throbber
The Journal of Immunology
`
`Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/5/3449/1267196/zim01708003449.pdf by guest on 08 February 2023
`
`Adoptive Transfer of Gene-Modified Primary NK Cells Can
`Specifically Inhibit Tumor Progression In Vivo1
`
`Hollie J. Pegram,* Jacob T. Jackson,* Mark J. Smyth,*† Michael H. Kershaw,2*†
`and Phillip K. Darcy2,3*†
`
`NK cells hold great potential for improving the immunotherapy of cancer. Nevertheless, tumor cells can effectively escape NK
`cell-mediated apoptosis through interaction of MHC molecules with NK cell inhibitory receptors. Thus, to harness NK cell effector
`function against tumors, we used Amaxa gene transfer technology to gene-modify primary mouse NK cells with a chimeric
`single-chain variable fragment (scFv) receptor specific for the human erbB2 tumor-associated Ag. The chimeric receptor was
`composed of the extracellular scFv anti-erbB2 Ab linked to the transmembrane and cytoplasmic CD28 and TCR-␨ signaling
`domains (scFv-CD28-␨). In this study we demonstrated that mouse NK cells gene-modified with this chimera could specifically
`mediate enhanced killing of an erbB2ⴙ MHC class Iⴙ lymphoma in a perforin-dependent manner. Expression of the chimera did
`not interfere with NK cell-mediated cytotoxicity mediated by endogenous NK receptors. Furthermore, adoptive transfer of gene-
`modified NK cells significantly enhanced the survival of RAG mice bearing established i.p. RMA-erbB2ⴙ lymphoma. In summary,
`these data suggest that use of genetically modified NK cells could broaden the scope of cancer immunotherapy for patients. The
`Journal of Immunology, 2008, 181: 3449 –3455.
`
`N atural killer cells comprise 5–10% of PBLs and play an
`
`important role in the body’s first line of defense against
`pathogen invasion and malignant transformation (1, 2).
`Unlike the exquisite Ag specificity observed for T and B lympho-
`cytes, NK cells instead express a number of different activation
`and inhibition receptors which provide a balance of signals that
`dictate their overall response (3). This recognition system used by
`NK cells and the fact that they do not require prior sensitization
`provides a degree of flexibility to rapidly recognize different
`pathogens.
`Several elegant studies have demonstrated that NK cells can
`effectively control tumor growth in mice mediated through release
`of perforin and cytokines (4 – 6). Their importance in anti-tumor
`immunity is further illustrated by the increased incidence of leu-
`kemia in patients with dysfunctional NK cells (7). The observation
`that NK cells can effectively respond to tumor cells exhibiting
`defective or altered MHC class I has made them promising effec-
`tors for immunotherapeutic strategies that target tumor escape
`
`*Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne,
`Victoria, Australia; and †Department of Pathology, University of Melbourne, Mel-
`bourne, Australia
`
`Received for publication April 17, 2008. Accepted for publication July 3, 2008.
`
`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.
`1 This work was supported by a National Health and Medical Research Council Pro-
`gram Grant and a Cancer Council of Victoria research grant. M.H.K. and P.K.D. were
`supported by National Health and Medical Research Council of Australia R.D. Wright
`Research Fellowships. M.J.S. was supported by a National Health and Medical Re-
`search Council Senior Principal Research Fellowship.
`
`H.J.P. performed the research, analyzed the data, and wrote first draft of the paper.
`J.T.J. performed the research. M.J.S. analyzed the data. M.H.K. designed the research
`and analyzed the data. P.K.D. designed the research, analyzed the data, and wrote the
`manuscript.
`2 M.H.K. and P.K.D. contributed equally as senior authors.
`3 Address correspondence and reprint requests to Dr. Phillip K. Darcy and Dr. Mi-
`chael H. Kershaw, Peter MacCallum Cancer Institute, Locked Bag 1, A’Beckett
`Street, Victoria, 8006, Australia. E-mail addresses: phil.darcy@petermac.org and
`michael.kershaw@petermac.org
`
`www.jimmunol.org
`
`variants (8). Nevertheless, tumor cells have developed several
`mechanisms to impede NK cell function, which include the ex-
`pression of ligands that interact with NK cell inhibitory recep-
`tors (9).
`Immunotherapeutic strategies to enhance NK cell anti-tumor ac-
`tivity have included the use of specific cytokines (10) or adoptive
`transfer of autologous ex vivo IL-2-activated lymphokine killer
`cells (LAK4; Ref.11). However, these approaches have only re-
`sulted in moderate success in restricted numbers of patients (12).
