refractory B-cell leukemias
`*Renier J. Brentjens,1-3 *Isabelle Rivie` re,1-4 Jae H. Park,1,2 Marco L. Davila,1,2 Xiuyan Wang,2-4 Jolanta Stefanski,2-4
`Clare Taylor,2-4 Raymond Yeh,1,2 Shirley Bartido,2,3 Oriana Borquez-Ojeda,2-4 Malgorzata Olszewska,2-4 Yvette Bernal,1
`Hollie Pegram,1,2 Mark Przybylowski,2-4 Daniel Hollyman,2-4 Yelena Usachenko,1,2 Domenick Pirraglia,2-4 James Hosey,2-4
`Elmer Santos,3,5 Elizabeth Halton,1 Peter Maslak,1 David Scheinberg,1-3 Joseph Jurcic,1 Mark Heaney,1 Glenn Heller,6
`Mark Frattini,1 and Michel Sadelain1-3
`
`1Department of Medicine, 2Center for Cell Engineering, 3Molecular Pharmacology and Chemistry Program, 4Cell Therapy and Cell Engineering Facility,
`5Department of Radiology, and 6Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY
`
`We report the findings from the first 10
`patients with chemotherapy-refractory
`chronic lymphocytic leukemia (CLL) or
`relapsed B-cell acute lymphoblastic leuke-
`mia (ALL) we have enrolled for treatment
`with autologous T cells modified to ex-
`press 19-28z, a second-generation chime-
`ric antigen (Ag) receptor specific to the
`B-cell lineage Ag CD19. Eight of the 9
`treated patients tolerated 19-28zⴙ T-cell
`infusions well. Three of 4 evaluable pa-
`tients with bulky CLL who received prior
`conditioning with cyclophosphamide ex-
`Introduction
`
`hibited either a significant reduction or a
`mixed response in lymphadenopathy
`without concomitant development of B-
`cell aplasia. In contrast, one patient with
`relapsed ALL who was treated in remis-
`sion with a similar T-cell dose developed
`a predicted B-cell aplasia. The short-term
`persistence of infused T cells was enhanced
`by prior cyclophosphamide administration
`and inversely proportional to the peripheral
`blood tumor burden. Further analyses
`showed rapid trafficking of modified T cells
`to tumor and retained ex vivo cytotoxic
`
`potential of CD19-targeted T cells retrieved
`8 days after infusion. We conclude that this
`adoptive T-cell approach is promising and
`more likely to show clinical benefit in
`the setting of prior conditioning chemo-
`therapy and low tumor burden or mini-
`mal residual disease. These studies are
`registered at www.clinicaltrials.org as
`#NCT00466531 (CLL protocol) and
`#NCT01044069 (B-ALL protocol). (Blood.
`2011;118(18):4817-4828)
`
`Despite currently available therapies, most patients with B-cell
`leukemias, including chronic lymphocytic leukemia (CLL) and
`B-cell acute lymphoblastic leukemia (B-ALL), are incurable.1,2 For
`this reason, novel therapeutic strategies are needed. The adoptive
`transfer of genetically engineered immune effector cells aims to
`rapidly establish T cell–mediated tumor immunity.3,4 In this ap-
`proach, the patient’s own T cells are targeted to tumor cells through
`a transgene-encoded Ag receptor consisting of either TCR chains or
`a chimeric Ag receptor (CAR). CARs are composed of an
`extracellular Ag recognition domain, most commonly a single
`chain fragment variable derived from a mAb, fused to a transmem-
`brane domain, and a cytoplasmic signaling domain, most com-
`monly including that of the CD3␨ chain.3-10 When expressed in
`T cells, CARs efficiently redirect T-cell specificity and cytotoxicity
`to cells expressing the targeted Ag in HLA-independent manner.11-18
`We have previously generated a series of CARs specific for the
`CD19 Ag,11,12 a member of the Ig superfamily and component of a
`B-cell surface signal transduction complex.19 Expression of CD19
`is restricted to B-lineage cells and possibly follicular dendritic cells
`and is found in most B-cell malignancies, including CLL and
`B-ALL.19-23 Significantly, CD19 is not expressed in hematopoietic
`stem cells. The immunologic targeting of CD19 therefore carries a
`
`minimal risk of autoimmunity or BM toxicity other than the
`potential induction of B-cell aplasias.
`In preclinical studies, human T cells expressing CD19-specific
`CARs efficiently lysed a wide panel of human CD19⫹ tumor cell
`lines as well as freshly isolated patient B-cell tumors.11 Signifi-
`cantly, intravenously administered CD19-targeted human periph-
`eral blood T cells eradicated systemic CD19⫹ tumors established in
`SCID-Beige mice.11,12 Our in vivo studies further showed enhanced
`antitumor efficacy by providing costimulatory signals to adoptively
`transferred T cells. Because most B-cell leukemias fail to express
`ligands for activating costimulatory receptors,24,25 we overcame
`this limitation by replacing the inert CD8 transmembrane domain
`with the transmembrane and cytoplasmic signaling domains of the
`T-cell costimulatory CD28 receptor,26 resulting in the 19-28z CAR,
`which enhances antitumor efficacy in SCID-Beige mice bearing
`CD19⫹ leukemias.12 On the basis of these preclinical data we chose
`to translate this approach to the clinical setting with the use of the
`19-28z CAR.
