throbber
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Cancer Therapy: Preclinical
`
`Genetically Targeted T Cells Eradicate Systemic Acute
`Lymphoblastic Leukemia Xenografts
`RenierJ. Brentjens,1Elmer Santos,2 Yan Nikhamin,1Raymond Yeh,1Maiko Matsushita,1Krista La Perle,3
`Alfonso Quinta¤ s-Cardama,1Steven M. Larson,2 and Michel Sadelain1,4,5
`
`Abstract Purpose: HumanTcells targeted to the B cell ^ specific CD19 antigen through retroviral-mediated
`transfer of a chimeric antigen receptor (CAR), termed 19z1, have shown significant but partial
`in vivo antitumor efficacy in a severe combined immunodeficient (SCID)-Beige systemic human
`acute lymphoblastic leukemia (NALM-6) tumor model. Here, we investigate the etiologies of
`treatment failure in this model and design approaches to enhance the efficacy of this adoptive
`strategy.
`Experimental Design: A panel of modified CD19-targeted CARs designed to deliver combined
`activating and costimulatory signals to theT cell was generated and tested in vitro to identify an
`optimal second-generation CAR. Antitumor efficacy of Tcells expressing this optimal costimula-
`tory CAR,19-28z, was analyzed in mice bearing systemic costimulatory ligand-deficient NALM-6
`tumors.
`Results: Expression of the 19-28z CAR, containing the signaling domain of the CD28 receptor,
`enhanced systemic T-cell antitumor activity when compared with 19z1 in treated mice. A treat-
`ment schedule of 4 weekly T-cell injections, designed to prolong in vivo T-cell function, further
`improved long-term survival. Bioluminescent imaging of tumor in treated mice failed to identify a
`conserved site of tumor relapse, consistent with successful homing by tumor-specific T cells to
`systemic sites of tumor involvement.
`Conclusions: Both in vivo costimulation and repeated administration enhance eradication of
`systemic tumor by genetically targeted T cells. The finding that modifications in CAR design as
`well asT-cell dosing allowed for the complete eradication of systemic disease affects the design
`of clinical trials using this treatment strategy.
`
`The majority of adult B-cell malignancies, including acute
`lymphoblastic leukemia (ALL), chronic lymphocytic leukemia,
`and non – Hodgkin’s lymphoma, are incurable despite currently
`available therapies. For this reason, novel therapeutic strategies
`are needed to treat
`these diseases. Adoptive therapy with
`genetically engineered autologous T cells is one such approach.
`T cells may be modified to target tumor-associated antigens
`
`Authors’ Affiliations: Departments of 1Medicine and 2Radiology; 3Research
`Animal Resource Center; 4Immunology Program; and 5Gene Transfer and Somatic
`Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York,
`New York
`Received 3/23/07; revised 5/17/07; accepted 5/23/07.
`Grant support: CA95152, CA59350, CA08748, CA86438, and CA96945;
`The Alliance for Cancer Gene Therapy (M. Sadelain); The Annual Terry Fox Run for
`Cancer Research (New York, NY) organized by the Canada Club of New York,
`William H. Goodwin and Alice Goodwin, and the Commonwealth Cancer
`Foundation for Research and the Experimental Therapeutics Center of Memorial
`Sloan Kettering Cancer Center (R.J. Brentjens and M. Sadelain); Amgen Career
`Development Award (R.J. Brentjens); and the Bocina Cancer Research Fund.
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance
`with 18 U.S.C. Section 1734 solely to indicate this fact.
`Requests for reprints: Renier J. Brentjens, Department of Medicine, Memorial
`Sloan Kettering Cancer Center, Box 242, 1275 York Avenue, New York, NY 10021.
`Phone: 212-639-7053; E-mail: brentjer@mskcc.org.
`F 2007 American Association for Cancer Research.
`doi:10.1158/1078-0432.CCR-07-0674
`
`through the introduction of genes encoding artificial T-cell
`receptors, termed chimeric antigen receptors (CAR), specific to
`such antigens (1 – 5).