`More promising results have been recently achieved in the trans-
`plant setting with the use of allogeneic NK cells against acute
`myeloid leukemia (13, 14). Another emerging approach to address
`the problem of NK cell-mediated inhibition by tumors involves the
`genetic modification of NK cells with chimeric single-chain vari-
`able fragment (scFv) receptors that directly target tumor-associ-
`ated Ags (TAA). This approach has successfully been used to en-
`hance tumor recognition by primary T cells (15–24), and several
`studies have demonstrated specific killing of tumor target cells
`following redirection of NK cell lines (25–27) or primary human
`NK cells (28) with chimeric receptors. Nevertheless, investigation
`of whether these genetically engineered primary NK cells can spe-
`cifically reject tumor in vivo has never been reported and has been
`hampered by lack of an efficient method for expressing transgenes
`in mouse NK cells.
`In this study we have used Amaxa Nucleofector technology, an
`electroporation-based procedure, to genetically engineer primary
`mouse NK cells with an scFv anti-erbB2-CD28-␨ chimeric recep-
`tor. We and others have shown that this novel receptor design
`incorporating both costimulatory CD28 and TCR-␨domains linked
`in the one intracellular domain could optimally trigger activation
`of transduced PBMC after Ag stimulation (15, 21, 29, 30). The
`Amaxa system uses optimized electrical parameters to enhance
`delivery of DNA to the cell nucleus, which increases transfection
`
`4 Abbreviations used in this paper: LAK, lymphokine killer cells; scFv, single-chain
`variable fragment; TAA, tumor-associated Ags; FasL, Fas ligand; WT, wild type.
`
`Copyright © 2008 by The American Association of Immunologists, Inc. 0022-1767/08/$2.00
`
`UPenn Ex. 2067
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3449
`
`

`

`3450
`
`ENHANCED TUMOR INHIBITION BY GENE-MODIFIED NK CELLS
`
`Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/5/3449/1267196/zim01708003449.pdf by guest on 08 February 2023
`
`FIGURE 1. Gene modification of primary murine NK cells with the
`anti-erbB2 chimeric receptor. A, Schematic representation of the scFv-anti-
`erbB2-CD28-␨receptor. The chimeric receptor consisted of the VH and VL
`regions of the anti-erbB2 mAb joined by a flexible linker, a CD8-␣ mem-
`brane-proximal hinge region (MP), and the transmembrane (TM) and cy-
`toplasmic regions of the mouse CD28 signaling chain fused to the intra-
`cellular domain of human TCR-␨. B, The expression of the chimeric scFv
`anti-erbB2 receptor in mouse NK cells was analyzed following staining
`with an anti-tag Ab mAb and PE-labeled sheep anti-mouse Ig (thick line)
`or a secondary PE-conjugated Ab (thin line). C, Expression of GFP in
`transfected (thick line) vs nontransfected (thin line) NK cells was analyzed
`by flow cytometry. Results shown are representative of seven experiments
`performed.
`
`51Cr-release assay. In brief, NK cells were incubated with 1 ⫻ 105 51Cr-
`labeled tumor targets at various E:T ratios in triplicate wells of a 96-well
`round-bottom plate (in 200 ␮l of complete DMEM). The percentage of
`specific release of 51Cr into the supernatant was assessed as described
`previously (31).
`
`Adoptive transfer
`
`The ability of gene-modified NK cells expressing the ␣-erbB2-CD28-␨
`receptor to enhance the survival of tumor bearing mice was investigated in
`the following model. C57BL/6 RAG-1⫺/⫺ mice were injected i.p. with 2 ⫻
`105 RMA-parental or RMA-erbB2 tumor cells. Mice were then treated on
`days 0, 1, days 0, 1, 2, 3 (early model), or days 3, 4 (delayed model) with
`2 ⫻ 106 (per injection) of ␣-erbB2 NK cells or GFP-NK control cells
`delivered i.p. In some experiments gene-modified NK cells were coadmin-
`istered with high dose IL-2 (200,000 IU/ml) injected i.p. on days 0, 1, and
`2. To investigate the persistence of NK cells in vivo, 2 ⫻ 106 donor gene-
`modified NK cells from congenic C57BL/6- ptprca (CD45.1⫹) mice were
`transferred into RMA-erbB2 tumor-bearing RAG-1⫺/⫺ recipient mice
`(CD45.2⫹) cell on days 0 and 1. Three mice at each time point were then
`sacrificed on days 1, 2, and 5 following tumor injection, and spleens were
`harvested and i.p. washes were performed to determine the number of
`CD45.1⫹ cells present.
`
`Statistical analysis
`The Mann-Whitney U test was used for statistical analysis. Values of p ⬍
`0.05 were considered significant.
