throbber
Cancer Immunol Immunother (2012) 61:953–962
`DOI 10.1007/s00262-012-1254-0
`
`REVIE W
`
`Targeted immunotherapy of cancer with CAR T cells:
`achievements and challenges
`
`Grazyna Lipowska-Bhalla · David E. Gilham ·
`Robert E. Hawkins · Dominic G. Rothwell
`
`Received: 26 January 2012 / Accepted: 25 March 2012 / Published online: 22 April 2012
` Springer-Verlag 2012
`
`Abstract The adoptive transfer of chimeric antigen receptor
`(CAR)-expressing T cells is a relatively new but promising
`approach in the Weld of cancer immunotherapy. This therapeu-
`tic strategy is based on the genetic reprogramming of T cells
`with an artiWcial immune receptor that redirects them against
`targets on malignant cells and enables their destruction by
`exerting T cell eVector functions. There has been an explosion
`of interest in the use of CAR T cells as an immunotherapy for
`cancer. In the pre-clinical setting, there has been a consider-
`able focus upon optimizing the structural and signaling
`potency of the CAR while advances in bio-processing tech-
`nology now mean that the clinical testing of these gene-modi-
`Wed T cells has become a reality. This review will summarize
`the concept of CAR-based immunotherapy and recent clinical
`trial activity and will further discuss some of the likely future
`challenges facing CAR-modiWed T cell therapies.
`
`Keywords T cell · Gene modiWcation · Chimeric antigen
`receptor · Cancer · Immunotherapy
`
`G. Lipowska-Bhalla · D. E. Gilham · R. E. Hawkins
`Clinical and Experimental Immunotherapy Group,
`School of Cancer and Enabling Sciences,
`Manchester Academic Health Science Centre,
`University of Manchester, Manchester, UK
`
`G. Lipowska-Bhalla · D. G. Rothwell
`Clinical and Molecular Monitoring Laboratory,
`Clinical and Experimental Pharmacology Group,
`Manchester Academic Health Science Centre,
`School of Cancer and Enabling Sciences,
`University of Manchester, Manchester, UK
`
`D. E. Gilham (&)
`Clinical and Experimental Immunotherapy Group,
`Paterson Institute for Cancer Research, Wilmslow Road,
`Withington, Manchester M20 4BX, UK
`e-mail: dgilham@picr.man.ac.uk
`
`Introduction
`
`The concept of the chimeric antigen receptor (CAR; also
`known as T-bodies or chimeric immune receptors) was
`originally described over 15 years ago by Zelig Eshhar and
`colleagues working at the Weissman Institute in Israel [1].
`The approach was based upon the idea of expressing novel
`receptors on the T cell surface that would enable the T cell
`to identify intact protein antigens present on the surface of a
`target cell. T cells generally recognize peptide antigens that
`are presented in association with major histocompatibility
`complex (MHC) proteins by the target cell. However, one
`well-documented tumor escape mechanism is the modula-
`tion or down-regulation of MHC on the surface of the
`tumor cell [2] which thereby eVectively renders the tumor
`“invisible” to T cells, since binding of the T cell receptor to
`peptide-MHC is a pre-requisite for T cell eVector function.
`However, the direct recognition of protein antigens through
`a CAR would then make the tumor cell “visible” to T cell
`immune surveillance once more. Moreover, the use of a tar-
`geting system that functions independently of MHC means
`that the CAR can be used in a generic manner rather than
`having the restrictions that are imposed by the use of T cell
`receptor (TCR) approaches where the speciWc receptor is
`only suitable for patients expressing a speciWc MHC. In
`addition, CARs can substantially broadened the range of
`antigens recognizable by T cells to include carbohydrate [3]
`and glycolipid tumor antigens that are not within the scope
`of TCR-based recognition [4, 5]. Consequently, these fac-
`tors make the use of CARs highly attractive for adoptive
`gene-modiWed T cell therapy. The reader is also directed to
`other highly relevant and recent reviews of CAR T cell
`biology [6–8] and clinical application and focused reviews
`detailing novel directions of CAR T cell
`therapy
`approaches [9].