`CD19 is an attractive target for immune-mediated therapies
`as it is expressed on most B-cell malignancies and normal B
`cells, but not on bone marrow stem cells. We previously
`constructed a ‘‘first-generation’’ CAR, termed 19z1, specific to
`the CD19 antigen. The 19z1 CAR contains a CD19-specific
`murine single-chain fragment length antibody (scFv) fused to
`the extracellular and transmembrane regions of CD8, which, in
`turn, is fused to the intracellular signaling domain of the CD3 ~
`chain. Human T cells retrovirally transduced to express the
`19z1 receptor specifically lyse heterologous and autologous
`CD19+ human tumor cells in vitro (6). A single i.v. injection of
`19z1+ T cells into SCID-Beige mice bearing established systemic
`Raji tumor, a human Burkitt lymphoma tumor cell line that
`expresses the costimulatory ligands CD80 and CD86, success-
`fully eradicates disease in 50% of mice (6). Unfortunately, a
`similar therapy fails to fully eradicate systemic NALM-6 tumor,
`a human pre-B cell ALL cell line that lacks expression of both
`CD80 and CD86. However, 19z1+ T-cell therapy of SCID-Beige
`mice bearing systemic NALM-6 tumors genetically engineered
`to express CD80 (NALM-6/CD80) enhanced long-term survival
`and resulted in complete NALM-6/CD80 tumor eradication in
`40% of mice (6). Although these data show a role for in vivo
`costimulation in tumor eradication by genetically modified
`
`Clin Cancer Res 2007;13(18) September 15, 2007
`
`5426
`
`www.aacrjournals.org
`
`UPenn Ex. 2041
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Eradication of Systemic ALL with CD19-Targeted T Cells
`
`elsewhere (8). For ex vivo expansion studies and cytokine release assays,
`transduced T cells were cocultured for 7 days after
`retroviral
`transduction in 24-well tissue culture plates (Falcon, Becton Dickinson)
`with confluent NIH 3T3 AAPCs in RPMI medium supplemented with
`10% FCS, L-glutamine, streptomycin, and penicillin, with no added
`cytokines. For in vivo experiments, transduced T cells were injected after
`for the Pz1+ T cell control,
`expansion on 3T3(CD19/CD80) or,
`3T3[prostate-specific membrane antigen (PSMA)/CD80] AAPCs
`in RPMI medium as above, supplemented with 20 IU IL-2/mL and
`10 ng/mL IL-15. For mice treated with multiple injections of modified
`T cells for 4 weeks, CAR+ T cells were generated by weekly restimulation
`on AAPCs as described above.
`Western blot analysis. Western blot analysis of T-cell lysates under
`reducing conditions with 0.1 mol/L DTT (Sigma) was done as
`previously described (10). Briefly, transduced T cells were washed in
`PBS and resuspended in radioimmunoprecipitation assay buffer
`(Boston Bioproducts) with mini complete protease inhibitor as per
`the manufacturer’s instructions (Roche Diagnostics). Resulting proteins
`were separated on 12% SDS-PAGE mini gels (Bio-Rad) after the
`
`addition of 6 reducing loading buffer (Boston Bioproducts) and
`heating at 100jC for 10 min. Separated proteins were subsequently
`transferred to Immobilon membranes and probed using an anti-human
`CD3 ~ chain monoclonal antibody (BD Biosciences). Antibody binding
`was detected by probing the blot with goat anti-mouse horse radish
`peroxidase – conjugated antibody followed by luminescent detection
`using Western Lighting Chemiluminescence Reagent Plus (Perkin-Elmer
`Life Sciences) as per the manufacturer’s instructions.
`Cytotoxicity assays. We determined the cytotoxic activity of trans-
`duced T cells by standard 51Cr release assays as described elsewhere (8).
`Briefly, transduced T cells were assessed by fluorescence-activated cell
`sorting analysis for CAR expression as well as CD4:CD8 ratio on day 4
`after transduction. NALM-6 tumor cells were labeled with 51Cr for 1 h at
`37jC, washed with RPMI medium supplemented with 10% FCS, and
`resuspended in the same medium at a concentration of 1  105 tumor
`
`cells/mL. Transduced T cells were added to tumor cells at varying
`effector to target cell ratios in 96-well tissue culture plates in a final
`volume of 200 AL, and incubated for 4 h at 37jC. Thereafter, 30 AL of
`supernatant from each well was analyzed using Lumaplate-96 micro-
`plates (Packard Bioscience) by a Top Count NXT microplate scintilla-
`tion counter (Packard Bioscience). Effector cell number in all assays was
`calculated based on the total number of CD8+ CAR+ T cells.