`
`Results
`Expression of the chimeric anti-erbB2 receptor in primary
`mouse NK cells
`
`The genetic modification of primary mouse NK cells with scFv
`chimeric receptors using retroviral-based transduction methods has
`proven difficult. To address this, we used the Amaxa Nucleofector
`system to gene modify mouse NK cells with the scFv ␣-erbB2-
`CD28-␨chimeric receptor (Fig. 1A). Using this method, high level
`expression of the ␣-erbB2 receptor was achieved in mouse NK
`
`efficiency and gene expression levels. NK cells expressing the chi-
`meric receptor were demonstrated to enhance target cell killing
`following receptor ligation. Furthermore, adoptive transfer of
`scFv-receptor gene-modified NK cells led to significant growth
`inhibition of erbB2⫹ T cell lymphomas in mice. These data sug-
`gest that gene-modified NK cells may have significant potential as
`an effective immunotherapy for cancer.
`
`Materials and Methods
`Cell lines
`
`The C57BL/6 murine lymphoma cell lines RMA and RMA-S are T cell
`lymphomas derived from the Rauscher murine leukemia virus-induced
`RBL-5 cell line (8). The murine melanoma cell line B16-F10 was obtained
`from American Type Culture Collection. The erbB2-expressing cell lines
`RMA-erbB2 and B16-F10-erbB2 were generated by transduction with a
`retroviral vector (murine stem cell vector) encoding the cDNA for human
`erbB2. All cell lines were maintained in complete DMEM medium con-
`taining 10% (v/v) FCS, 2 mM L-glutamine, 100 U/ml penicillin, and 100
`␮g/ml streptomycin (Invitrogen). The murine lymphoma cell line YAC-1
`was maintained in RPMI 1640 medium (Invitrogen), with 10% FCS (v/v),
`2 mM L-glutamine, 0.1 mM non-essential amino acids (Life Technologies),
`1 mM sodium pyruvate (Life Technologies), 100 U/ml penicillin, 100
`␮g/ml streptomycin (Life Technologies), and 5 ⫻ 10⫺5 mM 2ME.
`
`Mice
`C57BL/6, C57BL/6-ptprca, and C57BL/6 RAG-1-deficient (RAG-1⫺/⫺)
`mice were purchased from the Walter and Eliza Hall Institute of Medical
`(C57BL/6-pfp⫺/⫺) and
`Research. C57BL/6 perforin (pfp)-deficient
`C57BL/6 gld (Fas ligand (FasL) mutant) were bred at the Peter MacCallum
`Cancer Centre. All mice were housed in specific pathogen free conditions
`at the Peter MacCallum Cancer Centre and mice 6 –12 wk of age were used
`in all experiments.
`
`Isolation of NK cells
`
`Dissected spleens from C57BL/6 mice were crushed into hypotonic lysis
`buffer and filtered to create a single cell suspension. NK cells were then
`selected using anti-DX5 Microbeads or an NK cell isolation beading kit
`(Miltenyi Biotec) according to manufacturer’s specifications. The cells
`were then grown in RPMI 1640 medium containing 10% (v/v) FCS, 2 mM
`L-glutamine, 5 ⫻ 10⫺5 mM 2ME, 100 U/ml penicillin, 100 ␮g/ml strep-
`tomycin (Life Technologies), 2 mM HEPES, and 1000 IU/ml recombinant
`human IL-2 (Biological Resources Branch Preclinical Repository, National
`Cancer Institute).
`
`Gene modification of NK cells
`
`Seven-day IL-2-activated mouse NK cells were gene modified by electro-
`poration using the Amaxa Nucleofector system (Amaxa Biosystems). In
`brief, NK cells were placed in 0.1 ml electroporation solution with either
`4 ␮g pMAX plasmid DNA encoding the scFv ␣-erbB2-CD28-␨ chimeric
`receptor or GFP. Following electroporation, the cells were placed into 2 ml
`Amaxa recovery medium with 600 IU/ml recombinant human IL-2 for 24 h
`before being used in experiments.
`
`Flow cytometry
`
`Expression of the chimeric scFv receptor on the surface of NK cells was
`determined by indirect immunofluorescence with a primary c-myc tag Ab
`(Cell Signaling Technology), followed by staining with a secondary PE-
`labeled anti-mouse Ig mAb (BD Biosciences). Background fluorescence
`was determined by staining cells with an isotype control Ab followed by a
`secondary PE-conjugated anti-mouse Ig mAb. Direct detection of GFP by
`flow cytometry was examined in transfected vs non-transfected NK cells.