`
`123
`
`UPenn Ex. 2035
`Miltenyi v. UPenn
`IPR2022-00855
`

`

`

`954
`
`Cancer Immunol Immunother (2012) 61:953–962
`
`The basic CAR structure: extracellular
`and transmembrane protein domains
`
`The basic CAR consists of an extracellular antigen-recogni-
`tion domain attached to an extracellular spacer domain, a
`trans-membrane region that anchors the receptor to the cell
`surface and a signaling endodomain. Single-chain antibody
`fragments (scFv) consisting of the variable heavy (VH) and
`a variable light (VL) chain isolated from an antibody linked
`by a Xexible linker have been used extensively as the anti-
`gen-recognition domain in many CARs due to their small
`size, which facilitates both the genetic manipulation and
`expression of the CAR (Fig. 1). The scFv determines the
`CAR antigen speciWcity and binds the target protein in a
`MHC-independent, non-restricted manner, most commonly
`with the speciWcity and aYnity similar to that of the anti-
`body from which it was derived [10]. More recently, non-
`scFv-based ligand-binding domains have also been suc-
`cessfully utilized in a CAR format including endothelial
`growth factor polypeptide, an integrin binding peptide,
`heregulin, IL-13 mutein, and NK cell receptor NKG2D
`[11–14].
`In most CARs, the antigen-recognition domain is con-
`nected to the transmembrane region by means of an extra-
`cellular spacer domain. The rationale for this is to distance
`the recognition domain from the membrane and to poten-
`tially make it more “accessible” to bind target. Spacer
`
`regions used tend to be comprised of common Ig-like
`domains due to the stability of the protein domain and have
`included immunoglobulin Fc or an extracellular fragment
`derived from CD28, TCR♢ chain, CD8♡, or NKG2D [13–
`19]. However, the absolute requirement for an extracellular
`spacer domain most likely depends upon the position of the
`target epitope with respect to the target cell surface. CARs
`targeting epitopes toward the proximal end of protein anti-
`gens tend to function well without spacer domains while
`CARs targeting epitopes buried closer to the target cell
`membrane appear to demonstrate improved function when
`a Xexible spacer region is included [20, 21]. These observa-
`tions suggest that there is still a requirement for the empiri-
`cal testing of CARs with varying extracellular domains in
`order to clearly identify the optimal receptor for each target
`antigen.
`In a similar manner, various transmembrane regions have
`also been employed in CARs including those derived from
`CD28, CD3♩, CD8, CD4, or Fc♦RI♤ [15, 17, 19, 22, 23].
`While CARs bearing any of these protein domains have
`been shown to function in terms of down-stream signaling
`after antigen ligation, the structural and biochemical impact
`of the transmembrane domain upon the CAR remains
`largely unknown. Recent studies by Bridgeman et al. [24]
`investigating CARs employing the CD3♩ transmembrane
`domain indicate that the biochemical interactions that occur
`between the wild-type CD3♩ transmembrane domain and
`other components of the endogenous TCR/CD3 complex are
`important for the optimal activity of the CD3♩ CAR. These
`studies suggest that the over-expression of the CD3♩ con-
`taining CAR permits an increased level of endogenous TCR
`expression on the transduced T cell since the availability of
`CD3♩ protein represents the rate-limiting factor controlling
`cell surface TCR/CD3 complex expression in T cells; this
`translates to an increased responsiveness to mitogenic anti-
`CD3 antibody stimulation. Whether this confers an advanta-
`geous or deleterious eVect upon T cells engrafted with CD3♩
`transmembrane domain containing CARs in vivo remains
`unknown. However, there is a general lack of understanding
`of the speciWc structural and biochemical nature of the
`majority of CARs and, in particular, the speciWc eVects that
`the range of extracellular and transmembrane domains
`impart upon the endogenously expressed TCR in the CAR-
`expressing T cell.
`
`Basic CAR structure: signaling domains
`
`Fig. 1 The structure of a CAR. CARs consist of a scFv derived from
`an antibody variable heavy (VH) and variable light (VL) fragments, a
`hinge region linking the scFv with a transmembrane (TM) domain and
`signaling domain, most often CD3♩ or Fc♦RI♤. CL—constant region of
`light immunoglobulin chain, CH1–CH3—constant region of heavy
`immunoglobulin chain, dotted line corresponds to disulWde bound
`
`In keeping with the plethora of options available for the
`extracellular domains of the CAR, there has been an ever-
`increasing range of intracellular signaling domains shown
`to function in the context of a CAR. Initial studies
`employed a single signaling domain, most commonly
`
`123
`
`UPenn Ex. 2035
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Cancer Immunol Immunother (2012) 61:953–962
`
`955
`
`derived from the CD3♩ chain or the ♤ chain of the high-
`aYnity IgE Fc receptor (Fc♦RI), referred to as FcR♤
`(Fig. 1). CARs consisting of either signaling domain
`proved capable of activating T cell eVector function includ-
`ing target cell cytotoxicity and cytokine release in response
`to target antigen binding [25]. However, head to head in
`vivo studies suggested that mouse T cells engrafted with
`CD3♩ CARs demonstrated improved anti-tumor activity as
`compared to those bearing the FcR♤ signaling domain [26].
