throbber
Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen
`receptor
`Bipulendu Jena, 1 Gianpietro Dotti, 2 and Laurence J. N. Cooper1
`'Division of Pediatrics, University of Texas M. D. Anderson Cancer Center, Houston; and 2Center for Cell and Gene Therapy, Baylor College of Medicine,
`Houston, TX
`
`Infusions of antigen-specific T cells have
`yielded therapeutic responses in patients
`with pathogens and tumors. To broaden
`the clinical application of adoptive immu(cid:173)
`notherapy against malignancies, investi(cid:173)
`gators have developed robust systems
`for the genetic modification and character(cid:173)
`ization of T cells expressing introduced
`chimeric antigen receptors (CA Rs) to redi(cid:173)
`rect specificity. Human trials are under
`way in patients with aggressive malignan-
`
`cies to test the hypothesis that manipulat(cid:173)
`ing the recipient and reprogramming
`T cells before adoptive transfer may im(cid:173)
`prove their therapeutic effect. These ex(cid:173)
`amples of personalized medicine infuse
`T cells designed to meet patients' needs
`by redirecting their specificity to target
`molecular determinants on the underly(cid:173)
`ing malignancy. The generation of clinical
`grade CAR+ T cells is an example of
`bench-to-bedside translational science
`
`that has been accomplished using inves(cid:173)
`tigator-initiated trials operating largely
`without industry support. The next-gen(cid:173)
`eration trials will deliver designer T cells
`with improved homing, CAR-mediated
`signaling, and replicative potential, as
`investigators move from the bedside to
`the bench and back again. {Blood. 201 0;
`116(7):1035-1044)
`
`1 ntrod uction
`
`The systematic development and clinical application of genetically
`modified T cells is an example of how academic scientists working
`primarily at nonprofit medical centers are generating a new class of
`therapeutics. In this context, gene therapy has been used to
`overcome one of the major barriers to T-cell therapy of cancer,
`namely tolerance to desired target tumor-associated antigens (TAAs).
`This was achieved by the introduction of a chimeric antigen
`receptor (CAR) to redirect T-cell specificity to a TAA expressed on
`the cell surface. The prototypical CAR uses a mouse monoclonal
`antibody (mAb) that docks with a designated TAA and this binding
`event is reproduced by the CAR to trigger desired T-cell activation
`and effector functions. Multiple early-phase clinical trials are now
`under way or have been completed to evaluate the safety and
`feasibility of adoptive transfer of CAR+ T cells (Table 1). These
`pilot studies have revealed challenges in achieving reproducible
`therapeutic successes which may be solved by (1) reprogramming
`the T cells themselves for improved replicative potential, effector
`function, and in vivo persistence, (2) manipulating the recipient to
`improve TAA expression and survival of infused T cells, and
`(3) adapting the gene therapy platform to deliver (1) CARs capable
`of initiating an antigen-dependent fully-competent activation sig(cid:173)
`nal, and (2) transgenes to improve safety, persistence, homing, and
`effector functions within the tumor microenvironment. Academic
`investigators who work to both develop and deliver investigational
`biologic agents such as CAR+ T cells are poised to further tighten
`the pace of discovery between the bench and the bedside to
`improve the therapeutic potential of genetically modified T cells
`with redirected specificity. This review builds upon recent articles
`that describe the immunobiology of CAR+ T cells (Table 2) and we
`highlight how the CAR technology has been adapted to meet the
`challenges of infusing genetically modified T cells in medically
`fragile patients with aggressive malignancies and what new
`
`directions the field will need to embrace to undertake multicenter
`trials to prove their therapeutic efficacy.