`Phenotyping of cell surface marker expression on NK cells was determined
`by staining cells with allophycocyanin-conjugated Abs specific for NK1.1,
`DX5, CD11b, and CD27 (eBioscience) and biotin-conjugated KLRG1,
`NKG2D (eBioscience), and biotin- conjugated CD94, CD25, and CD69
`(BD Pharmingen). This was the followed by staining with a PerCPCy5.5-
`streptavidin (BD Pharmingen) Ab. MHC class I expression on tumor cells
`was determined by staining cells with a PE-conjugated Ab specific for
`mouse H2kb (BD Biosciences).
`
`Cytotoxicity
`
`The ability of gene-modified mouse NK cells from wild-type (WT) and/or
`gene-targeted mice to specifically kill tumor targets was assessed in a 4 h
`
`UPenn Ex. 2067
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3450
`
`

`

`Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/5/3449/1267196/zim01708003449.pdf by guest on 08 February 2023
`
`3451
`
`RMA-erbB2
`RMA-S-erbB2
`
`*
`
`20:1
`
`10:1
`E:T ratio
`
`5:1
`
`30
`
`25
`
`20
`
`15
`
`10
`
`05
`
`Specificlysis(%)
`
`FIGURE 3. MHC class I inhibition of NK cell cytotoxicity. GFP-NK
`effector cells were used in a 4 h 51Cr release assay against the MHC class
`I⫹ RMA-erbB2 cell line or class I deficient RMA-S-erbB2 tumor cell line.
`GFP-NK cells were able to more effectively kill the RMA-S-erbB2 cell line
`(open squares) compared with the RMA-erbB2 cell line (closed squares; ⴱ,
`p ⬍ 0.05, as determined by a Mann-Whitney U test). Results are repre-
`sentative of three independent experiments.
`
`etry to compare expression of a number of molecules expressed by
`␣-erbB2-NK and control GFP-NK cells, including activation and
`inhibition receptors. In three independently performed experi-
`ments, we observed no difference in the expression of NK cell
`markers NK1.1 or DX5 between GFP-NK and ␣-erbB2-NK cells
`(Fig. 2, A and B). There was also no difference in the level of
`expression of the CD11b marker between transfected NK cells
`(Fig. 2C). In addition, comparable expression of inhibitory recep-
`tors Ly49A, KLRG1, CD94, and the activatory receptor NKG2D,
`was observed between ␣-erbB2-NK and GFP-NK cells (Fig. 2,
`D–F). The levels of expression of the costimulatory receptor CD27
`and activation markers CD69 and CD25 were also expressed at
`similar levels between the transfected NK cell types (Fig. 2, H–J).
`These data indicated that transfection of mouse NK cells with the
`scFv chimeric receptor has not phenotypically altered expression
`of a number of important NK cell-associated markers.
`
`Ag-specific cytotoxicity mediated by anti-erbB2-NK cells
`
`Although NK cells can mediate effective killing of target cells,
`they are often inhibited by recognition of MHC class I molecule.
`Indeed, this is supported by our data demonstrating low killing by
`GFP-NK effector cells of the MHC class I⫹ lymphoma cell line
`RMA-erbB2 compared with the class I-deficient RMA-S-erbB2
`cell line (Fig. 3). To determine whether our gene-modified NK
`cells expressing the ␣-erbB2 chimeric receptor could overcome
`MHC-class I inhibition we assessed their ability to kill RMA cells,
`either expressing the erbB2 TAA (RMA-erbB2) or not. Impor-
`tantly, the level of MHC class I expression on RMA-erbB2 and
`RMA cells was equivalent (Fig. 4). We demonstrated at least a two
`fold increase in the level of killing of RMA-erbB2 cells by
`␣-erbB2-NK cells compared with control GFP-NK cells (Fig. 5A).