`After the validation of a robust process for large-scale human
`T-cell transduction and expansion,27 we enrolled 10 patients with
`either chemotherapy refractory CLL or relapsed B-ALL on 2 phase
`1 dose escalation clinical trials. The primary objective of these
`
`Submitted April 13, 2011; accepted August 5, 2011. Prepublished online as
`Blood First Edition paper, August 17, 2011; DOI 10.1182/blood-2011-04-348540.
`
`*R.J.B. and I.R. contributed equally to this study.
`
`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.
`
`An Inside Blood analysis of this article appears at the front of this issue.
`
`© 2011 by The American Society of Hematology
`
`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`4817
`
`UPenn Ex. 2012
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`4818
`
`BRENTJENS et al
`
`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`trials is to assess the safety of infusing 19-28z⫹ T cells with or
`without prior cyclophosphamide-conditioning chemotherapy. Sec-
`ondary objectives include assessment of clinical responses to
`therapy and the effect of cyclophosphamide conditioning on
`disease response, T-cell persistence, and T-cell function. Herein, we
`report our findings on the first 10 patients, 9 of which were infused
`with the manufactured T cells. We were able to generate sufficient
`CAR-transduced T cells from leukapheresis products derived from
`all enrolled patients. Either a marked reduction in tumor burden
`stable disease or B-cell aplasia was observed in 4 of the 5 evaluable
`patients given cyclophosphamide before T-cell infusion. We dem-
`onstrate rapid trafficking of 19-28z⫹ T cells to sites of tumor
`involvement and report on the short-term persistence and function
`of the adoptively transferred T cells. Collectively, these data show
`the promise of CD19-targeted T cells for the treatment of B-cell
`malignancies and provide insights into how to optimally apply this
`strategy in the clinical setting.
`
`rus by PCR and marker rescue cell culture assay; residual Dynabeads
`ClinExVivo CD3/CD28; negative bacterial, fungal, and Mycoplasma
`cultures; endotoxin level no ⬎ 5 EU/kg; Gram stain–negative on day of
`infusion; ⬎ 80% cell viability; and CD3⫹, CD8⫹, CD5⫹ (patients with
`CLL), CD10⫹ (patients with ALL), and CD19⫹ phenotype by flow
`cytometry and CD19-specific cytotoxicity. The assays were performed as
`described in Hollyman et al27 and Taylor et al.29
`
`Restimulation of T cells from postinfusion PBMCs with
`Dynabeads ClinExVivo CD3/CD28
`
`PBMCs collected in cell preparation tubes (BD Biosciences) were purified
`from whole blood according to the manufacturer’s recommendations.
`Patient CD3⫹ T cells were subsequently selected and activated with
`Dynabeads ClinExVivo CD3/CD28 (Invitrogen) at ratio of 3 beads to
`1 CD3⫹ T cell. CD3⫹ T cells were cultured in X-VIVO 15 medium (Lonza)
`supplemented with 100 U/mL IL-2. Cell samples were taken at different
`time points to determine the average vector copy number by quantitative
`RT-PCR and the expression of 19-28z CAR by flow cytometry.
`
`Methods
`
`Clinical protocols
`
`CLL protocol (NCT00466531). Patients with relapsed purine analog-
`refractory CLL are eligible for enrollment. This is a phase 1 dose escalation
`trial. Patients initially undergo leukapheresis for T-cell collection. In the
`first step, one cohort of 3-6 patients is treated with the lowest initial dose of
`T cells, level 1 (1.2-3.0 ⫻ 107 19-28z⫹ T cells/kg), without prior cyclophos-
`phamide administration. In the second step with 2 cohorts, patients are
`treated with dose-escalating cyclophosphamide chemotherapy (1.5 and
`3.0 g/m2) followed 2 days later by infusion of modified T cells at dose level
`1. After the death on study of the first patient treated in the second cohort,28
`the ⫺1 T-cell dose level
`the subsequent 3 patients were treated at
`(0.4-1.0 ⫻ 107 19-28z⫹ T cells/kg) with T-cell infusions split over 2 days to
`enhance safety. Enrollment on this second cohort has been completed.
`B-ALL protocol (NCT01044069). Adult patients with CD19⫹ B-ALL
`are eligible for enrollment. Patients can be either enrolled in first complete
`remission (CR1) after ⱖ 1 round of consolidation therapy or on presenta-
`tion with relapsed or refractory disease, defined as no CR after ⱖ 2 induc-
`tion regimens. Patients enrolled in CR1 underwent leukapheresis but are
`treated with T cells only after relapse. Patients enrolled with relapsed or
`refractory B-ALL leukapheresed, treated with a salvage reinduction regi-
`men, and regardless of remission status, receive cyclophosphamide (3.0 g/
`m2) followed 2 days later by split dose infusion of autologous 19-28z⫹
`T cells. This is a standard phase 1 dose-escalation trial with 3 planned T-cell
`doses, 3 ⫻ 106, 1 ⫻ 107, and 3 ⫻ 107 19-28z⫹ T cells/kg. Patients enrolled
`on this protocol are not precluded, if eligible, to undergo additional therapy
`with allogeneic stem cell transplantation after the modified T-cell infusion.