`
`Redirecting T-cell specificity through the
`introduction of a CAR
`
`The generation of T bodies, or CARs, by Eshhar et al has been
`adapted by investigators as a tool to enable T cells, as well as other
`immune cells, to overcome mechanisms (eg, loss of human
`leukocyte antigen [HLA]1) by which tumors escape from immune
`surveillance of the patient's endogenous (unmanipulated) T-cell
`repertoire. 2•3 The specificity of a CAR is achieved by its exodomain
`which is typically derived from the antigen binding of a mAb
`linking the V Hand V L domains to construct a single-chain fragment
`variable (scFv) region. The exodomains of CARs have also been
`fashioned from ligands or peptides (eg, cytokines) to redirect
`specificity to receptors (eg, cytokine receptors), such as the
`IL-13Ru2-specific "zetakine."4 The exodomain is completed by
`the inclusion of a flexible (hinge) sequence, such as from CD8u or
`immunoglobulin sequence5·6 and via a transmembrane sequence,
`the exodomain is fused to 1 or more endodomain(s) which may
`include cytoplasmic domains from CD3-E, CD3--y, or CD3-1; from
`the T-cell receptor (TCR) complex or high-affinity receptor FcERI.7·9
`When CARs are expressed on the cell surface of genetically
`modified T cells, they redirect specificity to TAA (including TAA
`on tumor progenitor cells 10) independent of major histocompatibil(cid:173)
`ity complex (MHC). This direct binding of CAR to antigen ideally
`provides the genetically modified T cell with a fully-competent
`activation signal, minimally defined as CAR-dependent killing,
`proliferation, and cytokine production. Given that T cells targeting
`tumor through an endogenous ul3TCR (and even introduced TCR
`
`Submitted January 8, 2010; accepted April 26, 2010. Prepublished online as
`Blood First Edition paper, May 3, 201 O; DOI 10.1182/blood-2010-01-043737.
`
`© 201 O by The American Society of Hematology
`
`BLOOD, 19AUGUST 2010 • VOLUME 116, NUMBER 7
`
`1035
`
`Miltenyi Ex. 1031 Page 1
`
`

`

`1036
`
`JENA et al
`
`BLOOD, 19 AUGUST 2010 • VOLUME 116, NUMBER 7
`
`Table 1. Clinical trials in the United States infusing CAR+ T cells under IND
`Viral-
`specific Lympho-
`Teen
`depletion
`
`Tumor target
`
`Antigen
`
`CAR
`generation
`
`ClinicalTrial.gov
`identifier
`
`Enrolling
`
`SAE
`
`Gene transfer
`
`Kappa light chain B-NHL and B-CLL
`2 CD19
`Lymphoma/leukemia (B-NHL) and CLL
`3 CD19
`Advanced B-NHUCLL
`4 CD19
`Lymphoma and leukemia
`5 CD19
`ALL (post-HSCT)
`6 CD19
`Follicular NHL
`7 CD19
`CLL
`
`8 CD19
`9 CD19
`10 CD19
`11 CD19
`
`B-NHUleukemia
`B-cell leukemia, CLL and B-NHL
`B-ALL
`B-lymphoid malignancies
`
`No
`No
`Yes
`No
`Yes
`No
`No
`
`No
`No
`No
`No
`
`Yes
`No
`No
`Yes
`No
`Yes
`Yes/no
`
`Yes
`No
`Yes
`Yes
`
`First and second NCT00881920
`First and second NCT00586391
`First and second NCT00709033
`Second
`NCT00924326•
`Second
`NCT00840853
`First
`NCT00182650
`Second
`NCT00466531 •
`
`First and second NCT00891215
`Second
`NCT01087294
`Second
`NCT01044069
`Second
`NCT00968760
`
`Yes
`Yes
`Yes
`Yes
`Yes
`No
`Yes
`
`Yes
`Yes
`Yes
`No
`
`TBM
`TBM
`TBM
`TBM
`TBM
`No
`Yes
`(1 Death)
`TBM
`TBM
`TBM
`TBM
`
`Virus
`Virus
`Virus
`Virus
`Virus
`Electroporation
`Virus
`
`Virus
`Virus
`Virus
`Electroporation
`(SB system)
`Electroporation
`Electro po ration
`Virus
`Virus
`Virus
`Electroporation
`Virus
`Virus
`Virus
`Virus
`Electroporation
`Virus
`
`NCT00012207'
`NCT00621452
`NCT00085930•
`NCT00004178
`NCT00664 196
`NCT00006480
`NCT00019136
`NCT00429078
`NCT00673829
`NCT00673322
`NCT00730613
`NCT00924287
`
`No
`Yes
`Yes
`No
`Yes
`No
`No
`Yes
`Yes
`Yes
`No
`No
`
`12 CD20
`Relapsed/refractory B-NHL
`13 CD20
`Mantle cell lymphoma or indolent B-NHL
`14 GD2
`Neuroblastoma
`15 CEA
`Adenocarcinoma
`16 PSMA
`Prostate cancer
`17 CD171/L 1-CAM Neuroblastoma
`18 FR
`Ovarian epithelial cancer
`19 CEA
`Stomach carcinoma
`20 CEA
`Breast cancer
`21 CEA
`Colorectal carcinoma
`IL-13Ra2
`22
`Glioblastoma
`23 ERBB2
`Metastatic cancer
`(HER2/neu)