`This enhanced killing was erbB2 Ag-specific because ␣-erbB2-NK
`and GFP-NK cells mediated comparable lysis of RMA parental
`cells (Fig. 5B). We next determined whether anti-erbB2-NK cells
`could increase killing of another erbB2⫹ cell line. In this experi-
`ment we demonstrated enhanced killing by anti-erbB2-NK cells of
`a mouse melanoma cell line B16 expressing erbB2 Ag (B16-F10-
`erbB2) compared with control GFP-NK cells (Fig. 5C). Again this
`was erbB2-specific because equivalent killing of parental B16-F10
`cells by anti-erbB2 or GFP-NK cells was observed (Fig. 5D). We
`also showed that expression of the scFv receptor or GFP had no
`impact on the endogenous cytotoxic ability of NK cells. We dem-
`onstrated comparable cytotoxicity of a NK cell-sensitive target cell
`line, YAC-1, by either non-transfected 7-day IL-2-activated NK
`
`The Journal of Immunology
`
`A
`
`NK1.1
`
`B
`
`DX5
`
`C
`
`C 11bD
`
`D
`
`Ly49A
`
`E
`
`KLRG1
`
`F
`
`NKG2D
`
`G
`
`CD94
`
`H
`
`CD27
`
`I
`
`CD69
`
`J
`
`CD25
`
`100
`
`100
`101
`102
`103
`104
`101
`102
`103
`Mean fluorescence intensity
`
`104
`
`120
`
`90
`
`60
`
`30
`
`0
`120
`
`90
`
`60
`
`30
`
`0
`120
`
`90
`
`60
`
`30
`
`0
`120
`
`90
`
`60
`
`30
`
`0
`120
`
`90
`
`60
`
`30
`
`0
`
`Relativecellcount
`
`FIGURE 2. Phenotypic characterization of gene-modified primary
`mouse NK cells. The surface expression of various NK cell markers, ac-
`tivation markers, and activation/inhibition receptors was analyzed by flow
`cytometry following staining with appropriate Abs. Cells used for analysis
`were gated on either anti-tag positive cells (representing anti-erbB2 recep-
`tor expressing cells; thick line) or gated on GFP positive cells (representing
`control NK cells; thin line), or on unstained anti-erbB2-NK cells (dotted
`line). There was no significant difference in expression of the following
`molecules between anti-erbB2 or GFP transfected NK cells; NK1.1 (A),
`DX5 (B), CD11b (C), Ly49A (D), KLRG1 (E), NKG2D (F), CD94 (G),
`CD27 (H), CD69 (I), or CD25 (J). Results shown are representative of
`three independent experiments.
`
`cells following staining with a c-myc tag mAb specifically recog-
`nizing a c-myc tag epitope incorporated into the extracellular do-
`main of the chimeric receptor (46 ⫾ 10%, n ⫽ 7; Fig. 1B). Equiv-
`alent levels of expression of autonomous GFP were also observed
`in control mouse NK cells (49 ⫾ 11%, n ⫽ 7; Fig. 1C). Impor-
`tantly, the transfected NK cell populations were TCR␤ negative
`(data not shown). Cell viability ranged between 60 and 90% fol-
`lowing electroporation.
`
`Phenotypic characterization of gene-modified primary mouse
`NK cells
`
`We next investigated whether expression of the chimeric scFv re-
`ceptor had any affect on NK cell phenotype. We used flow cytom-
`
`UPenn Ex. 2067
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3451
`
`

`

`ENHANCED TUMOR INHIBITION BY GENE-MODIFIED NK CELLS
`
`Unstained
`RMA-erbB2
`RMA-parental
`
`100
`
`75
`
`50
`
`25
`
`3452
`
`Relativecellcount
`
`Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/5/3449/1267196/zim01708003449.pdf by guest on 08 February 2023
`
`**
`
`*
`
`10:1
`
`5:1
`
`1:1
`
`WT NK
`gld NK
`pfp NK
`
`10:1
`
`5:1
`
`1:1
`
`B
`60
`
`40
`
`20
`
`0
`
`D
`60
`
`40
`
`20
`
`10:1
`
`5:1
`
`1:1
`
`0
`
`10:1
`E:T Ratio
`
`5:1
`
`1:1
`
`A
`60
`
`40
`
`20
`
`0
`
`C
`60
`
`40
`
`20
`
`0
`
`Specificlysis(%)
`
`FIGURE 6. Cytotoxicity of erbB2⫹ tumor cells by anti-erbB2 NK cells
`is perforin-dependent. Gene-modified NK cells derived from WT, perforin-
`deficient (pfp⫺/⫺), or gld (Fas ligand mutant) mice were used in a 4 h 51Cr
`release assay. A, Killing of RMA-erbB2 target cells by anti-erbB2 NK cells
`derived from pfp⫺/⫺, but not WT or gld mice, was completely abrogated.
`Equivalent background killing of RMA-erbB2 tumor cells (B) and RMA
`parental cells (C) by GFP-NK cells or RMA-parental cells by ␣-erbB2 NK
`cells (D) derived from either WT or gld mice. (ⴱ, p ⬍ 0.05, ⴱⴱ, p ⬎ 0.05
`as determined by a Mann-Whitney U test). Results shown are representa-
`tive of two independent experiments.