`Both trials were approved by the Memorial Sloan-Kettering Cancer
`Center Institutional Review Board,
`the Recombinant DNA Advisory
`Committee of the National Institutes of Health, and are supported by
`BB-IND 13266 approved by the US Food and Drug Administration. In both
`trials, informed consent is obtained from eligible patients in accordance
`with the Declaration of Helsinki. Patients enrolled on these trials do not
`receive IL-2. Adverse events during and after therapy were assessed
`according to the National Institutes of Health Common Terminology
`Criteria for Adverse Events Version 3.0 (http://ctep.cancer.gov/). The
`Memorial Sloan-Kettering Cancer Center Data and Safety Monitoring
`Board review all safety data every 6 months.
`
`Generation and expansion of genetically modified T cells
`
`19-28z–transduced T cells were generated as described.27 Briefly, T cells
`were isolated from leukapheresis product and activated with Dynabeads
`ClinExVivo CD3/CD28. Release criteria include 19-28z CAR expression
`by flow cytometry; average vector copy number by quantitative PCR;
`vector identity by Southern blot; absence of replication competent retrovi-
`
`Restimulation of T cells from postinfusion PBMCs with
`CD19ⴙCD80ⴙ artificial APCs
`
`Human CD19 and CD80 expressing fibroblasts (3T3-CD19-CD80)11 were
`irradiated at 30 Gy. Postinfusion PBMCs were thawed and plated on the
`3T3-CD19-CD80 cells in X-VIVO 15 media (Lonza) supplemented with
`5% human AB serum (GEMINI) and 100 U/mL IL-2. Lysis of 3T3 cells
`was monitored, and PBMCs were fed on day 3. Cells were counted 7 days
`after restimulation and stained with anti–human CD3-FITC, CD8-eFluor
`450, CD4-PE-Cy7, biotinylated goat anti–mouse IgG F(ab)2 followed by
`APC-labeled streptavidin, and 7-amino-actinomycin D (7-AAD) with the
`use of standard staining procedures. Data acquisition was performed on a
`LSRII flow cytometer, and data analysis was performed with FlowJo
`software (TreeStar Inc).
`
`Characterization of cytokine profiles secreted by EOP
`19-28z–transduced T cells
`
`Samples of patient T cells transduced with 19-28z CAR were taken at the
`end of production (EOP). T cells (1.0 ⫻ 106) were plated on 3T3 artificial
`APCs (AAPCs) expressing CD19 or untransduced 3T3 fibroblasts as
`controls, in complete X-Vivo culture media in 5% human AB serum and in
`the absence of ILs. After 48 hours supernatants were collected and assayed
`for human cytokines on a Luminex IS100 instrument as per manufacturer’s
`instructions (Millipore Corp). The cytokine levels were normalized to
`transduction efficiency. In addition, background levels measured on untrans-
`duced 3T3 fibroblasts were subtracted.
`
`Other methods
`
`including manufacture of clinical grade PG13-19-28z
`Other methods,
`vector stocks, serum cytokine analyses, cytotoxic T-lymphocyte assays,
`quantitative real-time PCR, Abs and reagents, flow cytometry, statistics,
`and IHC can be found in supplemental Methods (available on the Blood
`Web site; see the Supplemental Materials link at the top of the online
`article).
`
`Results
`
`Patient characteristics
`
`Eight patients with relapsed purine analog-refractory CLL have
`been enrolled and treated (Table 1). The median age of patients was
`68 years with 7 of 8 being men. All patients were previously treated
`with 1-4 different chemotherapy regimens, with 7 of 8 patients
`having received ⱖ 2 different regimens before enrollment. Chromo-
`somal and genetic analyses of CLL tumor cells showed adverse
`prognostic features, including del17p with concomitant p53 gene
`
`UPenn Ex. 2012
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`AUTOLOGOUS CD19-TARGETED T CELLS IN CLL/ALL
`
`4819
`
`Table 1. Patient characteristics
`Age at
`Age at