`24 HER2/neu
`25 HER2/neu
`
`Lung malignancy
`Advanced osteosarcoma
`
`No
`No
`Yes
`No
`No
`No
`No
`No
`No
`No
`No
`No
`
`Yes
`No
`
`Yes
`Yes
`Yes
`No
`Yes
`No
`No
`No
`No
`No
`NA
`Yes
`
`First
`Third
`First
`First
`First
`First
`First
`Second
`Second
`Second
`Second
`Third
`
`No
`TBM
`No
`No
`TBM
`No
`No
`TBM
`TBM
`TBM
`No
`Yes
`(1 Death)
`No
`Second
`NCT00889954
`Yes
`Virus
`TBM
`No
`Second
`NCT00902044
`Virus
`Yes
`TBM
`ALL indicates acute lymphoblastic leukemia; NHL, non-Hodgkin lymphoma; CLL, chronic lymphocytic leukemia; GD2, disialoganglioside; FR, alpha folate receptor; CEA,
`carcinoembryonic antigen; L 1-CAM, L 1 cell adhesion molecule; PSMA, prostate-specific membrane antigen; ERBB2, receptor tyrosine-protein kinase erbB-2; Her2, human
`epidermal growth factor receptor; HSCT, hematopoietic stem cell transplantation; NA, not available; SB, Sleeping Beauty; TBM, to be monitored; SAE, serious adverse event;
`and IND, investigational new drug.
`•studies have been described to demonstrate a CAR-mediated antitumor effect based on reduction in tumor size. Other trials demonstrated a biologic effect of CAR+ T
`cells based on reduction in biologic markers of tumor activity.
`
`Table 2. Published reviews since 2003 on immunobiology and
`clinical applications of CAR+ T cells
`References
`Authors
`
`Year published
`
`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`13
`14
`15
`16
`17
`18
`19
`20
`21
`22
`23
`
`11
`12
`13
`14
`15
`16
`17
`18
`19
`20
`21
`22
`23
`24
`25
`26
`27
`28
`29
`30
`31
`32
`33
`
`Rossig and Brenner
`Willemsen et al
`Baxevanis and Papamichail
`Rossig and Brenner
`Rivie re et al
`Thistlethwaite et al
`Kershaw et al
`Dotti and Heslop
`Cooper et al
`Foster and Rooney
`Biagi etal
`Rossi et al
`Varela-Rohena et al
`Eshhar
`Marcu-Malina et al
`Berry et al
`Sadelain et al
`June et al
`Dotti et al
`Till and Press
`Vera et al
`Brenner and Heslop
`Westwood and Kershaw
`
`2003
`2003
`2004
`2004
`2004
`2005
`2005
`2005
`2005
`2006
`2007
`2007
`2008
`2008
`2009
`2009
`2009
`2009
`2009
`2009
`2009
`2010
`2010
`
`chains34) exhibit such a fully-competent activation signal, investi(cid:173)
`gators have iteratively designed and tested CARs, for example,
`with 1 or more activation motifs, to try and recapitulate the
`signaling event mediated by al3TCR chains. These changes to the
`CAR are enabled by the modular structure of a prototypical CAR
`and this has resulted in first-, second-, and third-generation CARs
`designed with 1, 2, or 3 signaling endodomains (Figure 1)
`including a variety of signaling motifs, such as chimeric CD28,
`CD134, CD137, Lek, ICOS, and DAPl0.6-35-37 A listing of such
`CARs is provided by Berry et al26 and Sadelain et al.27 While the
`optimal CAR design remains to be determined, at present it is
`believed that the first-generation technology, in which a CAR
`signals solely through immunoreceptor tyrosine-based activation
`motif (ITAM) domains on CD3-1;, is insufficient to sustain in vivo
`persistence of T cells. 38 This is supported by early clinical data
`which demonstrate
`that CD19-specific,39 CD20-specific,40
`GD2-specific (not Epstein-Barr virus [EBY] bispecific),41 and
`LI-CAM (cell adhesion molecule)-specific42 T cells had appar(cid:173)
`ently short-lived persistence in peripheral blood. The decision
`regarding which second- or even third-generation CAR design to
`use in clinical trials is predicated on the ability of a CAR to activate
`T cells for desired T-cell effector function, which at a minimum
`includes CAR-dependent killing. However, it is possible that
`next-generation CARs can be engineered to provide a supraphysi(cid:173)
`ologic activation signal which may be detrimental to continued
`T-cell survival and perhaps even the well-being of the recipient.43
`
`Miltenyi Ex. 1031 Page 2
`
`

`

`BLOOD, 19AUGUST2010 • VOLUME 116, NUMBER 7
`
`IMMUNOBIOLOGY OFT CELLS EXPRESSING CAR
`
`1037
`
`A
`
`Figure 1. Modular structure of prototypical CAR.
