throbber
8#-w~t NIH Public Access
`~ tf:i,,-w Author Manuscript
`u 1111 11 m, , r r · J
`Published in final edited form as:
`Cancer Res.2011 May I; 71(9): 3175-3181. doi: I 0.1158/0008-5472.CAN-10-4035 .
`
`11l h • 11 I t _,
`
`II
`
`II I 11
`
`O,t:-1-fEf>..~
`
`,
`
`, ,
`
`r 1 ,,
`
`I,\~ j"""li\ t
`EXHIBIT N0~05Z.
`~1-~MG
`
`Considerations for the Clinical Application of Chimeric Antigen
`Receptor (CAR) T Cells: Observations from a Recombinant DNA
`Advisory Committee (RAC) Symposium June 15, 2010
`
`Hildegund C.J. Ert1 1, John Zaia2, Steven A. Rosenberg3, Carl H. June4, Gianpietro Dotti5,
`Jeffrey Kahn6, Laurence J.N. Cooper7, Jacqueline Corrigan-Curay8 , and Scott E. Strome9
`1Wistar Institute, Philadelphia, PA
`
`2Beckman Research Institute, City of Hope, Duarte, CA
`
`3National Cancer Institute, NIH
`
`4University of Pennsylvania, Philadelphia PA
`
`5Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
`
`6Center for Bioethics, University of Minnesota, MN
`
`7Division of Pediatrics, MD Anderson Cancer Center, Houston TX
`
`80ffice of Biotechnology Activities, Office of the Director, NIH
`
`9Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, MD
`
`Abstract
`
`T cells which are genetically modified to express single-chain chimeric antigen receptors (CARs)
`have shown promise in early cancer immunotherapy clinical trials. Unfortunately, two recent
`deaths in cancer patients treated with CAR-T cells have created some uncertainty on how to best
`mitigate patient risk, while continuing to advance this very promising therapeutic avenue. In order
`to address these concerns, the Recombinant DNA Advisory Committee (RAC) held a symposium,
`the objectives of which were to first review the reported treatment-associated toxicities and
`second, to discuss methods for improving safety and efficacy. This report highlights the issues
`raised as part of this discussion with a specific focus on protocols infusing CAR-T cells. Because
`this was not a consensus conference, the opinions described should not be construed to represent
`those of any individual RAC member, the RAC as a body, conference participants, NIH or the
`FDA.
`
`Keywords
`
`Chimeric antigen receptor; T-cell therapy; gene therapy; CD 19; adverse events
`
`Corresponding author: Scott E. Strome, MD, Department ofOtorhinolaryngology-Head and Neck Surgery, Cniversity of Maryland,
`16 South Eutaw Street: Suite 500. Baltimore, MD, 21201-168, CSA, sstrome(':llsmail.umaryland.edu.
`Conflict of Interest: Dr. Strome is the cofounder and a major stockholder in Gliknik, Inc., a biotechnolob'Y company. He also receives
`royalties through the Mayo Clinic College of Medicine for the Ii censure of IP relating to 4-1 BB (CD 13 7) and B7-H I (PD-LI).
`
`z
`J:
`I
`""CJ
`)>
`)>
`
`C --=s-
`
`0
`
`""I s::
`
`Dl
`::::::,
`C
`Cl)
`£.
`"S.
`
`z
`J:
`~
`
`I
`
`)>
`
`C --=s-
`
`0
`
`""I s::
`
`Dl
`::::::,
`C
`Cl)
`0
`
`""I -5· -
`
`z
`J:
`I
`""CJ
`)>
`)>
`
`C --=s-
`""I s::
`
`0
`
`Dl
`::::::,
`C
`(/)
`
`0 ~(cid:173)"C -
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 1
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C ....
`::,-
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C ....
`::,-
`0 -,
`s:
`
`Q)
`~
`
`C en
`(") -,
`....
`-5·
`
`z
`I
`I
`-0
`)>
`)>
`
`C ....
`::,-
`0 -,
`s:
`
`Q)
`~
`
`C en
`(") -,
`....
`-5·
`
`Ertl et al.