`Cytokine detection assays. Cytokine assays were done per manufac-
`turer’s specifications using the multiplex Human Cytokine Detection
`System (Upstate, Inc.). Luminescence was assessed using the Luminex
`IS100 system and analyzed for cytokine concentration using IS 2.2
`software (Luminex Corp.).
`Flow cytometry. We did flow cytometry using a FACScan cytometer
`with Cellquest software (BD Biosciences). Cells were labeled with either
`phycoerythrin-conjugated, CAR-specific polyclonal goat antibody (Cal-
`tag Laboratories) or phycoerythrin-labeled anti-human CD8 and FITC-
`labeled anti-human CD4 monoclonal antibodies (Caltag Laboratories).
`Retroviral transduction of NALM-6 tumor cells with GFP-FFLuc. The
`GFP-FFLuc gene (Clontech Laboratories) was subcloned into the SFG
`retroviral vector. VSV-G pseudotyped retroviral supernatants derived
`from gpg29 fibroblasts transduced with the resulting SFG (GFP-FFLuc)
`plasmid were used to transduce NALM-6 tumor cells as described
`elsewhere (10). Resulting tumor cells were sorted by fluorescence-
`activated cell sorting for GFP expression.
`In vivo SCID-Beige mouse tumor models. We inoculated 8- to 12-
`week-old FOX CHASE C.B-17 (SCID-Beige) mice (Taconic) with tumor
`cells by tail vein injection. We subsequently treated mice by tail vein
`injection with transduced T cells. In the Raji tumor model, mice were
`
`injected by tail vein with 5  105 Raji tumor cells on day 1, and on day
`6 were treated with a single i.v. dose of 1  107 CAR+ T cells. In the
`vein on day 1 with 1  106 NALM-6 tumor cells, and on days 2 to
`4 were injected i.v. with 1  107 CAR+ T cells daily. In the weekly
`
`NALM-6 upfront treatment tumor model, mice were injected by tail
`
`tumor-targeted T cells, they neither fully explain the etiologies
`of treatment failure in a majority of treated mice nor provide a
`strategy to overcome these limitations.
`To further investigate the in vivo limitations of this adoptive
`T-cell strategy, we chose to pursue the treatment of NALM-6
`tumors in SCID-Beige mice due to the fact that the tumor in
`this model has several features that mimic B-cell ALL disease in
`human subjects: First, the disease is systemic; second, similar to
`the clinical setting, NALM-6 tumor displays an anatomic
`disease pattern that includes involvement of the bone marrow
`and central nervous system (CNS; ref. 7); and third, the NALM-
`6 tumor cell line fails to express costimulatory ligands as do
`most B-cell leukemias, including B-cell ALL. In this report, we
`use this NALM-6 tumor model to address the limitation of
`failed in vivo T-cell costimulation, and further investigate T-cell
`persistence and homing as potential etiologies of treatment
`failure. We found that both in vivo costimulation as well as
`repeated T-cell administration were critical to the complete
`eradication of NALM-6 tumor in SCID-Beige mice. Subsequent
`modifications in our treatment strategy based on these findings
`resulted in a markedly improved rate of complete tumor
`eradication in treated mice.
`To our knowledge, this is the first report demonstrating
`complete eradication of a systemic human tumor lacking
`costimulatory ligands using genetically targeted T cells. Further-
`more, complete eradication is achieved in the absence of further
`in vivo therapy, including prior chemotherapy or subsequent
`cytokine support. These optimized treatment strategies are likely
`to be applicable to future human trials enrolling patients with
`B-cell malignancies, including B-cell ALL.
`
`Materials and Methods
`
`Cell lines and T cells. Raji and NALM-6 tumor cell lines were
`cultured in RPMI 1640 (Life Technologies) supplemented with 10%
`heat-inactivated FCS, nonessential amino acids, HEPES buffer, pyru-
`vate, and BME (Life Technologies). PG-13 and gpg29 retroviral
`producer cell
`lines were cultured in DMEM (Life Technologies)
`supplemented with 10% FCS, and NIH-3T3 artificial antigen-presenting
`cells (AAPC), described previously (6), were cultured in DMEM
`supplemented with 10% heat-inactivated donor calf serum. All media
`were supplemented with 2 mmol/L L-glutamine (Life Technologies),
`100 units/mL penicillin, and 100 Ag/mL streptomycin (Life Technol-
`ogies). Where indicated, medium was supplemented with 10 ng/mL
`interleukin 15 (IL-15; R&D Systems).