`CARs bearing signaling domains from a single receptor
`have been subsequently termed “Wrst-generation” receptors.
`The modular nature of the CAR lends itself to further engi-
`neering
`involving combining
`intracellular
`signaling
`domains that can potentially increase the potency of the
`CAR. The full activation of T cells requires multiple sig-
`nals, and it is clear that signaling from these Wrst-generation
`CARs only supplied the so-called “signal one” that could
`drive T cell eVector functions, but in the absence of further
`signals (signal two or co-stimulation), the T cells were
`unable to fully engage its eVector machinery and therefore
`undergoes apoptosis.
`The most studied co-stimulatory pathway has been liga-
`tion of T cell CD28 receptors with the B7 family of ligands.
`Ligation of CD28 on CAR T cells through the expression of
`B7 co-stimulatory ligands on target cells [15, 27, 28] or co-
`expression of the CD28 molecule together with the scFv
`and CD3♩ domains of the CAR [29] was shown to lead to
`proliferation of CAR-modiWed T cells and enhanced anti-
`tumor activity. Exploiting the modular nature of the CAR
`enabled the engineering of “second-generation” CARs
`
`where the signaling domains of two receptors were
`expressed within one CAR (Fig. 2). The fusion of CD28
`and CD3♩ into a single CAR has proven to be eVective in
`permitting the repeated antigen stimulation and prolifera-
`tion of CAR-expressing T cells in the absence of exogenous
`co-stimulatory ligands, maintaining the ability to demon-
`strate both antigen-speciWc cytolysis and to secrete IL-2 in
`vitro [16, 28, 30, 31]. Recent Wndings have also shown that
`CARs containing the CD28 domain could also rescue acti-
`vated T cells from antigen-induced cell death (AICD) [32]
`and enhance the resistance of CAR-modiWed T cells to reg-
`ulatory T cells (Treg) activity [33].
`Experiments in animal models conWrmed these in vitro
`observations and demonstrated that T cells engrafted with
`CARs containing the CD28 signaling domain had pro-
`longed survival, produced higher levels of cytokines, pro-
`liferated vigorously, and mediated enhanced anti-tumor
`eVect compared to T cells expressing the conventional
`CARs [34–36]. These reprogrammed T cells have also been
`observed to proliferate robustly, even without administra-
`tion of exogenous IL-2 [37].
`Several other co-stimulatory receptors have been studied
`including OX40 (CD134) [38], 4-1BB (CD137) [39],
`DAP10, and ICOS [31, 40]. Although all these constructs
`showed antigen-dependent cytotolysis in vitro, only 4-1BB
`CARs showed enhanced persistence and anti-tumor activity
`in vivo [39, 41]. All other second-generation CARs failed
`to produce suYcient amounts of IL-2 to promote T cell pro-
`liferation in the absence of exogenous co-stimulation. How-
`ever, when the exogenous B7 co-stimulation was delivered,
`
`Fig. 2 Clinical protocol of
`adoptive transfer of CAR-
`expressing T lymphocytes. T
`lymphocytes are collected from
`a cancer patient and retrovirally
`transduced with CAR genes,
`then expanded ex vivo to large
`numbers and infused back into
`the patient. To facilitate engraft-
`ment of CAR-modiWed T lym-
`phocytes in vivo, the patient is
`given a lymphodepletive chemo-
`therapy regimen prior to infu-
`sion and cytokine support post-
`infusion
`
`DNA
`encoding CIR
`
`Viral vector
`
`Gene
`modification
`
`T cell
`expansion
`
`Isolation of T cells
`
`Infusion of modified
`T cell
`
`Conditional
`chemotherapy
`
`IL-2 supportive
`therapy
`
`123
`
`UPenn Ex. 2035
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`956
`
`Cancer Immunol Immunother (2012) 61:953–962
`
`transgenic T cell expressing these CARs produced IL-2 and
`showed improved proliferation as compared to T cells
`engrafted with CD3♩ alone CARs under the same condi-
`tions [31].