`CAR shown dimerized on the cell surface demonstrat(cid:173)
`ing the key extracellular (A-B) and intracellular (C-E)
`A domains. CARs may express 1, 2, or 3 signaling
`motifs within an endodomain to achieve a CAR(cid:173)
`dependent fully-<:ompetent T-<:ell activation signal.
`The modular structure of the CAR's domains, for
`example, the scFv (VL linked to VH) region (A) and the
`flexible hinge and spacer, for example, from lgG4
`hinge, CH2, and CH3 regions (8), allow investigators
`to change specificity through swapping of exodo(cid:173)
`mains and achieve altered function by varying trans(cid:173)
`membrane and intracellular signaling moieties (C-E).
`
`B
`
`1 s1 Generation CAR
`One T -cell signaling endodomain
`
`2 nd Generation CAR
`Two T-cell signaling endodomains
`
`3"' Generation CAR
`Three T-cell signaling endodomains
`
`Modifying the CAR to achieve a fully competent
`activation signal and reduce immunogenicity
`
`The inclusion of 1 or more T-cell costimulatory molecules within
`the CAR endodomain is in response to the appreciation that
`genetically modified and ex vivo-propagated T cells may have
`down-regulated expression of desired endogenous costimulatory
`molecules ( eg, CD28) or that the ligands for these receptors may be
`missing on tumor targets (eg, absence of CD80/CD86 on blasts
`from B-lineage acute lymphoblastic leukemia). By way of ex(cid:173)
`ample, chimeric CD28 and CD3-t38,44 CD137 and CD3-t23-36 or
`CD134 and CD3-t,45,46 have been incorporated into the design of
`second-generation CARs with the result that these CARs with
`multiple chimeric signaling motifs exhibited effector function
`stemming from a both a primary signal (eg, killing) and costirnula(cid:173)
`tory signal (eg, IL-2 production) after an extracellular recognition
`event.47·48 Animal studies have demonstrated that T cells express(cid:173)
`ing a second-generation CD19-specific36,38 and carcinoembryonic
`antigen-specific CAR49 exhibited an improved antitumor effect
`compared with T cells bearing a first-generation CAR. In addition
`to altering the endodomains, investigators have also made changes
`to the scFv to improve affinity based on selecting high-affinity
`binding variants from phage arrays.50,51 The approach to develop(cid:173)
`ing CAR+ T cells with a calibrated increase in functional affinity
`may be necessary to enable genetically modified T cells to target
`tumors with low levels of antigen expression or perhaps to target a
`cell-surface molecule in the presence of soluble antigen.52.53
`Because a CAR typically contains a murine scFv sequence which
`may be subject to immune recognition leading to deletion of
`infused T cells, investigators have developed humanized scFv
`regions to target carcinoembryonic antigen (CEA)54-55 and ERBB2
`
`(a member of epidermal growth factor receptor family).6 However,
`a benefit for using human or humanized scFv regions is yet to be
`established in the clinical setting.