`diagnosis,
`treatment,
`y
`y
`
`Patient
`ID
`
`Sex
`
`Indication
`for treatment
`
`CLL-1
`CLL-2
`CLL-3
`CLL-4
`CLL-5
`CLL-6
`CLL-7
`CLL-8
`
`ALL-1
`
`ALL-2
`
`44
`66
`62
`63
`65
`56
`52
`58
`
`66
`
`45
`
`51
`72
`73
`69
`68
`68
`62
`61
`
`67
`
`48
`
`M
`M
`F
`M
`M
`M
`M
`M
`
`M
`
`F
`
`Bulky LAD
`Bulky LAD
`Bulky LAD
`Bulky LAD
`Bulky LAD
`Bulky LAD
`Bulky LAD
`Bulky LAD
`
`Prior therapies
`
`FCR, PCRM
`FR, RCVP, PCRM
`Chlorambucil, PCR, PCRM
`R, PCRM
`PCR
`RCVP, PCR, Bendamustine
`CVP, RC, PCR, PCRM
`RCVP, Alemtuzumab
`
`Genetic
`abnormalities/IgVH
`mutation status
`
`WBC
`count,
`ⴛ 103/␮L
`
`ALC,
`ⴛ 103/␮L
`
`Hgb
`level,
`g/dL
`
`PLT
`count,
`ⴛ103/␮L
`
`del11q
`Unmutated IgVH
`Normal karyotype
`del11q
`del11q, trisomy 12
`del11q, inv1, unmutated IgVH
`del17p, unmutated IgVH
`del17p, monosomy 14,
`monosomy 15
`Normal karyotype
`
`Normal karyotype
`
`200.6
`4.2
`136.4
`187.1
`76.3
`97.1
`1.9
`5.4
`
`2.9
`
`ⴱ
`
`196.6
`3.4
`132.3
`174
`66.4
`92.2
`1
`3.3
`
`7.1
`9.9
`8.9
`9.9
`10
`8.9
`10
`11.6
`
`26
`60
`100
`189
`162
`174
`61
`41
`
`0.7
`
`8.6
`
`126
`
`ⴱ
`
`ⴱ
`
`ⴱ
`
`Relapsed disease C, Mitoxantrone, vincristine,
`etoposide
`Relapsed disease HyperCVAD, mitoxantrone,
`cytarabine, vincristine
`
`IgVH indicates immunoglobulin heavy chain; WBC, white blood cell; ALC, absolute lymphocyte count; Hgb, hemoglobin; PLT, platelet; LAD, lymphadenopathy;
`FCR, fludarabine, cyclophosphamide, rituximab; PCRM, pentostatin, cyclophosphamide, rituximab, mitoxantrone; FR, fludarabine, rituximab; RCVP, rituximab, cyclophosph-
`amide, vincristine, prednisone; PCR, pentostatin, cyclophosphamide, rituximab; R, rituximab; CVP, cyclophosphamide, vincristine, prednisone; RC, rituximab, cyclophosph-
`amide; C, cyclophosphamide; and HyperCVAD, cyclophosphamide, vincristine, doxorubicin, dexamethasone.
`*This patient is yet to be treated with modified T cells.
`
`deletion, del11q, and/or an unmutated Ig variable heavy chain
`domain. All patients exhibited advanced disease and tumor burden
`as evidenced by marked bulky lymphadenopathy.
`Two patients with relapsed B-ALL have been enrolled and 1
`patient has been treated (Table 1). The latter achieved an initial
`remission after induction therapy but subsequently relapsed after
`the third cycle of consolidation therapy at which point the patient
`was enrolled and T cells were obtained by leukapheresis. The patient
`achieved a second remission after reinduction chemotherapy and
`was treated on protocol. The second patient had a relapse of disease
`after initial induction therapy but became ineligible for treatment
`with modified T cells because of intervening complications.
`
`Generation of patient-derived 19-28z–transduced T cells
`
`Despite all patients having been heavily pretreated, we were able to
`obtain adequate numbers of T cells from the leukapheresis products
`in every case (Table 2). The mean time in culture to achieve the
`19-28z⫹ T-cell dose was 16 days (range, 11-19 days) with a mean
`120-fold expansion of T cells (range, 24- to 385-fold). The
`transduction efficiency in CD3⫹ T cells, as assessed by flow
`cytometry, ranged from 23% to 70% in CLL patient cells and from
`4% to 8.6% in ALL patient cells. The average vector copy number
`
`per cell in these cell products ranged from 0.06 to 1.5 with a mean
`of 0.67.
`
`Characterization of final 19-28z–transduced T cells
`
`The final T-cell products exhibited a predominant CD4⫹ pheno-
`type, particularly pronounced in the CLL EOP cells (mean CD4⫹
`T-cell fraction of 88%, CD4/CD8 ratio of 10.5; Tables 3 and 4). The
`patients with B-ALL showed a mean CD4⫹ T-cell fraction of 63%
`(Table 3). Despite a marked prevalence of CD4⫹ T cells, all EOP
`T cells displayed robust in vitro cytotoxic activity against both