`
`Evolution of CARs
`
`Page 2
`
`Following appropriate preconditioning regimens that deplete circulating lymphocytes, the
`adoptive transfer of tumor infiltrating lymphocytes (TILs) into patients with metastatic
`melanoma leads to objective clinical responses in select individuals (Supplemental
`references (SR) 1,2). Building on this approach, recent studies suggest that autologous T
`cells, genetically engineered to express tumor associated antigen (T AA)-specific TC Rs, can
`replace TILs in certain clinical settings. (I) Importantly, tumor regressions are observed
`even under conditions of widely metastatic and bulky disease, and in patients who have
`failed both prior surgical extirpation and medical therapy. Unfortunately, the potential for
`widespread use of adoptive transfer strategies using Tl Ls or T cells with recombinant TC Rs
`is limited (SR#3 ).
`
`In order to overcome the historic problems associated with the cellular targeting of HLA
`restricted T AA, Eshhar et al. developed a strategy to redirect T-cell specificity using
`Chimeric Antigen Receptors (CARs) or T-bodies (Figure I). First generation vectors for the
`production of CAR-T cells contain the heavy and light chain immunoglobulin (lg) variable
`regions, fused as a single chain to the epsilon, gamma or zeta signaling sequences of the T
`cell receptor (TCR) or the signaling region of the Fey domain. (2-8) T cells expressing such
`first generation CARs recognize surface T AAs, independent of HLA restriction, but cannot
`recognize intracellular T AAs (Table I). Early clinical experiences with I st generation
`CAR-T cells demonstrated that (a) the survival of adoptively transferred CAR-T cells was
`limited in cancer patients and (b) few objective anti-tumor responses were observed. (8-10)
`
`To enhance survival and/or increase proliferation of transferred CAR-T cells, investigators
`have incorporated signaling moieties from costimulatory molecules including CD28 (87-1 ),
`CD 134 (OX40), and CD 13 7 ( 4-1 88), alone (2nd generation) or in sequence (3rd
`generation). ( I 1-15) A recent study comparing simultaneously infused CD 19-specific CAR(cid:173)
`T cells with or without the CD28 signaling sequence indeed demonstrates improved survival
`of CD28-modified cells .. (16) Additionally, combining CD28 with 4-188 signaling
`sequences further promotes engraftment of CAR-T cells. ( 17, 18) As an alternative to
`incorporating costimulatory moieties into the vector, some investigators express first
`generation CARs in T cells specific to endogenous viral antigens e.g. Epstein Barr virus
`(EBY). In this setting, antigen recognition by the CAR provides TCR ~ signaling, while
`recognition of processed EBY peptides in the context of appropriate HLA molecules by the
`physiologic urncR, allows for intrinsic costimulation.( 19)
`
`Focus Points for Future Consideration
`
`Reported serious adverse events (SAE) following CAR-T cell transfer
`
`At least 40 trials using CAR-T cells for treatment of cancer have been registered with the
`NIH Office of Biotechnology Activities (OBA) and undergone NIH RAC review and at least
`120 subjects have been dosed across all trials (Supplemental Table I). (20) Overall, most
`SA Es that were viewed as possibly related to transfer of CAR-T cells have been mild, self(cid:173)
`limited and occurred shortly after infusion (Supplemental Table 2). Unfortunately, 2 patients
`died shortly after adoptive transfer of CAR-T cells. (21, 22)
`
`Cancer Res. Author manuscript; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 2
`
`

`

`z
`::t
`~
`)>
`
`I
`
`C -::::r
`0 -,
`s: Q)
`:::,
`C en
`"C -
`C')
`:::::!.
`
`z
`::t
`I
`""C
`)>
`)>
`
`C -::::r
`0 ..,
`s: Q)
`:::,
`C en
`C') -,
`
`-6' -
`
`z
`::t
`~
`)>
`
`I
`
`C -::::r
`0 ..,
`s: Q)
`:::,
`C en
`C') ..,
`"§:
`
`Ertl et al.
`
`Page J
`
`Following RAC review of the death in the HER2-CAR-T cell trial at its December 2009
`meeting [http /, oba.od.nih.gov/rdna _ rac 'rac past meetings _ 2000.htrnl] the Committee
`recommended holding a safety symposium to review clinical trials employing CAR-T cells
`and to then formulate a report to both facilitate subsequent RAC and Institutional Biosafety
`Committee (IBC) reviews of these trials and to assist investigators in designing future
`studies. A planning committee, consisting of RAC members, leading investigators in this
`field, and OBA staff, identified key components of these trials based on their potential
`impact on patient safety and their role in therapy. These issues were discussed in a panel
`format and are summarized under the heading "Points to Consider in the Design and
`Implementation of Clinical Trials using CAR-T cells." A paradigm for considering how to
`incorporate these considerations into clinical trial design is provided in Table 2 and
`Supplemental Tables 3-5.