`
`gld (mutant FasL) mice. Importantly, the level of expression of the
`scFv chimeric receptor was comparable in NK cells derived from
`WT and gene-targeted mice (data not shown). It is also important
`to note that previous studies have shown that other functional path-
`ways of NK cells from perforin-deficient mice (i.e., FasL-mediated
`killing) are intact (33). In cytotoxicity assays, we demonstrated no
`killing of RMA-erbB2 target cells by ␣-erbB2 NK cells derived
`from pfp⫺/⫺ mice (Fig. 6A). In contrast, the sensitivity of RMA-
`erbB2 cells to ␣-erbB2 NK cells derived from gld mice or WT
`mice was similar (Fig. 6A). As further specificity controls we ob-
`served comparable background killing of RMA-erbB2 by GFP-NK
`cells and RMA-parental cells by either ␣-erbB2 or GFP-NK cells
`derived from WT and gld mice (Fig. 6, B–D). These data demon-
`strated that gene-modified primary mouse NK cells mediated Ag-
`specific cytotoxicity through a perforin-dependent mechanism.
`
`Ag-specific inhibition of tumor growth mediated by
`anti-erbB2-NK cells
`
`We next assessed the ability of gene-modified mouse NK cells
`expressing the ␣-erbB2 chimeric receptor to mediate Ag-specific
`inhibition of tumor growth in vivo. Tumor cells (RMA-parental or
`RMA-erbB2) were injected i.p. into RAG-1⫺/⫺ mice that then
`received early transfer (days 0, 1) or delayed transfer (days 3, 4) of
`2 ⫻ 106 gene-modified NK cells (␣-erbB2-NK or GFP-NK cells).
`In these experiments, we demonstrated significantly increased sur-
`vival of mice with RMA-erbB2 tumor that received ␣-erbB2 gene-
`modified NK cells delivered at early or at later time points (Fig. 7,
`A and B). This effect was Ag-specific because there was no sig-
`nificant increase in survival of mice with RMA-erbB2 that re-
`ceived control GFP-NK cells. Furthermore, ␣-erbB2 NK cells had
`no anti-tumor effect in mice injected with RMA parental tumor. In
`another experiment we demonstrated that coadministration of high
`dose IL-2 (200,000 IU/ml) with gene-modified NK cells did not
`improve the anti-tumor effect in mice (data not shown). We also
`investigated the persistence of our adoptively transferred gene-
`modified NK cells by using donor NK cells from congenic C57BL/
`
`0
`
`100
`
`101
`
`103
`
`104
`
`102
`H2Kb
`FIGURE 4. Expression of MHC class I on RMA tumor cells. MHC
`class I expression on RMA parental (thick line) and RMA-erbB2 (thin line)
`tumor cells was determined by staining cells with a PE-conjugated Ab
`specific for mouse H2kb. Unstained RMA tumor cells (dotted line) were
`used as a control.
`
`cells or gene-modified NK cells (Fig. 5E). These data demon-
`strated that expression of the scFv receptor targeting TAA could
`endow primary mouse NK cells with the ability to overcome MHC
`class I-mediated inhibition and kill NK cell-sensitive tumors.
`
`Target cell lysis mediated by gene-modified NK cells was
`perforin dependent
`
`It has been reported that NK cells lyse their targets predominantly
`via the granule exocytosis pathway involving perforin; however,
`they can also mediate apoptotic activity through FasL or TRAIL
`pathways (5, 32). To determine the mechanism of killing used by
`our gene-modified primary mouse NK cells, we genetically mod-
`ified NK cells from C57BL/6 WT, perforin-deficient (pfp⫺/⫺), and
`
`Anti-erbB2-NK
`GFP-NK
`
`*
`
`10:1
`
`5:1
`
`1:1
`
`Anti-erbB2-NK
`GFP-NK
`
`*
`
`20:1
`
`10:1
`
`1:1
`
`B
`60
`50
`40
`30
`20
`10
`0
`D
`50
`40
`30
`20
`10
`0
`
`Anti-erbB2-NK
`GFP-NK
`
`10:1
`
`5:1
`
`1:1
`
`Anti-erbB2-NK
`GFP-NK
`
`20:1
`
`10:1
`
`1:1
`
`Untransfected
`Anti-erbB2-NK
`GFP-NK
`10:1
`
`5:1
`E:T ratio
`
`1:1
`
`A
`60
`50
`40
`30
`20
`10
`0
`C
`50
`40
`30
`20
`10
`0
`E
`100
`80
`60
`40
`20
`0
`
`Specificlysis(%)
`
`FIGURE 5. Enhanced cytotoxicity of erbB2⫹ tumor cells by anti-erbB2
`NK cells. A, Gene modification of primary mouse NK cells with the scFv
`chimeric anti-erbB2 receptor enhanced killing of RMA-erbB2 target cells
`compared with GFP-NK cells. B, Anti-erbB2 NK and GFP-NK cells equiv-
`alently killed RMA parental cells. C, Gene modification of primary mouse
`NK cells with the scFv chimeric anti-erbB2 receptor enhanced killing of
`B16-F10-erbB2 target cells compared with GFP-NK cells. D, Anti-erbB2
`NK and GFP-NK cells equivalently killed B16-F10 parental cells. E, Anti-
`erbB2 NK, GFP-NK, or untransfected cells equivalently killed the NK cell
`sensitive target YAC-1. (ⴱ, p ⬍ 0.05, as determined by a Mann-Whitney U
`test). Results are expressed as average ⫾ SEM of triplicates from three
`independent experiments.