`Construction of second-generation CAR fusion genes. Construction of
`the 19z1 and Pz1 scFv-~ chain fusion proteins have been previously
`published (6, 8). The resulting fusion genes were cloned into the SFG
`retroviral vector (9). VSV-G pseudotyped retroviral supernatants derived
`from transduced gpg29 fibroblasts were used to construct stable PG-13
`gibbon ape leukemia virus (GaLV) envelope-pseudotyped retroviral
`producing cell lines using polybrene (Sigma) as described previously
`(8). All second-generation fusion receptors contain the scFv derived
`from 19z1. Human CD28, DAP10, 4-1BB, and OX40 coding regions
`were PCR amplified from a human activated T-cell cDNA library, and
`subcloned into the TopoTA PCR 2.1 cloning vector (Invitrogen). All
`receptor constructs were generated using overlapping PCR. The
`resulting cassettes were designed to facilitate the exchange of the
`transmembrane and signaling domains of the 19z1 construct by NotI/
`BamHI restriction sites encoded in flanking primers.
`Retroviral
`transduction and expansion of human T lymphocytes.
`Retroviral
`transduction of healthy donor T cells, obtained under
`institutional review board – approved protocol 90-095,
`is described
`
`www.aacrjournals.org
`
`5427
`
`Clin Cancer Res 2007;13(18) September 15, 2007
`
`UPenn Ex. 2041
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Cancer Therapy: Preclinical
`
`treatment model, mice were inoculated with NALM-6 tumor cells on
`
`day 1, and subsequently treated with i.v. injections of 1  107 CAR+
`
`T cells on days 2, 8, 15, and 22. In all experiments, mice that developed
`hind limb paralysis or decreased response to stimuli were sacrificed by
`CO2 asphyxiation. All murine studies were done in the context of
`an Institutional Animal Care and Use Committee – approved protocol
`(no. 00-05-065).
`In vivo bioluminescence of NALM-6 tumors. Bioluminescence
`imaging was done using Xenogen IVIS Imaging System (Xenogen) with
`Living Image software (Xenogen) for acquisition of imaging data sets.
`injection with 150 mg/kg D-luciferin
`Mice were infused by i.p.
`(Xenogen) suspended in 200 AL PBS. Ten minutes later, mice were
`imaged while under 2% isoflurane anesthesia. Image acquisition was
`done on a 15- or 25-cm field of view at medium binning level for
`0.5- to 3-min exposure time. Both dorsal and ventral views were
`obtained on all animals. Tumor bulk, as determined by IVIS imaging,
`was assessed as described previously (11).
`Histologic analysis of mouse tissue sections. Mouse tissues were fixed
`in 10% buffered formalin phosphate (Fisher Scientific). Osseous
`samples (head with brain, vertebral column with spinal cord, and
`hind limbs) were fixed and decalcified in SurgiPath Decalcifier I
`(SurgiPath Medical Industries) as per the manufacturer’s specifications.
`All tissues were processed by routine methods and embedded in
`paraffin wax. Five-micrometer sections were stained with H&E (Poly
`Scientific).
`Statistics. Statistical analysis of survival data by log-rank analysis
`was obtained using GB-STAT software (Dynamic Microsystems).
`
`Results
`
`Construction of second-generation costimulatory CARs. We
`have previously shown that T cells which express the first-
`generation 19z1 CAR successfully eradicate systemic CD80/
`CD86+ Raji tumor in SCID-Beige mice. However, in the same
`report, we further show the inability of 19z1+ T cells to fully
`eradicate systemic NALM-6 tumors, which fail to express the
`costimulatory CD80 and CD86 ligands. The genetic modifica-
`tion of NALM-6 tumors to express CD80 allowed for the
`complete eradication of tumor in a significant number of
`treated mice (6) consistent with the notion that in vivo T-cell
`costimulation enhances the antitumor efficacy of
`tumor-
`specific T cells.
`Because most B-cell tumors fail to express costimulatory
`ligands, we addressed this limitation of our treatment strategy
`by constructing a series of second-generation CARs designed to
`deliver an additional costimulatory signal in the absence of
`exogenous costimulatory ligand by inserting the transmem-
`brane and cytoplasmic signaling domains of the CD28, DAP10,
`and 4-1BB costimulatory receptors into the 19z1 CAR (Fig. 1A).