`This Wnding paved the way for the engineering of “third-
`generation” CARs that contain CD28 and one other co-
`stimulatory domain, most often OX40 or 4-1BB, fused with
`the activation domain (Fig. 2). These CARs were found to
`drive equivalent levels of cytolysis as observed in second-
`generation CARs [38, 39, 42] but drove higher and more
`prolonged levels of cytokine production and cell prolifera-
`tion [38].
`The hierarchy of CAR T cell co-stimulation, reXected in
`the sequence of CAR co-stimulatory and activation
`domains, appears to directly translate into in vivo anti-
`tumor eYcacy with second-generation CARs being supe-
`rior to Wrst-generation and third-generation outperforming
`the second-generation. However, the latest Wnding by
`Cheadle and colleagues suggests that functional activity of
`CAR-expressing T cells may be dependent not only upon
`the optimal combination of CAR signaling moieties but
`also upon the endogenous physiological receptor interac-
`tions provided by target cells [43]. Murine T cells
`engrafted with a Wrst-generation CD19-speciWc CAR were
`found to produce IL-2 following co-culture with CD19+
`B-cell lymphoma cells independently of CD28 receptor
`ligation, with the production of IL-2 driven by endogenous
`CD2 receptor activity. This Wnding suggests that the opti-
`mization of CAR signaling domains according to target
`cells could possibly be the way forward in achieving the
`best anti-tumor eVect rather than use of a universal CAR
`against all target cells.
`As previously mentioned, the majority of engineered
`CARs contain the CD3♩ signaling moiety that exploits TCR
`proximal kinases signaling pathway. CARs based on alter-
`native signaling domains utilizing TCR distal signaling
`events have also been engineered. These CARs include sig-
`naling domains derived mainly from the protein tyrosine
`kinase (PTK) Syk family and are capable of triggering anti-
`gen-dependent T cell activation with IL-2 production and
`target cell lysis [44]. Although the eYciency of Syk-based
`CARs is not superior to CD3♩-based CARs, their main
`potential advantage is the ability to bypass TCR proximal
`signaling events, which are often defective in cancer
`patients, and directly trigger downstream signal transduc-
`tion machinery.
`
`Initial clinical trials with CAR-modiWed T cells
`
`Clinical protocols for CAR T cell therapy usually involve
`the isolation of autologous T lymphocytes from cancer
`patients, their ex vivo modiWcation with the CAR genes
`
`123
`
`followed by large-scale expansion. These genetically modi-
`Wed T lymphocytes are then infused back into the patient,
`usually with administration of IL-2 to support their viability
`and function. To facilitate the engraftment and persistence
`of CAR-modiWed T cells, cancer patients are also pre-con-
`ditioned prior to the cell transfer (Fig. 2).
`A number of Wrst-generation CARs showed some suc-
`cess in pre-clinical models and subsequently entered phase
`I clinical trials. The tumors targeted included ovarian
`cancer [45], renal cell carcinoma [46, 47], neuroblastoma
`[48, 49], B-cell non-Hodgkin lymphoma (NHL), and man-
`tle cell lymphoma (MCL) [50]. Unfortunately, despite
`promising preclinical results, the majority of these initial
`CAR T cell trials showed little evidence of anti-tumor
`activity with limited activation, persistence, and homing to
`tumor sites being the main barriers. However, responses
`were seen in a small number of trials with some anti-tumor
`responses being reported in B-cell lymphoma patients
`treated with ♡CD20-CD3♩ T cells, in which two patients
`were reported to maintain a previous complete response,
`one patient achieved a partial response and four patients
`achieved stable disease [50]. Further anti-tumor responses
`were also seen in patients with neuroblastoma treated either
`with CAR T cells targeting L1-cell adhesion molecule
`(L1CAM) [48] or diasialo-ganglioside (GD2) antigen [49].
`To date, though, the GD2 targeting trail is the one in which
`Wrst-generation CAR T cells have mediated a durable, com-
`plete response in cancer patients [51].