`
`UPenn Ex. 2067
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3452
`
`

`

`Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/5/3449/1267196/zim01708003449.pdf by guest on 08 February 2023
`
`The Journal of Immunology
`
`3453
`
`doses of anti-erbB2-NK significantly enhanced the survival of
`mice (⬃35% mice tumor-free) compared with previous experi-
`ments involving two injections of gene-modified NK cells (Fig. 8).
`Mice that received control GFP-NK cells or RMA-erbB2 tumor
`alone rapidly succumbed to disease. Collectively, this data dem-
`onstrated for the first time that adoptive transfer of gene-modified
`primary mouse NK cells could mediate an effective Ag-specific
`tumor response in vivo.
`
`Discussion
`The use of NK cells for cancer immunotherapy is gaining much
`attention. The most promising developments have come from the
`transfer of allogeneic NK cells in the allogeneic transplant stem
`cell setting (34). Recent results have demonstrated that treatment
`of acute myeloid leukemia patients with alloreactive NK cells
`could substantially increase their survival without associated graft-
`vs-host effects (14). Nevertheless, improvements in the use of al-
`loreactive NK cells are required given that these cells had no ap-
`parent effect in patients with acute lymphoid leukemia (14). Other
`therapies involving the transfer of IL-2-activated LAK cells have
`shown only modest anti-tumor effects in patients. This is due in
`part to their nonspecific nature and to HLA-mediated inhibitory
`signals induced by interaction with NK cell inhibitory receptors
`(9). A novel way to overcome these problems and enhance NK cell
`anti-tumor activity involves their genetic modification with scFv
`chimeric receptors that can specifically recognize TAA. To test
`this we used the Amaxa transfection system to genetically engineer
`primary mouse NK cells with a chimeric scFv receptor with spe-
`cific recognition for the erbB2 TAA. Importantly, the expression
`of the chimeric receptor in mouse NK cells did not interfere with
`their natural cytotoxic capability against NK-sensitive target cells.
`We demonstrated that NK cells engineered with the scFv anti-
`erbB2 receptor could significantly enhance killing of an essentially
`NK-insensitive lymphoma cell line in an erbB2⫹ Ag-specific man-
`ner. Furthermore, for the first time, we demonstrated that adoptive
`transfer of receptor-modified primary mouse NK cells could spe-
`cifically enhance the survival of tumor-bearing mice.
`A number of studies have shown that gene modification of var-
`ious mouse and human NK cell lines with scFv chimeric receptors
`could specifically enhance their anti-tumor activity in vitro (25, 27,
`35, 36). Another report demonstrated that human primary NK cells
`expressing an anti-CD19 scFv receptor could specifically kill
`CD19⫹ leukemic cells (28). Nevertheless, the ability of primary
`NK cells to mediate Ag-specific anti-tumor effects in vivo has not
`been formally tested. This has been due to difficulties in using
`retroviral-based approaches to efficiently express chimeric scFv
`receptors in NK cells, which is particularly the case for primary
`mouse NK cells. In our study, we were able to demonstrate proof
`of principle that adoptively transferred gene-modified primary
`mouse NK cells could specifically mediate anti-tumor inhibition in
`vivo. Indeed, mice treated with four doses of anti-erbB2-NK cells
`resulted in ⬃35% long term survivors. Nevertheless, these exper-
`iments were performed in RAG-1⫺/⫺ mice where the presence of
`endogenous NK cells may have competed for important growth
`factors and cytokines limiting both the persistence and activity of
`gene-modified NK cells. Persistence could potentially be improved
`by a non-myeloablative conditioning regimen before adoptive NK
`cell transfer to produce a conducive cytokine environment. This
`type of approach has been demonstrated to enhance the therapeutic
`efficacy of adoptively transferred T cells in both mouse models and
`in patients (37– 41). Alternatively, in future experiments, the use of
`⫺/⫺ recipient mice (that also lack NK cells) may overcome
`RAG␥
`c
`this problem in an experimental setting and result in increased
`
`RMA-erbB2 +
`Anti-erbB2 NK
`RMA-erbB2
`alone
`RMA +
`Anti-erbB2-NK
`RMA-erbB2 +
`GFP-NK
`5
`
`10
`
`*
`
`15
`
`20
`
`25
`
`30
`
`*
`
`20
`15
`10
`5
`Days after tumor inoculation
`
`25
`
`A
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`B
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`0
`
`0
`
`Survival(%)
`
`FIGURE 7. Enhanced survival of tumor-bearing mice after adoptive
`transfer of ␣-erbB2 NK cells. Groups of 5–10 mice were injected i.p. with
`2 ⫻ 105 RMA-erbB2 tumor alone (closed squares) or injected with RMA-
`erbB2 tumor and treated with two doses of 2 ⫻ 106 ␣-erbB2-NK cells
`(closed triangles) or GFP-NK cells (open triangles) on days 0 and 1 (A) or
`days 3 and 4 (B). Mice bearing RMA tumor were treated with 2 ⫻ 106
`␣-erbB2-NK cells (open squares). Mice bearing RMA-erbB2 tumor and
`treated with ␣-erbB2 NK cells showed significantly increased survival
`compared with mice treated with control GFP NK cells (ⴱ, p ⬍ 0.05, as
`determined by a Mann-Whitney U test). Results shown are representative
`of two experiments performed. Arrows indicated days of NK cell transfer.