`Alternative DAP-10 – and 4-1BB – containing receptors, as
`well as the OX-40 – containing CAR, were designed to contain
`the CD8 transmembrane domain from the original 19z1
`
`Fig. 1. T-cell expression of second-generation CARs. A, pictorial representations of 19z1with second-generation CAR genes demonstrating the genetic fragments used to
`generate the costimulatory CARs. B, Western blot analysis of Tcells retrovirally transduced with CAR genes under reducing conditions. Membranes were probed with a
`monoclonal antibody specific to the cytoplasmic domain of the human ~ chain and show the expression of CARs at the expected molecular weights. The nativeT-cell ~ chain
`is indicated. Lane 1, 19z1; lane 2, 19-28z; lane 3, 19(DAPic)z; lane 4, 19z(DAPic); lane 5, 19(DAP)z; lane 6, 19(4-1BBic)z; lane 7, 19(4-1BB)z; and lane 8, 19(OX-40ic)z.
`C, NALM-6 tumor lysis by CAR+ Tcells was assessed by standard 4 h 51Cr release assays. All Tcells expressing CD19-targeted second-generation CARs lysed target tumor
`cells equally well when compared withTcells expressing the first-generation 19z1CAR. Control Tcells expressing the irrelevant Pz1CAR did not significantly lyse NALM-6
`tumors. Effector to target ratios (E:T Ratio) representTcells normalized to the CD8+ CAR+ T-cell fraction. Data represent one of three different experiments using three different
`healthy donorT-cell populations with similar results.
`
`Clin Cancer Res 2007;13(18) September 15, 2007
`
`5428
`
`www.aacrjournals.org
`
`UPenn Ex. 2041
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Eradication of Systemic ALL with CD19-Targeted T Cells
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Fig. 2. In vitro T-cell costimulation of CAR+ Tcells as assessed by proliferation and cytokine secretion after coculture on NIH 3T3 fibroblast AAPCs. A, CAR-transduced
`Tcells, normalized to the CAR+ T-cell fraction, were cocultured on AAPC monolayers either without exogenous costimulation [3T3(CD19); left], or with exogenous
`costimulation [3T3(CD19/CD80); right]. On day 1, 5  105 CAR+ Tcells were cocultured on AAPC monolayers in 24-well tissue culture plates in cytokine-free medium.
`On days 4 and 7, total viableT-cell counts were obtained by trypan blue exclusion assays, and FACS analysis was done on these samples to calculate the total number of
`CAR+ Tcells.Whereas 19-28z, 19(4-1BB)z, and 19(4-1BBic)z transduced Tcells expanded by day 4, only 19-28z+ Tcells continued to expand in the absence of exogenous
`CD80 costimulation (left). Control Pz1+ Tcells failed to expand in either setting. B and C, equal numbers of CAR+ Tcells were incubated in the absence of additional cytokines
`AAPC monolayers. Cell-free tissue culture supernatants were analyzed at 24 h after coculture for the presence of cytokines: lane 1, 19z1; lane 2, 19-28z; lane 3, Pz1;
`lane 4, 19(DAP)z; lane 5, 19(DAPic)z; lane 6, 19z(DAPic); lane 7, 19(OX-40ic)z; lane 8, 19(4-1BB)z; and lane 9, 19(4-1BBic)z. OnlyTcells that expressed the 19-28z CAR
`secreted significant levels of IL-2 (B) and IFNg (C) in the absence of exogenous CD80.These data are representative of one of three different experiments using three different
`healthy donorT-cell populations, all of which showed the same proliferation and cytokine secretion patterns.
`
`construct to assess whether this CAR design could enhance
`costimulatory signaling. Finally, we also assessed whether
`placing the DAP-10 signaling domain either proximal or distal
`to the ~ chain signaling domain improved CAR function.
`T cells from healthy human donors were retrovirally
`transduced to express these CAR constructs. T-cell transduction
`efficiency, as assessed by flow cytometric analysis, ranged from
`50% to 80% (data not shown). Western blot analysis of
`transduced T cells, normalized to the CAR+ fraction, showed
`comparable levels of CAR proteins present at the predicted
`molecular weights (Fig. 1B).