`
`Points to Consider in the Design and Implementation of Clinical Trials
`using CAR-T cells
`
`1. Unwanted On-Target Effects ofCAR-T Cells
`
`Recognition of antigen expressed on non-tumor cells by CAR-T cells is emerging as the
`major risk factor of CAR-T-cell transfer. Such recognition may become manifest as: (a)
`immediate toxicity and (b) late or sustained toxicity resulting from long term depletion of
`cells with important homeostatic functions.
`
`A. Mitigating the potential for early toxicity-A feared complication after infusion of
`CAR-T cells is their massive activation leading to the research participant's death. This is
`especially of concern with CARs directed against untested and/or endogenously prevalent
`T AAs, as T cells with high avidity receptors can respond to cells that express their targets at
`levels that are currently too low for detection by conventional means. One approach to limit
`such toxicity is an inter-patient (and sometimes intra-patient) dose-escalation scheme.
`Alternatively one could conduct the initial dose escalation with I st generation CAR-T cells
`without conditioning or cytokine supplementation under the expectation that stimulation of
`these adoptively transferred T cells may be suboptimal, although this is not yet proven. Once
`the I st generation CAR-T cells are shown to be safe, 2nd (or perhaps 3rd) generation CAR-T
`cells, with a potential for more pronounced responses and prolonged persistence, could be
`explored in combination with supplementary treatments. (23) An important caveat lo this
`statement is that it is currently unclear whether lack of toxicity using I st generation CAR-T
`cells will translate into a similar safety profile for 2nd or 3rd generation CAR-T cells
`combined with lymphodepletion and/or cytokine support. As an alternative approach, if 2nd
`or 3rd generation CARs targeting a new antigen are evaluated without previous experience
`with a I st generation CAR, a very conservative dose escalation strategy should be adopted
`(see: Starting dose ofCAR-T cells in phase I trials).
`
`The question of whether the co-expression of conditional suicide genes might safeguard
`against some of the potential side effects of non-tumor cell recognition by CAR-T cells was
`discussed. (24) As exemplified by the fatal SAE using HER2-CAR-T cells, the CAR-T cells
`may act within minutes after engagement of their target antigen and symptoms are not
`
`Cancer Res. Author manuscript; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3
`
`

`

`z
`I
`I
`~
`
`► C -::T o -,
`
`~
`!l)
`:::,
`C en
`
`C') -, -a· -
`
`z
`I
`I
`'"O
`►
`
`► C -::T o -,
`
`~
`!l)
`:::,
`C en
`
`C') -, -a· -
`
`z
`I
`I
`~
`
`► C -::T o -,
`
`~
`!l)
`:::,
`C en
`
`C') -, -a· -
`
`Ertl et al.
`
`Page 4
`
`expected until significant damage has occurred. Therefore, inclusion of a suicide gene,
`whose benefit will necessarily take time, was deemed to have limited potential for
`preventing acute toxicity although it may have utility in modulating late toxicities.
`
`A third approach to reduce risks of immediate toxicity is splitting of the T-ccll dose over
`two or more days as is being tested in recipients of CD 19-specific T cells. (21) Using this
`scheme it would be desirable to monitor cytokines or chemokines in the serum as potential
`indicators for toxicity after the first of the split doses. A fourth approach using a very
`conservative dose escalation strategy is discussed below under the heading: Starting dose of
`CAR-T cells in Phase I trials. Importantly, in all trials, pre-clinical studies should carefully
`test for expression of the TCR's target antigen in healthy tissues. Furthermore, the use of
`CART cells against antigens that are widely expressed on non-tumor cells essential for
`important physiological functions should, to the extent feasible, not be chosen as targets
`unless there is convincing preclinical data that their expression are at such low levels that
`on-target toxicity is unlikely to occur.