`autologous CLL tumor cells and the CD19⫹ Raji Burkitt lymphoma
`cell line (Table 2). Further analyses of final T-cell products showed
`minimal numbers of CD4⫹ FoxP3⫹ T regulatory cells and a
`significant retention of cell surface markers,
`including CD27,
`CD28, and CD62L. Although the percentage of these markers
`showed some variability (Table 3), the absolute numbers of infused
`T cells were overall similar in all patients (supplemental Figure 1)
`as well as their distribution in CAR– versus CAR⫹ T cells (Table 3;
`supplemental Figure 1). To further assess the infused cell products,
`all EOP cells were activated in Ag-specific fashion in vitro under
`the same conditions with the use of CD19⫹ AAPCs.11,30 The
`cytokine signatures induced by exposure to CD19 were overall
`
`Table 2. Characteristics of the infused 19-28z–transduced T cells
`Test
`CLL-1
`CLL-2
`CLL-3
`
`CLL-4
`
`CLL-5
`
`CLL-6
`
`CLL-7
`
`CLL-8
`
`ALL-1
`
`ALL-2
`
`CD3ⴙ T cells, %
`Apheresis product
`EOP cells
`Tumor cells, %
`Apheresis product
`EOP cells
`CD3⫹/19-28z CAR⫹, %
`Cytotoxicity (25:1)
`Auto-B tumor cells
`Raji tumor cells
`Fold expansion after transduction
`(days in culture)
`Total 1928z⫹/CD3⫹ cells infused
`Total CD3⫹ cells infused
`
`5.4
`98
`
`94
`0.1
`23
`
`45
`100
`
`4
`0
`31
`
`7.2
`100
`
`90
`0
`53
`
`5.1
`99
`
`94
`0
`70
`
`15.9
`98
`
`50
`1
`32
`
`6.2
`99
`
`94
`0
`39
`
`25
`85
`
`28
`1.2
`25
`
`22.5
`100
`
`69
`0
`39.3
`
`45
`99
`
`3.1
`0
`8.6
`
`67
`100
`
`0
`0.2
`4
`
`58.8
`51.1
`53 (17)
`
`ND
`47.1
`184 (19)
`
`52.3
`52.3
`23.6 (18)
`
`48.7
`46.5
`112 (15)
`
`33.7
`43.3
`191 (18)
`
`59.5
`73.8
`51 (15)
`
`35.7
`72.9
`62.4 (15)
`
`28.3
`24.8
`385 (16)
`
`ND
`29.2
`49.4 (d11)
`
`ND
`8.6
`88.5 (d11)
`
`2.5 ⫻ 109
`11.1 ⫻ 109
`
`1.2 ⫻ 109
`3.7 ⫻ 109
`
`1.1 ⫻ 109
`2.1 ⫻ 109
`
`3.2 ⫻ 109
`4.6 ⫻ 109
`
`4 ⫻ 108
`1.2 ⫻ 109
`
`4 ⫻ 108
`1.0 ⫻ 109
`
`7.6 ⫻ 108
`3.0 ⫻ 109
`
`1.4 ⫻ 109
`3.5 ⫻ 109
`
`1.8 ⫻ 108
`2.1 ⫻ 109
`
`NA
`NA
`
`The average 19-28z vector copy number per cell from CLL-1 to ALL-2 patients was 0.34, 0.54, 0.66, 1.15, 0.81, 1.5, 0.86, 0.62, 0.14, and 0.06, respectively.
`
`UPenn Ex. 2012
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`4820
`
`BRENTJENS et al
`
`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`Table 3. Phenotype of the infused 19-28z–transduced T cells
`CD4ⴙ
`CD8ⴙ
`CD28ⴙCD27ⴚ
`CD28ⴙCD27ⴙ
`Patient ID
`
`CD62Lⴙ
`
`CCR7ⴙ
`
`CD25ⴙCD4ⴙFoxp3ⴙ
`
`Days in culture
`
`CLL-1
`CLL-2
`CLL-3
`CLL-4
`CLL-5
`CLL-5 (CAR⫹)
`CLL-6
`CLL-6 (CAR⫹)
`CLL-7
`CLL-7 (CAR⫹)
`CLL-8
`CLL-8 (CAR⫹)
`ALL-1
`ALL-1 (CAR⫹)
`ALL-2
`ALL-2 (CAR⫹)
`
`94
`96
`93
`99
`87
`87.4
`79
`78.6
`58
`66.6
`91.5
`90.3
`74
`62.8
`52
`47.9
`
`5
`5
`8
`0.7
`12
`11.9
`21
`20.8
`27
`21.5
`8.5
`9.7
`26
`25.6
`48
`51.8
`
`83.2
`86.1
`40.6
`86.9
`56.6
`62.9
`45.8
`57
`41.3
`49.1
`81.7
`82.4
`43.4
`52.3
`5.2
`28.5
`
`11.5
`6.1
`2
`5.9
`20.8
`16.6
`30.8
`19.3
`13.5
`13.3
`9.1
`7.8
`16.5
`15.9
`84.8
`64.1
`
`9.1
`15.5
`17.7
`34.4
`40
`40.4
`51.5
`52
`63.4
`54.3
`33.1
`28.1
`64.3
`78.1
`94.8
`86.1
`
`ND
`ND
`ND
`ND
`7.7
`10.1
`3.4
`3.7
`1.1
`1.4
`19.6
`16.6
`1.3
`1.3
`47.4
`36.5
`
`1.2
`0.61
`2.4
`0.9
`0.7
`ND
`1.3
`ND
`1.2
`ND
`ND
`ND
`1.7
`ND
`ND
`ND
`
`17
`19
`18
`15
`18
`ND
`15
`ND
`15
`ND
`16
`ND
`11
`ND
`13
`ND
`
`For patients CLL-1, CLL-2, CLL-3, and CLL-4, the CD4, CD8, and memory phenotypes were analyzed in 7-AAD⫺ populations. For patients CLL-5, CLL-6, CLL-7, ALL-1,
`and ALL-2, the CD4, CD8, and memory phenotypes were analyzed in both 7-AAD⫺ and CAR⫹ populations. All numbers are expressed in percentage of 7-AAD⫺ or CAR⫹
`populations as indicated.
`
`similar, showing in particular robust secretion of GM-CSF, IFN-␥,
`TNF-␣, MIP-1␣, MIP-1␤, and little or no IL-2, IL-4, and IL-10 in
`most patients (Figure 1).