`To verify that second-generation CARs when expressed in
`human T cells retained the ability to lyse CD19+ tumor cells
`
`in vitro, we conducted standard 4 h 51Cr release assays using
`transduced healthy donor T cells targeting 51Cr-labeled CD19+
`NALM-6 tumor cells (Fig. 1C). Effector to target ratio between
`different CAR constructs was normalized to the CD8+ CAR+
`T-cell population. All tested CARs, with the exception of the
`Pz1+ T-cell control, specific to the PSMA, were able to mediate
`tumor cell lysis equally well when compared with the first-
`generation 19z1 CAR.
`Characterization of second-generation CAR costimulatory
`function. We have previously generated a series of AAPCs
`derived from NIH-3T3 murine fibroblasts genetically engi-
`neered to express either CD19 alone [3T3(CD19)] or both
`CD19 and CD80 [3T3(CD19/CD80); ref. 6]. When cocultured
`
`www.aacrjournals.org
`
`5429
`
`Clin Cancer Res 2007;13(18) September 15, 2007
`
`UPenn Ex. 2041
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Cancer Therapy: Preclinical
`
`with CD19-specific CAR+ T cells, the former [3T3(CD19)] pro-
`vides T cells with a CAR-mediated activation signal (signal 1)
`alone, whereas the latter [3T3(CD19/CD80)] provides T cells
`with signal 1 as well as an exogenous costimulatory signal
`(signal 2) mediated through CD80 on the AAPC binding to
`CD28 on the T cell.
`transduced
`To assess CAR-mediated T-cell proliferation,
`T cells were cocultured on both 3T3(CD19) and 3T3(CD19/
`CD80) AAPCs in the absence of exogenous cytokine. CAR+
`T-cell number was determined after coculture on 3T3(CD19)
`and 3T3(CD19/CD80) AAPCs (Fig. 2A) on days 4 and 7.
`Following coculture on 3T3(CD19) AAPCs, only T cells
`transduced with the 19-28z, 19(4-1BB)z, and 19(4-1BBic)z
`CARs expanded after 4 days of coculture, whereas at day 7, only
`T cells transduced with the 19-28z CAR had undergone
`significant proliferation (10-fold expansion) consistent with
`CAR-mediated T-cell costimulation. All other constructs failed
`to promote T-cell proliferation in the absence of exogenous
`costimulation. Significantly, 19-28z CAR+ T cells proliferated
`equally well on either 3T3(CD19) or 3T3(CD19/CD80) AAPCs,
`and proliferated equally well when compared with 19z1+ T cells
`in the setting of 3T3(CD19/CD80) AAPC stimulation.
`We next assessed CAR+ T-cell secretion of IL-2 and IFN-g as
`surrogate makers of costimulation. Equal numbers of CAR+
`T cells with similar CD4:CD8 ratios were cocultured on
`3T3(CD19) and 3T3(CD19/CD80) AAPCs in cytokine-free
`medium after initial T-cell transduction. At 24 h after T-cell
`coculture on AAPCs, tissue culture supernatants were analyzed
`for the presence of IL-2 and IFN-g. Coculture of 19-28z+ T cells
`on 3T3(CD19) AAPCs resulted in significantly elevated levels of
`IL-2 (Fig. 2B) and IFN-g (Fig. 2C) when compared with the
`first-generation 19z1 CAR as well as the other six tested second-
`generation CARs. These data further confirm the ability of the
`19-28z CAR to elicit a costimulatory signal independent of
`exogenous costimulatory ligand. Significantly, 4-1BB and DAP-
`10 constructs containing either the native or CD8 transmem-
`brane domains showed no significant differences in function,
`
`and in the setting of the DAP-10 constructs, no differences in
`function were observed when the signaling domain was placed
`either proximal or distal to the ~ chain.