`Although clinical studies of Wrst-generation CARs
`produced rather modest clinical outcomes, they established
`the feasibility and safety of CAR-adoptive transfer therapy.
`This in turn paved the way for further improvements of
`CAR signaling capacity and resulted in the engineering of
`second- and third-generation CARs. These improved CARs
`are capable of delivering superior strength and quality acti-
`vation signal, resulting in increased proliferation, cytokine
`release, and eVector functions of CAR-modiWed T cells in
`vitro and in pre-clinical models. Both the second- and the
`third-generation CARs are now entering the clinical arena,
`and their therapeutic potential is under intense investigation
`(Table 1). Encouragingly, the Wrst clinical reports from
`these studies have been published and have shown some
`promising results [52–56].
`In a pilot clinical study with ♡CD19.4-1BB.CD3♩ T cells
`in three patients with advanced chronic lymphocytic leuke-
`mia (CLL), two complete remissions and one partial
`response that has been ongoing 10 months after the treat-
`ment has been achieved [52, 54]. The engineered T cells
`were found to expand over 3-logs in these patients, inWl-
`trated and lysed tumor cells, and persisted at high levels for
`over 6 months. Interestingly, a fraction of these cells dis-
`played a memory T cell phenotype, suggesting the potential
`for preventing tumor relapses.
`
`UPenn Ex. 2035
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Cancer Immunol Immunother (2012) 61:953–962
`
`957
`
`+Trial suspended temporarily due to re-location of cell processing facility
`*Trials due to open
`Williams Medical Center, Providence, Rhode Island, USA, UCL University College London, UK, UP University of Pennsylvania, Philadelphia, USA, CHMAN Christie Hospital, Manchester, UK
`MDACC M.D. Anderson Cancer Center, Houston, Texas, USA, MSKCC Memorial Sloan-Kettering Cancer Center, New York, USA, NCI National Cancer Institute, Bethesda, USA, RWMC Roger
`eBCM Baylor College of Medicine, Houston, Texas, USA, COH City of Hope Medical Center, Duarte, California, USA, FHCRC Fred Hutchinson Cancer Research Center, Seattle, Washington, USA,
`dLV lentiviral, RV ♤-retroviral, SB sleeping beauty transposon/transposase DNA plasmid system
`cEBV Epstein-barr virus-speciWc CAR-modiWed T cells
`cord blood transplantation
`bB-ALL B-lineage acute lymphoblastic leukemia, B-NHL B-lineage non-Hodgkin’s lymphoma, CLL chronic lymphocytic leukemia, HSCT hematopoietic stem cell transplantation, UCBT umbilical
`aCEA carcinoembryonic antigen, GD2 diasialoganglioside, Her2 human epidermal growth factor receptor, PSMA prostate-speciWc membrane antigen
`
`NCT00881920
`BCM
`NCT01109095
`BCM
`NCT00902044
`BCM
`NCT00889954
`BCM
`NCT00673322
`RWMC
`NCT00673829
`RWMC
`NCT01140373
`MSKCC
`NCT00664196
`RWMC
`NCT00085930
`BCM
`NCT00621452
`FHCRC
`NCT01318317*
`COH
`CHMAN
`NCT01493453+
`NCT01362452*MDACC
`MDACC
`NCT00968760
`NCI
`NCT01087294
`UP
`NCT00891215
`BCM
`NCT00840853
`NCI
`NCT00924326
`UCL
`NCT01195480*
`BCM
`NCT00709033
`BCM
`NCT00608270
`BCM
`NCT00586391
`MSKCC
`NCT01416974
`MSKCC
`NCT01044069
`MSKCC
`NCT00466531
`
`I
`
`I/II
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I/II
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I/II
`I/II
`
`I
`
`I
`
`I
`
`I
`
`I
`
`I
`
`RV
`RV
`RV
`RV
`RV
`RV
`RV
`RV
`RV
`Electroporation
`LV
`RV
`Electroporation/SB plasmid
`Electroporation/SB plasmid
`RV
`LV
`RV
`RV
`RV
`RV
`RV
`RV
`RV
`RV
`RV
`
`CD3♩/CD28 versus CD3♩
`CD3♩/CD28 (EBV)
`CD3♩/CD28
`CD3♩/CD28
`CD3♩/CD28
`CD3♩/CD28
`CD3♩/CD28
`CD3♩
`CD3♩ (EBV)
`CD3♩/CD137/CD28
`CD3♩
`CD3♩
`CD3♩/CD28
`CD3♩/CD28
`CD3♩/CD28
`CD3♩/4-1BB versus CD3♩
`CD3♩/CD28