`
`B. Mitigating the potential for late toxicity-Another risk ofCAR-T cells is the long
`term depletion of cells, which are important for normal human function. For example, CAR(cid:173)
`T cells for treatment of 8-cell malignancies have targeted CD 19 and CD20, markers
`expressed on normal 8 cells. (25) Treatment with CAR-T cells directed against either of
`these antigens has the potential to deplete the patients' 8 cells.(26) Furthermore, unlike the
`u-CD20 monoclonal antibody (mAb), RituxanTM, which has a defined half-life, these CAR(cid:173)
`T cells may potentially survive and function for the life of the patient. The lack of CD 19 or
`CD20 expression on most plasma cells allows the maintenance of physiologic antibody
`levels in the majority of patients following depletion of CD 19+ or CD20- cells ( SR#4 ). In
`addition, it is expected that 8 cells would rapidly be replaced from lymphoid progenitors
`once the CAR-T cells die or become functionally impaired. Finally, even under conditions
`that would allow for long-term persistence of functioning CDI 9- or CD20-specific CAR-T
`cells, resulting in continuous depiction of 8 cells, this could be managed by lg transfer and
`would thus be preferable to a fatal cancer.
`
`A related concern is the potential for CAR-T cells to negatively impact organ function
`secondary to low levels of target antigen expression in a non-tumor site e.g. vascular
`endothelial growth factor receptor 2 (VEGFR2)-specific CARs targeting the tumor
`vasculature (SR#5,6 ). VEGFR2 is expressed on endothelial cells during physiological
`angiogenesis, vasculogenesis, arteriogencsis, and lymphangiogencsis and is needed for
`processes such as wound healing and embryogencsis (SR#7). As such, VEGFR2-spccific
`persisting C AR-T cells could interfere with formation of indispensable vasculature. While
`there is no universal strategy for mitigating these late CAR-T-cell associated toxicities, the
`resultant conditions could potentially be managed medically. Alternatively, risks of long(cid:173)
`term toxicity could be mitigated by the insertion of suicide genes into the CAR-T cells.
`
`2. Effects of Costimulatory Signaling
`Results to date demonstrate that while target cell lysis by I st generation CAR-T cells is
`independent of costimulatory signaling, their proliferation, production of cytokines and up-
`
`Cancer Res. Author manuscript; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 4
`
`

`

`z
`:r:
`I
`"'tJ
`)>
`)>
`
`C: st
`0 ..,
`s: Q)
`
`::,
`C:
`(/J
`C')
`::::!.
`
`"O ....
`
`I
`
`z
`:r:
`~
`)>
`C: ....
`::,-
`0 ..,
`s: Q)
`
`::,
`C:
`~
`"O ....
`::::!.
`
`I
`
`z
`:r:
`~
`C: -::,-
`)>
`0 ..,
`s: Q)
`
`::,
`C:
`(/J
`C')
`::::!.
`
`"O -
`
`Ertl et al.
`
`Page 5
`
`regulation of other effector cell molecules depends on and/or is improved by co-stimulation.
`(27) Furthermore, based on what is understood regarding natural functions of CD28 and
`4-188 in vivo, it is anticipated that CAR-T cells bearing these domains might be more
`resistant to activation-induced cell death (SR#8). Therefore, the majority of participants
`concluded that the incorporation of CD28 and/or 4-188 into CAR constructs offers an
`important potential for therapeutic benefit. Evaluations of other cosignaling moieties, which
`may enhance the proliferation and/or survival of CAR-T cells are appropriate. In addition to
`modifying the CAR to improve T cell engraftment it may also be possible to a priori identify
`T-cell sub-populations with the capacity for sustained proliferation. In fact, there is evidence
`that the starting population ofT cells used for CAR modification may affect their ability to
`survive and expand (SR#9). For example, in non-human primates, effector cos+ T cells
`generated ex vivo from central rather than effector memory populations, enjoy enhanced
`survival following adoptive transfer (SR# I 0). Because the ratios of different T-cell subsets
`varies between patients, selective use of T-cell subsets prior to genetic manipulation may
`provide a more discriminating path to predict the long-term outcome of CAR-T cell transfer.
`Finally, clinical experience with the expression of CARs in non-T cell populations e.g. NK
`cells, is limited. (28) Therefore, special care must be taken with CAR use in T-cell subsets
`and non-T cell lymphocyte populations.