`In vivo antitumor activity of alternative costimulatory
`CARs. To define the in vivo costimulatory activity of the
`19-28z CAR, we initially compared the antitumor activity of
`19z1+ and 19-28z+ T cells in a previously established systemic
`Raji tumor model. Because both 19z1+ T cells and 19-28z+
`T cells are costimulated in vivo by CD80/CD86+ Raji tumor
`cells, we predicted and observed equal long-term survival (50%)
`in both treatment groups (Fig. 3A). We next compared the same
`treatment groups (19z1 versus 19-28z) in mice bearing systemic
`NALM-6 tumor, which fails to express the CD80 and CD86
`costimulatory ligands. We observed improved antitumor
`efficacy in the 19-28z treatment group (Fig. 3B). However, the
`improved long-term overall survival was modest (0 of 15 19z1+
`T cell – treated mice versus 3 of 17 or 18% of 19-28z+ T cell –
`treated mice; P < 0.03). Treatment with 19(4-1BB)z+ and
`19(4-1BBic)z+ T cells enhanced survival when compared with
`mice treated with the control Pz1+ T cells, but overall survival
`was similar to 19z1+ T cell – treated mice, with no long-term
`surviving mice (data not shown). Fluorescence-activated cell
`sorting analysis of single-cell suspensions of tissues derived
`from mice that failed 19-28z+ T cell treatment showed persistent
`expression of CD19 on the tumor cells, ruling out the possibility
`that down-regulation of the CD19 target antigen was a source of
`treatment failure (data not shown).
`Repeated administration of CD19-targeted T cells enhances
`complete NALM-6 tumor eradication. Using bioluminescent
`imaging of treated SCID-Beige mice bearing NALM-6(GFP-
`FFLuc) tumors, we found a 10- to 14-day delay of tumor
`progression after 19z1+ T-cell therapy when compared with
`Pz1+ T-cell
`therapy (Fig. 4A-B). A similar delay of tumor
`progression was seen after 19-28z+ T-cell therapy in mice with
`relapsed disease (data not shown). Fluorescence-activated cell
`sorting analysis of single-cell suspensions derived from tissues
`of 19-28z+ T cell – treated tumor-bearing mice confirmed a
`
`Fig. 3. In vivo analysis of CD19-specific CAR+ Tcells in SCID-Beige mice bearing systemic human CD19+ tumors. A, treatment with 19z1+ or 19-28z+ Tcells eradicated
`systemic Raji tumors in SCID-Beige mice equally well. Briefly, mice were injected by tail vein with 5  105 Raji tumor cells on day 1. Mice were subsequently treated with a
`single dose of 1 107 CAR+ Tcells by tail vein injection on day 6. Mice treated with either 19z1+ or 19-28z+ Tcells eradicated tumor equally well with an overall 50% long-term
`survival in both treatment groups. B, treatment of NALM-6 bearing SCID-Beige mice with 19-28z+ Tcells (1 107 CAR+ Tcells injected daily  3 d) statistically enhanced
`survival when compared with similar treatment with 19z1+ Tcells (18% versus 0% long-term survival; P < 0.03).
`
`Clin Cancer Res 2007;13(18) September 15, 2007
`
`5430
`
`www.aacrjournals.org
`
`UPenn Ex. 2041
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Eradication of Systemic ALL with CD19-Targeted T Cells
`
`Fig. 4. Enhanced antitumor activity through weekly
`CD19-targeted T-cell infusions. A, treatment of
`NALM-6 tumor-bearing mice with daily targeted
`T-cell injections on days 1to 3 after tumor cell
`injection delays tumor growth for 10 to 14 d as
`assessed by analysis of IVIS images of mice treated
`with either 19z1+ Tcells or the control Pz1+ Tcells.
`NALM-6 (GFP-Luc) tumor burden was assessed
`based on ventral mouse images in photons per
`second emitted after luciferin injection. Similar
`data were obtained in mice treated with 19-28z+
`Tcell ^ treated mice (not shown). B, representative
`dorsal and ventral images of NALM-6 (GFP-Luc) ^
`bearing mice obtained weekly afterT-cell therapy
`shows delayed tumor growth in 19z1+ Tcell ^ treated
`mice when compared with the Pz1+ T-cell control
`mice. C, weekly 19-28z+ Tcell tail vein injection for
`4 wks on days 2, 9, 16, and 22, after NALM-6 tumor
`injection on day 1, enhanced long-term survival of
`treated mice >4-fold (44% versus 10%) when
`compared with similar treatment with either 19z1+
`Tcells (P < 0.002) or the control Pz1+ Tcell ^ treated
`mice. Results represent pooled data from two
`separate experiments.
`
`short in vivo T-cell survival (data not shown), consistent with
`previously published data demonstrating limited in vivo
`survival of human T cells in an immunocompromised murine
`host (11 – 14).