`
`Imaging CAR+ T cells by positron emission
`tomography
`
`The ability to genetically modify T cells to redirect specificity
`provides investigators with a platform to express other transgenes
`such as for noninvasive imaging. Such temporal-spatial imaging is
`desired as a surrogate marker for a CAR-mediated antitumor effect
`and to serially determine number and localization of infused
`T cells. One imaging transgene coexpressed with CAR is thymi(cid:173)
`dine kinase (TK) and associated mutants from herpes simplex
`virus-I (HSV-1)56 which can be used to enzymatically trap
`radioactive substrates within the cytoplasm to image the locore(cid:173)
`gional biodistribution of T cells using positron emission tomogra(cid:173)
`phy.57-59 The expression ofTK also renders CAR+ T cells sensitive
`to conditional ablation using ganciclovir in a cell-cycle-dependent
`manner.60-62
`
`Gene transfer of CARs
`
`Approaches to the genetic manipulation of T cells for the introduc(cid:173)
`tion of CAR transgene use either viral-mediated transduction, or
`nonviral gene transfer of DNA plasmids or in vitro-transcribed
`mRNA species, The advantage of retrovirus63-65 or lentivi(cid:173)
`rus23·36·66·67 to modify populations of T cells lies in the efficiency of
`gene transfer, which shortens the time for culturing T cells to reach
`clinically-significant numbers (Figure 2), Gamma retroviruses, the
`
`Electroporation
`(Non-viral gene transfer of
`
`Numeric expansion using aAPC
`
`Harvest
`
`Figure 2. Timeline for in vitro gene transfer and propagation
`of CAR+ T cells. The electrotransfer of transposon/transposase
`systems has narrowed the gap between nonviral and viral-based
`gene transfer for the amount of time in tissue culture needed to
`generate a clinically sufficient number of genetically modified
`CAR+ T cells. Cells transduced with virus (blue text) are typically
`propagated for 3 weeks before infusion. 68,69 T cells that undergo
`nonviral gene transfer with the SB system (red text) can be
`typically harvested within 4 weeks.
`
`Woo~~ "f--s-y:_~_e_~_: _ _.__ _ _ _ _ _ _ ,__ _ _ _ _ _ __. _ _ __c:..:.;_ __ . . . , _ _ )
`I
`I
`Week4
`Week3
`Week2
`l
`J
`
`y
`Harvest
`
`Transduction
`(Viral-based gene transfer
`ofy-retrovirus)
`
`------------------
`
`Numeric expansion using 0KB and IL-2
`
`Miltenyi Ex. 1031 Page 3
`
`

`

`1038
`
`JENA et al
`
`Figure 3. Schematic of vector systems to express A
`CAR transgenes used in clinical trials. (A) Two SB
`DNA plasmids expressing a (CAR) transposon and a
`hyperactive transposase (eg, SB11 ). Transposition oc-
`curs at a TA dinucleotide sequence when the trans(cid:173)
`posase enzymatically acts on the internal repeat flank-
`ing the transposon. (B) A recombinant retroviral vector
`showing the long terminal repeats (LTR) containing the
`promoter flanking the CAR. SD and SA are the splice 8
`donor and splice acceptor sites, respectively, and tJ, is
`the viral packaging signal. (C) A self-inactivating recom(cid:173)
`binant lentiviral vector construct containing the LTR, tf,,
`SD and SA s~es, HIV Rev response element (ARE),
`HIV central polypurine tract (cPPT), CAR under control
`of an internal promoter, and the wood-chuck hepatitis
`virus posttranscriptional regulatory element (WPRE).
`
`... IR/OR
`
`Promoter
`
`Promoter
`
`BLOOD, 19AUGUST2010 • VOLUME 116, NUMBER 7
`
`Transposon ,
`
`CAR
`
`Poly A
`
`S'LTR
`
`CAR
`
`3'LTR
`
`C ,______sd ~l-0-c::$--Es:JC __ }- -{-J"_ -
`
`S'LTR
`
`'i' HIV RRE HIV cPPT
`
`Promoter
`
`CAR
`
`WPRE
`
`3'LTR
`
`most common vector system used to genetically modify clinical
`grade T cells (Figure 3), have introduced CAR into T cells with a
`proven ability to exert a therapeutic effect.41 Self-inactivating
`lentiviral vectors (Figure 3) hold particular appeal as they appar(cid:173)
`ently integrate into quiescent T cells.28 These recombinant viruses
`often have a high cost to manufacture at clinical grade in
`specialized facilities skilled in current good manufacturing practice
`(cGMP) which may preclude investigators from undertaking
`clinical trials. As an alternative to transduction, we and others have
`adapted electroporation as an approach to the nonviral gene transfer
`of DNA42,70,71 resulting in CAR+ T cells that have been attributed
`to have an antitumor effect in a clinical triaI.40 Previously, the
`electrotransfer and integration of naked plasmid DNA into T cells
`was considered inefficient because it depended on illegitimate
`recombination for stable genomic insertion of nonviral sequences.