`CD3♩/CD28
`CD3♩ EBV
`CD3♩/CD28 versus CD3♩ (EBV)
`CD3♩/CD28 versus CD3♩
`CD3♩/CD28 versus CD3♩
`CD3♩/CD28
`CD3♩/CD28
`CD3♩/CD28
`
`First and second
`Second
`Second
`Second
`Second
`Second
`Second
`First
`First
`Third
`First
`First
`Second
`Second
`Second
`First and second
`Second
`Second
`First
`First and second
`First and second
`First and second
`Second
`Second
`Second
`
`B-NHL and B-CLL
`Glioblastoma
`Advanced osteosarcoma
`Lung malignancy
`Colorectal carcinoma
`Breast cancer
`Prostate cancer
`Prostate cancer
`Neuroblastoma
`Mantle cell lymphoma/indolent B-NHL
`B-NHL
`B-NHL
`B-lineage lymphoid malignancies post-UCBT
`B-lymphoid malignancies post-HSCT
`B-cell leukemia/CLL/B-NHL
`Lymphoma/leukemia
`ALL post-HSCT
`Lymphoma/leukemia
`ALL
`Advanced B-NHL/CLL
`B-NHL/CLL
`Lymphoma/leukemia (B-NHL)/CLL
`Leukemia
`B-ALL
`CLL
`
`Kappa light chain
`Her2/neu
`Her2/neu
`Her2/neu
`CEA
`CEA
`PSMA
`PSMA
`GD2
`CD20
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`CD19
`
`Centere
`
`gov identiWer
`Clinical trial.
`
`Phase
`
`CAR gene transferd
`
`CAR endodomainc
`
`CAR generation
`
`Diseaseb
`
`targeteda
`Antigen
`
`Table1Representative list of CAR T cell clinical trials (generated from a review of the clinical trials.gov database)
`
`123
`
`UPenn Ex. 2035
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`958
`
`Cancer Immunol Immunother (2012) 61:953–962
`
`Although ♡CD19.4-1BB.CD3♩ T cells produced
`signiWcant therapeutic eVect, their potent cytotoxic
`activity led to the delayed development of potentially
`life-threatening tumor lysis syndrome in one of the
`patients [58]. The patient developed the syndrome
`22 days following the Wrst infusion of ♡CD19 T cells
`showing elevated levels of uric acid, lactate dehydroge-
`nase, and acute kidney injury. In two patients, elevated
`levels of certain serum cytokines were also detected;
`however, no classical symptoms of cytokine storm were
`observed [52, 54].
`Also, T cells modiWed with second-generation CD19-
`speciWc CARs containing a CD28 costimulatory endodo-
`main demonstrated enhanced expansion and persistence in
`lymphoma patients, compared to CAR T cells lacking this
`endodomain [57]. Infusions of ♡CD19.CD28.CD3♩ T cells
`were well tolerated, and no acute toxicity was seen.
`However, recently two serious adverse events were
`reported which highlight the need for caution while using
`activation potent second- and third-generation CARs. One
`occurred following the adoptive transfer of T cells modiWed
`with a CD19-speciWc second-generation CAR (♡-CD19-
`CD28-CD3♩) into a patient with advanced CLL [58], and
`the second in a patient with metastatic colon cancer treated
`with a third-generation CAR targeting HER-2/neu (♡-HER2/
`neu.CD28.4-1BB.CD3♩) [59].
`The treatment-related death in the anti-CD19 T cell
`recipient occurred shortly after cyclophosphamide-based
`chemotherapy and infusion of the anti-CD19 T cells. The
`patient developed acute sepsis, persistent fever, hypoten-
`sion, acute renal failure and expired 44 h following infusion
`of the T cells [58]. Although the cause of the patient’s death
`remains uncertain, the subsequent investigation has impli-
`cated an underlying infection which was exacerbated due to
`the cyclophosphamide-based pre-conditioning regime. Fol-
`lowing extensive re-appraisal, the trial has now been
`reopened and two subsequent patients have been treated
`with only minor modiWcations to the protocol [55]. These
`include the single infusion of CD19-reactive T cells follow-
`ing cyclophosphamide chemotherapy being split into two,
`with a 24-h delay between infusions. Both patients treated
`tolerated the treatment well, and no notable toxicity was
`seen.