`To assess whether enhanced in vivo T-cell persistence
`promoted long-term survival, we generated a sustained in vivo
`population of tumor-targeted T cells in NALM-6 tumor bearing
`
`SCID-Beige mice by injecting CAR+ T cells weekly for 4 weeks.
`The incidence of long-term survival with 19-28z+ T-cell therapy
`increased from 18% as seen in the upfront daily 3 treatment
`
`schedule to 44% in the weekly treatment schedule (Figs. 3B
`and 4C, respectively). Consistent with an additive effect of
`in vivo T-cell persistence and costimulation, we found superior
`antitumor activity of 19-28z+ T cells, when compared with
`
`www.aacrjournals.org
`
`5431
`
`Clin Cancer Res 2007;13(18) September 15, 2007
`
`UPenn Ex. 2041
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/13/18/5426/1972030/5426.pdf by guest on 17 July 2022
`
`Cancer Therapy: Preclinical
`
`19z1+ T cells, as shown by 44% versus 10% long-term survival
`(P < 0.002), using this weekly T-cell dosing schedule (Fig. 4C).
`CD19-targeted T cells eradicate NALM-6 tumor cells at
`disparate anatomic sites in SCID-Beige mice after adoptive
`T-cell therapy. Despite improved survival with the weekly
`T-cell dosing treatment schedule, 56% of 19-28+ T cell – treated
`mice ultimately failed therapy. To assess whether these mice
`developed persistent or recurrent tumor in consistent anatomic
`sites potentially restricted from T-cell penetration or detection,
`we monitored in vivo NALM-6(GFP-FFLuc) tumor progression
`by weekly bioluminescent (IVIS) imaging in the setting of the
`weekly T-cell treatment regimen. In these studies, all control
`mice treated weekly with Pz1+ T cells showed diffuse tumor
`progression at multiple sites over the treatment period (Fig. 5A).
`By weekly IVIS monitoring of the 19z1+ T cell – treated mice,
`40% (4 of 10) had no detectable disease upon completion of
`therapy (day 28). In the nine mice that developed progressive
`disease in this treatment group, initial disease persistence or
`relapse was noted at distinct anatomic sites in different mice,
`including the bone marrow (n = 2; Fig. 5B), the periodontal
`region (n = 3), the spleen/calvarium (n = 1), the abdomen
`(n = 1), and the CNS/calvarium (n = 1). Overall, 69% (11 of 16)
`of mice treated with weekly 19-28z+ T-cell infusions showed no
`evidence of disease by weekly IVIS imaging upon completion
`
`of therapy. The 56% (9 of 16) of 19-28z+ T cell – treated mice,
`which ultimately failed therapy, likewise showed detectable
`disease at disparate anatomic sites, including the spleen (n = 1;
`Fig. 5C), the CNS/calvarium (n = 2; Fig. 5D), the periodontal
`region (n = 4; Fig. 5E), and the bone marrow (n = 2; Fig. 5F).
`Significantly, persistent disease in the periodontal region,
`despite ongoing T-cell therapy, was noted in 30% of 19z1-
`treated mice, and 25% of 19-28z – treated mice, accounting for
`a large majority of the mice with evidence of disease upon
`completion of treatment. The presence of tumor as assessed
`by bioluminescent imaging in 19-28z+ T cell – treated mice was
`subsequently confirmed by histologic analysis, demonstrating
`NALM-6 tumor infiltration in the bone marrow (Fig. 6A-B), the
`spleen (Fig. 6C-D), and the periodontal region (Fig. 6E-F).
`
`Discussion
`
`The successful clinical application of adoptive therapy of
`cancer with genetically targeted T cells requires both a better
`understanding of the in vivo biology of genetically modified
`T cells, as well as innovative means of enhancing the in vivo
`potency of these T cells. In this report, we use the clinically
`relevant NALM-6 systemic murine model of human B-cell ALL,
`which involves the bone marrow and CNS, and lacks
`
`Fig. 5. Bioluminescent imaging of NALM-6(GFP-
`FFLuc) tumor cells after weekly CAR+ T-cell
`treatment for 4 wks.Weekly treatment with Pz1+
`Tcells in mice bearing systemic NALM-6
`(GFP-FFLuc) resulted in a typical diffuse pattern of
`tumor cell expansion involving the bone marrow,
`spleen, liver, lymph nodes, and periodontal

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