`As a result, lengthy in vitro culturing times were required to select
`for T cells bearing stable integrants72•73 during which period the
`T cell became differentiated, and perhaps terminally differentiated,
`into effector cells which may have entered into replicative senes(cid:173)
`cence. This time in tissue culture can now be considerably
`shortened by using transposon/transposase systems such as Sleep(cid:173)
`ing Beauty (SB)7o,74,75 and piggyback16•11 to stably introduce CAR
`from DNA plasmids (Figure 3).71 •76•78· 84 We have shown that the SB
`system can be used to introduce CAR and other transgenes into
`human T cells with an approximately 60-fold improved integration
`efficiency compared with electrotransfer of DNA transposon
`plasmid without transposase74 and this provided the impetus to
`adapt the SB system for use in clinical trials. 85 After electropora(cid:173)
`tion, T-cell numbers from peripheral and umbilical cord blood can
`be rapidly increased in a CAR-dependent manner by recursive
`culture on -y-irradiated artificial antigen-presenting cells (aAPC)
`achieving clinically sufficient numbers of cells for infusion within
`3 to 4 weeks after electroporation (Figure 2). Given that T cells
`transduced with -y-retrovirus are typically cultured with OKT3 and
`IL-2 for approximately 3 weeks before infusion, the use of the SB
`system with aAPC does not appear to greatly lengthen the ex vivo
`culturing process.
`
`Improving the therapeutic potential of CAR+
`T cells
`
`T cells and polarized T-cell subsets, such as TH I and TH 17 cells,
`may be genetically modified to be specific for a catalog of
`
`cell-surface TAAs as described in recent reviews (Table 2). To
`enable these CAR+ T cells to achieve their full therapeutic
`potential in clinical trials, there are 3 major challenges to be
`overcome.
`
`Persistence
`
`The adoptively transferred CAR+ T cells must survive and perhaps
`also numerically expand to achieve a robust antitumor effect. One
`approach to improving persistence is to alter the host environment
`into which the T cells are infused. For example, rendering the
`recipient lymphopenic, or even aplastic by chemotherapy and/or
`radiation therapy, improves the persistence of adoptively trans(cid:173)
`ferred T cells (and natural killer [NK] cells).86·90 Presumably, the
`infused T cells proliferate in the lymphopenic recipient through
`homeostatic mechanisms mediated by the removal of regulatory/
`suppressor cells and the ready availability of previously scarce
`homeostatic cytokines. This approach likely improves in vivo
`persistence and thus efficacy of CAR+ T cells as a published trial
`and animal studies have hinted.40 However, combining chemo(cid:173)
`therapy and T-cell therapy might increase toxicity and compromise
`the interpretation of an antitumor effect for it may be difficult to
`definitively attribute efficacy to the infused CAR+ T cells in trials
`enrolling small numbers of patients who received concomitant
`chemotherapy. Trials infusing T cells expressing a first-generation
`CAR design (signaling solely through chimeric CD3-t endodo(cid:173)
`main) revealed that these T cells appear to have limited long-term
`persistence and one way of circumventing this limitation is to
`engineer CAR endodomains to deliver an activation signal for
`sustained proliferation. This has been achieved by the design of
`second-generation CARs that signal through 2 signaling domains
`and these are now in multiple clinical trials. Third-generation
`CARs are now being evaluated in humans in a few trials (Table 1)
`which are composed of 3 chimeric signaling moieties.6•35 Rather
`than modifying the CAR design to sustain proliferation, investiga(cid:173)
`tors have used signaling molecules alongside CARs to achieve
`improved persistence. For example, constitutively expressed recom(cid:173)
`binant CDS0 and CD137L alongside the CAR demonstrated
`coordinated signaling between these 2 introduced receptors with
`endogenous ligands CD28 and CD137 within the immunologic
`synapse on or between T cells, which could improve antitumor
`effects.91 CAR has been introduced into T cells expressing endoge(cid:173)
`nous al3TCR that recognize allo-92 or viral antigens.41 •93·95 Trigger(cid:173)
`ing such TCRs in vivo can be used to numerically expand T cells to
`
`Miltenyi Ex. 1031 Page 4
`
`

`

`BLOOD, 19AUGUST 2010 • VOLUME 116, NUMBER 7
`
`IMMUNOBIOLOGY OFT CELLS EXPRESSING CAR
`
`1039
`
`achieve an improved antitumor effect delivered by the CAR as was
`recently demonstrated by infusing bispecific T cells which recog(cid:173)
`nized EBY-specific antigens via the endogenous al3TCR and GD2
`on neuroblastoma cells by the introduced CAR.41 Because T cells