`
`6-PTPRCa mice. In these experiments we could not detect signif-
`icant persistence of these cells 7 days post transfer in recipient
`mice (data not shown). To determine whether increasing the num-
`ber of doses of anti-erbB2-NK cells could enhance the anti-tumor
`response, RAG-1⫺/⫺ recipient mice bearing RMA-erbB2 tumor
`were injected i.p with 2 ⫻ 106 anti-erbB2-NK or GFP-NK cells on
`days 0, 1, 2, 3. We demonstrated that increasing the number of
`
`RMA-erbB2+ Anti-
`erbB2-NK
`RMA-erbB2+ GFP-NK
`RMA-erbB2 alone
`
`*
`
` 40
`30
`20
`10
`
`Days after tumor inoculation
`
`50
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`0
`
`Survival(%)
`
`Increased transfer of ␣-erbB2 NK cells leads to tumor free
`FIGURE 8.
`survival of mice. Groups of six mice were injected i.p. with 2 ⫻ 105
`RMA-erbB2 tumor alone (closed squares) or injected with RMA-erbB2
`tumor and treated with four doses of 2 ⫻ 106 ␣-erbB2-NK cells (closed
`triangles) or GFP-NK cells (open triangles) on days 0, 1, 2, and 3. Mice
`bearing RMA-erbB2 tumor and treated with four doses of ␣-erbB2 NK
`cells showed significantly increased survival compared with mice treated
`with control GFP NK cells (ⴱ, p ⬍ 0.05, as determined by a Mann-Whitney
`U test). Arrows indicated days of NK cell transfer.
`
`UPenn Ex. 2067
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3453
`
`

`

`Downloaded from http://journals.aai.org/jimmunol/article-pdf/181/5/3449/1267196/zim01708003449.pdf by guest on 08 February 2023
`
`3454
`
`ENHANCED TUMOR INHIBITION BY GENE-MODIFIED NK CELLS
`
`persistence of gene-modified NK cells. The combination of lym-
`phodepletion and CD34⫹ hematopoietic stem cells has shown to
`further enhance the activity of transferred T cells (42). Thus, it
`would be of interest in future studies to test whether these types of
`regimens can similarly increase persistence and anti-tumor effects
`of transferred gene-modified NK cells.
`The anti-tumor effects observed with adoptive transfer of LAK
`cells in melanoma patients have been dependent on coadministra-
`tion of high dose IL-2 (43). However, in our mouse model coad-
`ministration of high dose IL-2 did not improve the anti-tumor ef-
`fects by gene-modified NK cells. There have been reports that
`other cytokines such as IL-15 are required for the persistence of
`NK cells (44, 45). Thus, it would be interesting to test in our model
`whether cytokines such as IL-15 may be of some benefit.
`To achieve receptor expression in this study we used a non-viral
`vector system, which has attractive safety aspects compared with
`viral-based systems, particularly when considering clinical appli-
`cations. In addition, expression levels using this method, although
`high, were largely transient, lasting ⬃72 h. This provides added
`safety by reducing the risk of long-term autoimmunity associated
`with prolonged presence of potentially autoreactive cells. Never-
`theless, improvements in safety and efficiency of gene transfer
`technology for primary mouse NK cells may lead to increased
`anti-tumor effects. A recent report using lentivirus transduction
`demonstrated long term and stable expression of GFP in mouse
`NK cells in vitro without affecting NK cell phenotype and function
`(46). It will be of interest in future experiments to test whether the
`use of lentiviral vectors can maintain stable

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