`
`Infusion of 19-28z–transduced T cells is well tolerated
`
`Nine patients have received intravenous infusion of 19-28z–
`transduced autologous T cells. Overall, patients tolerated therapy
`well with most patients experiencing rigors, chills, and transient
`fevers within 24 hours of modified T-cell infusions. In all cases,
`patient blood and urine cultures were obtained, and intravenous
`antibiotic therapy was initiated. However, with the exception of
`patient CLL-4, all patients readily recovered from these symptoms
`with negative blood and urine cultures and were released after an
`additional 48 hours of in-patient observation (Table 5). Patient
`CLL-4 experienced persistent fevers, developed a sepsis-like
`syndrome with hypotension and renal failure, and died within
`48 hours of T-cell infusion. Previously published extensive analy-
`ses of this case point to an infectious cause as evidenced by marked
`serum cytokine abnormalities that preceded T-cell infusion, argu-
`ing against the 19-28z⫹ T cells being the primary cause of this
`
`adverse outcome.28 The subsequent cohort of 4 patients with CLL
`was treated at a 3-fold lower T-cell dose than CLL-4 and
`administered in a split-dose manner over 2 consecutive days. In
`addition, serum cytokine levels were assessed before both cyclo-
`phosphamide and T-cell infusions. Cytokine elevations, as noted in
`CLL-4, were not observed in the subsequently treated patients
`before or after cyclophosphamide therapy or after T-cell infusion.
`Significantly, serial serum analyses in these patients did not show
`evidence of cyclophosphamide-induced alterations in cytokine
`profiles compared with patients treated with T cells without
`cyclophosphamide conditioning (Figure 2).
`
`Clinical responses to 19-28zⴙ T-cell infusion
`
`Eight patients with CLL have been treated. The first cohort of
`3 patients was treated without cyclophosphamide conditioning at
`the lowest planned T-cell dose of 1.2-3.0 ⫻ 107 19-28z⫹ T cells/kg
`with no objective disease responses (Table 6). Collectively, all
`3 patients on this cohort exhibited further progressive disease
`and soon required additional salvage chemotherapy. All patients
`on this cohort have died of their disease. Patient CLL-4 died soon after
`
`Table 4. CD4/CD8 ratio in apheresis, EOP 19-28z transduced T cells, and blood samples after infusion
`Apheresis
`EOP 19-28z T cells
`
`Patient ID
`
`CLL-1
`CLL-2
`CLL-3
`CLL-4
`CLL-5
`CLL-6
`CLL-7
`CLL-8
`Mean ⫾ SD
`ALL-1
`ALL-2
`
`CD4/CD8
`
`2.4
`0.3
`3.5
`0.5
`0.8
`1.5
`2.5
`4.5
`2.0 ⫾ 1.5
`1.0
`1.8
`
`CD4ⴙCD28ⴙ/CD8ⴙCD28ⴙ
`
`2.4
`0.3
`6.8
`4.0
`1.0
`2.3
`7.0
`6.0
`3.7 ⫾ 2.6
`2.1
`1.8
`
`CD4/CD8
`
`18.8
`19.2
`11.6
`141.4
`7.3
`3.8
`2.5
`10.1
`10.5 ⫾ 6.6ⴱ
`2.5
`0.9
`
`19-28z T cells after infusion
`
`CD4/CD8 (time after infusion)
`
`NA
`NA
`NA
`70.4 (1 h); 33.5 (24 h)
`NA
`NA
`NA
`19 (2 wk); 8.7 (4 wk); 8.8 (5 wk)
`NA
`1.1 (8 d)
`NA
`
`All phenotypes are analyzed by flow cytometry in 7-AAD⫺ and CD45⫹ populations. All numbers are expressed in ratio of percentages of CD4/CD8 or
`CD4⫹CD28⫹/CD8⫹CD28⫹.
`*The average CD4/CD8 ratio excludes the CD4/CD8 ratio for CLL-4. With CLL-4, the average CD4/CD8 ratio is 26.8 ⫾ 46.7. CLL-4 had the lowest CD8⫹CD28⫹ fraction
`(see supplemental Table 1).
`
`UPenn Ex. 2012
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`AUTOLOGOUS CD19-TARGETED T CELLS IN CLL/ALL
`
`4821
`
`15000
`
`150
`
`15000
`
`1500
`
`2000
`
`25000
`
`20000
`
`10000
`
`100
`
`10000
`
`1000
`
`5000
`
`50
`
`5000
`
`500
`
`G-C SF
`
`0
`
`G M-C SF
`
`0
`
`IF N-alpha2
`
`m a
`
`0
`
`IF N-ga m
`
`0
`
`IL-3
`
`4000
`
`25000
`
`1500
`
`1000
`
`500
`
`0
`
`IL-17
`
`20000
`
`15000
`
`10000
`
`5000
`
`0
`
`MIP-1alpha
`
`300
`
`15000
`
`10000
`
`5000
`
`0
`
`MIP-1beta
`
`80
`
`60
`
`40
`
`20
`
`0
`
`pg/ml
`
`20000
`
`15000
`
`10000
`
`5000
`
`0
`
`3000
`
`2000
`
`1000
`
`0
`
`IL-10
`
`2000
`
`1500
`
`1000
`
`500
`
`200
`
`100
`
`CLL1
`CLL2
`CLL3
`CLL4
`CLL5
`CLL6
`CLL7
`CLL8
`ALL1
`ALL2
`3T3 CD19 ALONE
`
`IL-13
`
`0
`
`sC D40L
`
`0
`
`T N F-alpha
`
`2500
`
`2000
`
`1500
`
`1000
`
`500
`
`0
`
`150
`
`100
`
`50
`
`0
`
`pg/ml
`
`IL-2
`
`IL-4
`
`Figure 1. Cytokine analysis of EOP 19-28z–transduced T cells stimulated on CD19ⴙ AAPCs. EOP 19-28z–transduced T cells were plated on 3T3 fibroblast AAPCs
`expressing CD19. After 48 hours in culture, cell supernatants were assayed for cytokine levels. The cytokine levels were normalized to transduction efficiency. In addition,
`background cytokine levels measured on 3T3 fibroblasts that did not express CD19 were subtracted.