`may require exogenous cytokines to sustain in vivo persistence,96
`investigators have enforced expression of cytokines that signal
`through the common 'Y cytokine receptor chain. Animal experi(cid:173)
`ments97 as well as clinical experience98,99 have shown that long(cid:173)
`lived T cells are associated with expression of the IL-7Ra chain
`(CD127), but genetically modified and cultured T cells tend to
`down-regulate this receptor. Therefore, to enhance the ability of
`T cells to respond to IL-7, made available after inducing lymphope(cid:173)
`nia, 100·102 investigators have enforced the expression of IL-7Ra to
`demonstrate improved survival of EBY-specific T cells in an
`animal model103 or introduced a novel membrane-bound variant of
`IL-7 that when expressed on the cell surface improved persistence
`of CAR+ T cells. 104 Furthermore, overexpression of receptors for
`IL-2105 and IL-15 106 as well as enforced expression of the cytokines
`themselves107 have improved persistence ofT cells. 108·110 However,
`when this approach was tested in a clinical trial infusing tumor(cid:173)
`infiltrating lymphocytes (TIL) genetically modified to constitu(cid:173)
`tively secrete IL-2, the persistence of the adoptively transferred
`T cells was not improved compared with genetically unmodified
`TIL. 111 As constitutive expression of cytokines and/or cytokine
`receptors is tested in early clinical trials, investigators will need to
`consider coexpressing transgenes for conditional ablation of these
`genetically modified T cells to guard against aberrant proliferation
`as unopposed cytokine signaling may lead to aberrant T-cell
`growth. 109,112 The type of T cell into which the CAR is introduced
`may also impact persistence after adoptive immunotherapy. This
`has been demonstrated in the monkey by infusing autologous
`preselected central memory T cells which despite ex vivo numeric
`expansion retained superior in vivo persistence compared with
`adoptive transfer of differentiated effector T cells. 113 These observa(cid:173)
`tions have been expanded upon by Hinrichs et al who showed that
`an infusion of naive murine T cells was associated with improved
`T-cell persistence.114 These animal observations are likely to
`influence the design of trials infusing genetically modified T cells
`as investigators seek to introduce CARs into T cells that preserve
`the functional capacity of central memory or naive T cells.
`
`Homing
`
`an ability to overcome the adverse regulatory effects within the
`tumor microenvironment. For example, investigators have intro(cid:173)
`duced a dominant-negative receptor for TGFl3 to enable genetically
`modified T cells can resist the suppressive effects of this pleiotropic
`cytokine. 119 Recognizing that the tumor microenvironment con(cid:173)
`tains regulatory T cells (T,egs), the CAR signaling motif has been
`adapted to resist the suppressive effects of these cells by the
`expression of chimeric CD28. 120 The ability to engineer CAR+
`T cells to successfully function within tumor deposits remains
`relatively underexplored as most clinical trials to date have used
`CAR+ T cells with specificity for hematopoietic malignancies.
`However, trials have been published and are under way which
`seek to treat solid tumors 41•42·121 (Table 1) and systematic
`approaches to enabling infused T cells to effectively operate
`within a tumor microenvironment will be needed to reliably
`eliminate large tumor masses.
`
`Reprogramming T cells
`
`The ex vivo gene transfer and propagation (Figure 2) of T cells
`provides an opportunity to further manipulate T cells before
`infusion. The in vitro propagation of T cells provides investigators
`with an opportunity to numerically expand T cells from scant
`starting numbers, such as when T cells are genetically modified
`from umbilical cord blood with the intent to augment the graft(cid:173)
`versus-tumor-effect after allogeneic hematopoietic stem cell trans(cid:173)
`plantation (HSCT).61 However, the in vitro culturing process can
`also be adapted to modify T cells for desired effector function by
`the selective addition of a subset of soluble cytokines, for example,
`that bind via common 'Y chain receptor to the culture media during
`ex vivo culture. 122,123 In addition, to help maintain a desired T-cell
`phenotype after gene transfer, investigators have provided costimu(cid:173)
`latory signals by the addition of CD28-specific mAb in addition to
`OKT3, often using beads conjugated to these mAbs. As an
`alternative, we and others have used immortalized cells in tissue
`culture, such as 3T3 124·125 and K56274·126·128 which can be geneti(cid:173)
`cally modified to express desired T-cell costimulatory molecules
`and function when irradiated as aAPC.