`In contrast, the second fatality associated with a
`HER-2/neu-speciWc CAR seems to be a direct eVect of
`administration of large numbers of HER-2/neu-reactive
`T cells which
`targeted
`low
`levels of HER-2/neu
`expressed on pulmonary endothelium [59]. Elevated lev-
`els of IFN-♤, GM-CSF, TNF-♡, IL-6, and IL-10 observed
`in the patient serum following therapy were consistent
`with a “cytokine storm” resulting from the massive “oV-
`target” activation of infused T cells inWltrating pulmo-
`nary endothelium. Within minutes following infusion,
`
`123
`
`the patient developed acute pulmonary toxicity followed
`by cardiac arrest and died 4 days later.
`
`Challenges of CAR-modiWed T cell therapy
`
`Recent clinical trials have revealed several potential pitfalls
`to CAR-modiWed T cells that potentially still bar the way to
`an eVective CAR therapy. A major concern is the risk of
`“oV-target” toxicity, resulting in autoimmune reaction
`against self-tissues [46, 59]. This is because tumor-
`restricted antigens are very rare, and the majority of tumor-
`associated antigens that are targets for immunotherapy are
`shared with normal tissues. Although physiologically these
`antigens are usually expressed at low levels in normal tis-
`sue and are over-expressed in malignant cells, the CAR
`response is highly speciWc and can target both “on-target”
`and “oV-target” antigens. This is especially the case with
`second- and third-generation CARs containing potent com-
`bination of signaling and costimulatory molecules that have
`the potential to respond to low levels of targets physiologi-
`cally expressed, resulting in the generation of powerful
`activation signal, leading to a lethal “cytokine storm” [59].
`To adopt the lesson learned from donor lymphocyte
`infusion following hematopoietic stem cell transplantation,
`which showed dose-dependent toxicity [60], the autoim-
`mune reaction could be avoided or at least controlled by
`using split infusions of lower doses of therapeutic T cells.
`This has been already shown to abrogate the acute toxicity
`seen in the anti-CD19.CD28.CD3♩ CAR study in which,
`following a fatality, the large dose of 1010 CAR-modiWed T
`cells was split into two infusions and did not produce any
`notable toxicity in two subsequent patients [55].
`Interestingly, an autoimmune reaction was observed in
`clinical studies using a CAIX-speciWc CAR [46] and a
`HER-2/neu-speciWc CAR [59], but was not seen for a CAR
`targeting L1CAM on neuroblastoma cells [48], which is
`known to be also expressed on adrenal medulla and sympa-
`thetic ganglia, suggests that the aYnity with which the scFv
`binds target antigen could be critical in inducing an autoim-
`mune response. High-aYnity CARs are possibly more sen-
`sitive to low levels of antigen where binding generates a
`more potent activation signal compared to low aYnity
`CARs, which may require higher antigen levels to respond.
`Therefore, using CARs displaying a lower aYnity to their
`target antigen may be advantageous in preventing autoim-
`mune responses.
`Another way to obviate a potential autoimmune reaction
`is to include a suicide gene in CAR-modiWed T cells that
`could be activated in the event of acute toxicity leading to
`elimination of
`the CAR-modiWed T cells. Such an
`approach, involving the herpes simplex virus thymidine
`kinase (HV-TK) suicide gene, has already been proven to
`
`UPenn Ex. 2035
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Cancer Immunol Immunother (2012) 61:953–962
`
`959
`
`work in donor lymphocyte infused in patients after hemato-
`poietic stem cell transplantation to prevent graft-versus-
`host disease (GvHD) [61]. HV-TK transgenic cells are sen-
`sitive to the antiviral drugs ganciclovir and acyclovir,
`which upon phosphorylation by HV-TK interfere with
`DNA polymerase leading to cell death. This approach has
`now been utilized in CAR-modiWed T cells. Park and
`colleagues engineered CAR T cells targeting L-1CAM con-
`taining the HV-TK suicide gene and reported no HV-TK-
`related toxicity in patients with metastatic neuroblastoma
`[48]. However, the main shortcoming of the application of
`this HV-TK suicide system in humans is its potential
`immunogenicity due to the viral origin [61, 62]. This may
`result in a quick clearance of infused therapeutic T cells,
`limiting their therapeutic eYcacy. Recently, an anti-CD19
`CAR containing an alternative suicide gene, an inducible
`caspase-9 (icasp9), has also been generated [63]. The
`icasp9 is induced by the administration of a small-molecule
`dimerizer that merged two non-functional icasp9 molecules
`in the active enzyme. Activation of icasp9 was seen to rap-
`idly induce apoptosis of CAR-expressing T cells in vitro
`and in vivo. The main advantage of icasp9-based suicide
`system is the fact that icasp9 is of human origin, and its
`expression in CAR T cells is unlikely to induce unwanted
`immune response.