`
`infusion and therefore was not evaluable for clinical
`T-cell
`response.28
`
`Table 5. Adverse events
`Diagnosis-Patient
`
`Adverse events
`
`Grade
`
`CLL-1
`
`CLL-2
`
`CLL-3
`CLL-4
`
`CLL-5
`
`CLL-6
`
`CLL-7
`CLL-8
`ALL-1
`
`Febrile neutropenia
`Rigors, chills
`Fever, rigors, chills
`Chest pain
`Fever, rigors, chills
`Fever
`Rigors, chills, dyspnea
`Hypotension, renal failure
`Fever
`Rigors, chills
`Hyponatremia
`Fever
`Hypotension
`Febrile neutropenia
`Febrile neutropenia
`Fever
`Neutropenia
`Diarrhea
`Hypotension
`
`3
`2
`2
`2
`1
`2
`1
`5
`2
`1
`1
`1
`2
`3
`3
`2
`4
`2
`3
`
`Related
`
`Probable
`Probable
`Probable
`Probable
`Probable
`Probable
`Probable
`Possible
`Probable
`Probable
`Possible
`Probable
`Possible
`Possible
`Probable
`Probable
`Possible
`Possible
`Possible
`
`Patient ALL-2 is omitted because the patient became ineligible for modified T-cell
`infusion.
`
`The next 4 patients (CLL-5 to -8) were treated with
`cyclophosphamide-conditioning chemotherapy, followed by in-
`fusion of 0.4-1.0 ⫻ 107 19-28z⫹ T cells/kg. Patient CLL-5
`exhibited stable to progressive disease at 1 month after treatment as
`assessed by computed tomography scan but subsequently devel-
`oped marked objective reduction of peripheral lymphadenopathy in
`the absence of any further therapeutic interventions, as assessed by
`physical examination and computed tomography scans over the
`subsequent 2 months (Figure 3). This marked reduction of lymph-
`adenopathy remained stable over the subsequent 6 months, and
`thereafter the patient developed disease progression in the abdo-
`men with associated ascites and worsening cytopenias, which
`required further chemotherapy. The patient died 15 months after
`T-cell therapy with progressive chemotherapy refractory disease.
`Patient CLL-6 exhibited progressive disease ⬎ 1 month after
`therapy requiring salvage chemotherapy and died of infectious
`complications 2 months later. Patients CLL-7 and CLL-8, both
`treated in the setting of rapidly progressive disease with increasing
`lymphadenopathy and cytopenias, exhibited stable disease with
`respect to lymphadenopathy over a 4- and ⬎ 2-month period of
`expectant management, respectively, after cyclophosphamide and
`T-cell infusions (data not shown).
`Patient ALL-1 was a 67-year-old male who relapsed during
`consolidation therapy, achieved a second remission with salvage
`chemotherapy, and received a single consolidation with high-dose
`
`UPenn Ex. 2012
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`4822
`
`BRENTJENS et al
`
`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`CLL1
`CLL2
`CLL3
`CLL5
`CLL6
`CLL7
`ALL1
`
`No Cyclophosphamide
`
` Cyclophosphamide
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`10
`
`02468
`
`10
`
`02468
`
`250
`
`200
`
`150
`
`100
`
`50
`
`0
`80
`
`60
`
`pg/ml
`
`pg/ml
`
`pg/ml
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`10
`
`02468
`
`10
`
`02468
`
`250
`
`200
`
`150
`
`100
`
`50
`
`0
`80
`
`60
`
`40
`
`pg/ml
`
`pg/ml
`
`pg/ml
`
`IL-2
`
`IL-7
`
`IL-12 (p40)
`
`IL-15
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`40
`
`20
`
`0
`
`150
`
`100
`
`50
`
`0
`250
`
`200
`
`150
`
`100
`
`50
`
`pg/ml
`
`pg/ml
`
`pg/ml
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`0
`
`10
`
`20
`Days
`
`30
`
`40
`
`pg/ml
`
`20
`
`0
`150
`
`100
`
`50
`
`0
`250
`
`200
`
`150
`
`100
`
`50
`
`pg/ml
`
`pg/ml
`
`INF-
`
`TNF-
`
`0
`
`0
`
`10
`
`30
`
`40
`
`20
`20
`Days
`Days
`Figure 2. Patient serum cytokine levels before and after cyclophosphamide and T-cell infusions. Serum cytokines were determined before and after 19-28z⫹ T-cell
`infusion. The first set of patients (CLL-1 to CLL-3) did not receive cyclophosphamide treatment before T-cell infusion, whereas the second set (CLL-5 to CLL-7 and ALL-1)
`received cyclophosphamide before T-cell infusion. The x-axis represents treatment time course with day 0 being the day of T-cell infusion and day ⫺2 being the day of
`cyclophosphamide treatment.