`
`To act on and within the tumor, genetically modified T cells must
`home to the site(s) of malignancy. Migration may be compromised
`by the loss of desired chemokine receptors during genetic modifica(cid:173)
`tion and passage ex vivo, or may result from the selection ofT cells
`that are inherently unable to localize to certain tissues. Panels of
`tissue-specific homing receptors which are typically composed of
`integrins, chemokines, and chemokine receptors are associated
`with T-cell migration to anatomic sites of malignancy, and flow
`cytometry can therefore be used to describe the potential migration
`patterns of T cells before infusion. 115-116 It is unclear whether
`T cells capable of expressing a desired matrix of endogenous
`homing receptors can be genetically modified to express CAR.
`Therefore, investigators are manipulating the homing potential of
`T cells through the enforced expression of chemokine receptors
`such as CCR4. 117•118
`
`Overcoming mechanisms of resistance
`
`Once infused T cells persist and home, they must be able to execute
`and recycle their CAR-dependent effector function, which requires
`
`Expressing CARs in cells other than a(3 TCR+
`T cells
`
`Populations of hematopoietic cells other than T cells expressing
`al3TCR, such as NK cells, cytokine-induced killer (CIK) cells,
`monocytes, and neutrophils have been genetically modified to
`express CARs.27,28 Given the inherent lytic potential of NK cells,
`redirection of their specificity via a CAR is appealing. 129·133 CAR
`signaling endodomain(s) can be specifically adapted or chosen to
`enhance NK-cell signaling versus those that activate T cells. 134 One
`drawback to adoptive transfer of NK cells has been their limited
`survival in vivo after transfer. However, ex vivo NK-cell culturing
`using aAPC adapted from K562 may change this perception. 135-136
`Furthermore, the use of NK cells as a cellular platform for
`introducing CARs may be attractive after allogeneic HSCT as
`donor-derived NK cells do not appear to significantly contribute to
`graft-versus-host-disease (GYHD). 137-138 T cells expressing-y8TCR
`are another lymphocyte population with endogenous killing ability
`that has been modified to express CAR, and these cells can be
`
`Miltenyi Ex. 1031 Page 5
`
`

`

`1040
`
`JENAetal
`
`BLOOD, 19 AUGUST 2010 • VOLUME 116, NUMBER 7
`
`selectively proliferated using an aminobiphosphonate (Zoledr(cid:173)
`onate), 139 raising the possibility that a Food and Drug Administra(cid:173)
`tion (FDA)-approved phannacologic agent can be used to selec(cid:173)
`tively propagate these infused CAR+ T cells in vivo. Instead of
`genetically modifying mature lymphocytes to express CAR, inves(cid:173)
`tigators have introduced CAR into developing thymocytes140 and
`immunoreceptors into hematopoietic stem cells (HSC) 141·143 to
`avoid negative selection and improve immune-mediated surveil(cid:173)
`lance. The approach of genetically modifying lymphoid precursors
`has also been adapted by Zakrzewski et al to make "universal
`T cells" in which CAR+ HSC can be adoptively transferred across
`MHC barriers. 144 It is only a matter of time until embryonic stem
`cells and induced pluripotent stem cells are genetically modified to
`express CAR and differentiated into T cells with redirected and
`desired specificity. The question remains, however, whether these
`other populations of cells have a therapeutic advantage over
`genetic modification and infusion of o:~ + T cells that are geneti(cid:173)
`cally modified to express CAR.
`
`Safety of T cells expressing CAR
`
`There are 4 levels of concern associated with the infusion of
`genetically modified T cells. The first is whether the introduced
`genetic material can lead to genotoxicity. This remains a theoretical
`concern for genetically modified T cells, in contrast to HSC. 145 The
`stable expression of CAR requires the introduction of a promoter
`and the transgene cDNA which raises the possibility of insertional
`mutagenesis. However, to date, there have been no genotoxic
`events associated with serious adverse events attributed to geneti(cid:173)
`cally modified T

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