`Another controversial issue is related to the clinical
`safety of retroviral vectors encoding CARs that are used to
`transfect human T cells [64]. These vectors stably integrate
`into the host genome creating the risk of insertional muta-
`genesis that may lead to malignant transformation [65].
`Although no such event has been observed in patients
`treated with retroviral gene-modiWed T cells during the past
`20 years [66, 67], all possible precautions should be taken
`to minimize the risk. Thus, safer systems for the transfer of
`CAR genes are being developed including the use of lentiv-
`iral vectors [52, 54, 68, 69] and electroporation of RNA
`[70–73].
`The potential immunogenicity of the CAR molecules
`themselves is another potential limitation threatening CAR
`therapy. In cases where this occurs, most often the immune
`response is directed against the extracellular antigen-
`recognition domain that is usually derived from mouse
`monoclonal antibodies [45, 74]. Although, putative immune
`response against
`retroviral vector-encoded epitopes
`expressed by CAR-modiWed T cells has also been observed
`[74]. The immune rejection of CAR-modiWed T cells leads
`to their destruction, thereby reducing survival and anti-
`tumor eVect. This limitation, however, can be avoided by
`using humanized scFv’s or scFv’s derived from human
`monoclonal antibodies [75, 76]. Also, alternatives to viral
`transfection gene transfer techniques and modiWcation of
`clinical protocols to include immunosuppressive condition-
`
`ing regimens may be beneWcial to fully exploit the therapeu-
`tic potential of CAR-modiWed T cells.
`Finally, the use of autologous T cells requires CAR-
`modiWed T cells to be generated for each individual
`patient that is a logistically and economically demanding
`process. Therefore, much eVort is currently being directed
`to create universal eVector T cells that could serve as “oV-
`the-shelf” immunotherapeutic to be administered to
`patients irrespective of their MHC type. To this end, allo-
`geneic T cells have been the obvious candidate but their
`use is threatened by host-versus-graft (HvG) rejection and
`graft-versus-host (GvH) response [77]. However, the
`recent in vivo research by Marcus and colleagues showed
`that the GvH activity of allogeneic CAR T cells can be
`harnessed by some T cell traYcking modulators [78],
`potentially allowing exploitation of the full therapeutic
`eVect of allogeneic CAR T cells.
`
`Concluding remarks
`
`The adoptive transfer of CAR-engineered T cells represents
`a valuable and attractive therapeutic strategy that in the
`near future has the potential to give new prospects to cancer
`immunotherapy. While clinical trials with the Wrst-genera-
`tion CARs showed rather modest anti-tumor eVect, these
`trials established the feasibility and safety for the therapy
`and more importantly paved the way for further improve-
`ments of CAR function. This resulted in the engineering of
`second- and third-generation CARs capable of delivering
`greater strength and quality activation signals directly
`translating into superior anti-tumor properties of CAR-
`engineered T cells. Whether or not these second- and third-
`generations CARs will fulWll their clinical expectations is
`too early to speculate. These CARs are just entering the
`clinical arena, and results of their intense clinical assess-
`ment are much awaited in the Weld. Importantly, the further
`understanding of T cell subsets with particular respect to
`optimal T cell phenotypes for engraftment will impact upon
`future CAR T cell approaches [79–82]. So far, these Wrst
`clinical reports give some promise for tumor treatment but
`also emphasize challenges to be undertaken to establish
`CAR T cell approach as an eVective and safe cancer
`therapy.
`
`Acknowledgments DEG, REH, and DGR were funded by Cancer
`Research UK. This work was also supported by the EU FP6 pro-
`gramme ATTACK, FP7 training Network ATTRACT and the Kay
`Kendall

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