`
`0
`
`0
`
`10
`
`30
`
`40
`
`cyclophosphamide followed 2 days later with T-cell infusion. This
`patient was followed over the next 8 weeks before undergoing an
`allogeneic BM transplantation from a related sibling. Despite T-cell
`and neutrophil recovery, the patient exhibited a persistent B-cell
`
`aplasia in the BM and peripheral blood with only very low level
`CD19⫹ B-cell recovery just before transplantation (Figure 4).
`
`19-28zⴙ autologous T cells rapidly traffic to sites of CD19ⴙ tumor
`
`Table 6. Summary of patient responses
`Patient ID
`Response to T-cell infusions
`
`CLL-1
`CLL-2
`CLL-3
`CLL-4
`CLL-5
`
`CLL-6
`CLL-7
`CLL-8
`ALL-1
`
`No objective response
`No objective response
`No objective response
`Not evaluable
`Marked reduction in lymphadenopathy at 3 months, subsequently
`stable for 6 months
`Progressive disease
`Stable disease, lasting 4 months
`Stable disease, lasting ⬎ 8 weeks
`Persistent B-cell aplasia in BM and peripheral blood
`
`Cryopreserved autopsy tissue samples obtained from patient CLL-4
`were analyzed by IHC for 19-28z⫹ T cells. IHC studies showed
`infiltration of targeted 19-28z⫹ T cells into CLL tumor beds in
`lymph nodes, liver, and BM (Figure 5A), reflecting rapid CAR-
`mediated trafficking of 19-28z⫹ T cells to sites of CD19⫹ tumor.
`
`In vivo persistence of 19-28zⴙ autologous T cells
`
`19-28z⫹ T-cell persistence was measured by IHC of BM samples
`and by RT-PCR and flow cytometry of serial peripheral blood and
`BM aspirate samples. With the use of an anti-idiotypic monoclonal
`Ab specific for the 19-28z CAR, IHC analyses of BM samples from
`the first cohort showed either discrete (CLL-3) or no evidence
`(CLL-1 and CLL-2) of 19-28z⫹ T cells at 1 month (Figure 5C).
`
`UPenn Ex. 2012
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`BLOOD, 3 NOVEMBER 2011 䡠 VOLUME 118, NUMBER 18
`
`AUTOLOGOUS CD19-TARGETED T CELLS IN CLL/ALL
`
`4823
`
`Figure 3. Marked reduction of peripheral lymphadenopathy was observed after treatment with autologous 19-28zⴙ T cells. Representative computed tomography
`scan images of patient CLL-5 before treatment and from 4 weeks and 14 weeks after treatment show mild increase in axillary (top) and pelvic lymphadenopathy (bottom) at
`4 weeks but regression of lymphadenopathy at 14 weeks after therapy with modified T cells.
`
`However, in the second cohort, further treated with cyclophosph-
`amide conditioning, CLL-7 exhibited a significant number of
`retained 19-28z⫹ T cells within the BM ⱕ 6 weeks after T-cell
`infusion (Figure 5B-C). Between 1 and 0.02 vector copies per
`100 cells could also be detected by RT-PCR 2 and 3 weeks after
`infusion, respectively, in freshly obtained BM aspirates derived
`from the same patient. Similarly, patient ALL-1 exhibited 19-28z⫹
`T cells in the BM at 5 weeks after therapy consistent with the
`B-cell aplasia observed after T-cell infusion (Figure 5B-C).
`To overcome the high tumor burden background in most of the
`patients with CLL in postinfusion peripheral blood samples, T cells
`were selected and expanded with Dynabeads before analysis. RT-PCR
`(Figure 6A) and flow cytometry analyses (Figure 6B) of expanded
`peripheral blood samples showed an enhanced persistence of
`modified T cells in patients treated with prior cyclophosphamide
`(CLL-4 to CLL-8, ALL-1), despite a lower dose of infused 19-28z⫹
`T cells, compared with the first cohort of patients treated with
`19-28z⫹ T cells alone (CLL-1 to CLL-3).
`
`Persistent 19-28zⴙ T cells retrieved from patient peripheral
`blood samples proliferate in response to CD19 and are
`cytotoxic
`
`To assess the functional status of persisting 19-28z⫹ peripheral
`blood T cells, we investigated whether these cells could proliferate
`on coculture with CD19-expressing AAPCs (3T3-CD19-CD80).11,30
`Peripheral blood T cells collected 8 days after T-cell infusion from
`patient ALL-1 indeed expanded on 3T3-CD19-CD80 fibroblasts,
`with 19-28z⫹ T cells reaching 32.9% of the CD3⫹ T-cell fraction
`after 7 days in culture (Figure 6B). In contrast, the same blood
`sample expanded by nonspecific activation with the use of anti-CD3/
`CD28 beads showed that 4.2% of the CD3⫹ T-cell fraction
`expressed the 19-28z⫹ CAR. Confirming and extending these
`findings, quantitative measurements of vector sequences showed
`42.1

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