`
`3. Systemic Conditioning
`
`Adoptive transfer of CAR-T cells can be further modified by auxiliary therapies such as
`partial or complete myeloablation to improve preservation of the transferred T-cell
`populations. The rationale behind lymphodepletion prior to T-cell infusion is multifactorial
`and includes, but is not limited to, the elimination of"cytokine sinks," creation of space for
`the expansion of adoptively transferred cells, and removal of suppressor cell populations. In
`fact, depletion of lymphocytes in melanoma patients infused with ex vivo expanded TILs is
`requisite for treatment efficacy and recent studies suggest a correlation between the degree
`of lymphocyte depletion and objective response rates (SR# 11 ). Despite the recognized risks
`of myelosuppression, the majority of the RAC meeting participants felt that administration
`of CAR-T cells that do not carry an endogenous TCR to a persisting virus is unlikely to
`result in significant clinical benefit without prior conditioning.
`
`4. Cytokine supplements
`
`Recombinant soluble cytokines such as IL-2 are administered upon CAR-T cell transfer with
`the expectation that they will promote the expansion and survival of transferred cells. If and
`to what degree cytokines may exacerbate the potential toxicity of 2nd or 3rd generation
`CAR-T cells is uncertain. It is also unclear if 2nd and 3rd generation CAR-T cells need to be
`supplemented with IL-2 either at all or at the high and potentially toxic doses used for TIL
`transfer (SR# 12). For the initial evaluation of CAR-T cells to novel targets, dosing without
`addition of cytokines or with injection of low to moderate amounts of cytokines may be
`prudent and would facilitate differentiation of SA Es induced by CAR-T cells alone from
`those caused by the potential synergy between these drugs. Furthennore, the design of later
`Phase studies employing 2nd and 3rd generation CAR-T cells should consider incorporating
`treatment arms with and without cytokine support. Ongoing studies exploring the use of
`CAR-T cells genetically modified to secrete cytokines such as IL-15 or IL-12 may eliminate
`
`Cancer Res . Author manuscript ; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 5
`
`

`

`z
`::i:::
`I -a
`)>
`)>
`C .....
`-:::,-
`0 -,
`s::
`
`ll)
`::::,
`C:
`(J'J
`C') -,
`"§:
`
`z
`::i:::
`I -a
`)>
`)>
`C .....
`-:::,-
`0 -,
`s::
`
`ll)
`::::,
`C:
`(J'J
`
`C') -, -a·
`.....
`
`z
`::i:::
`I
`~
`)>
`.....
`C:
`-:::,-
`0 -,
`s::
`
`ll)
`::::,
`C
`(J'J
`C') -,
`"§:
`
`Ertl et al.
`
`Page 6
`
`the need for systemic cytokine administration. (29) If and to what degree this approach will
`improve the potential benefit of CAR-T cells or instead pose additional risks, is currently
`unknown.
`
`5. CARs expressed in T cells targeting persisting viruses
`
`Some groups have combined the advantages ofCAR-T cells with those of traditional
`antigen-specific T cells (SR#l3). Specifically, T cells to persisting viruses such as EBY
`have been enriched in vitro and then genetically modified by insertion ofa transgene
`encoding a T AA-specific CAR. In a neuroblastoma trial, 11 individuals were treated
`simultaneously with EBV-CTLs expressing a GD2-specific first generation CAR and
`activated T lymphocytes expressing the same CAR. Only CAR EBV-CTLs showed
`significant in vivo persistence, and four of eight patients with evaluable tumors had evidence
`of tumor necrosis or regressions, including a sustained complete remission."
`Lymphodcpletion did not improve CAR-T cell persistence or clinical outcome. ( 19)
`Potential deleterious effects due to virus-specific CAR-T cells without a costimulatory
`endodomain arc anticipated to be similar to those of other T cells expressing 2nd or 3rd
`generation CARs.
`
`6. Starting dose of CAR-T cells in Phase I trials
`
`Phase I trials are primarily designed to assess safety and feasibility rather than efficacy;
`biological activity and proof of concept are usually secondary aims. Phase I trials for CAR-T
`cells typically start with reduced cell doses which are then gradually escalated. The starting
`dose should be adjusted depending on the type of CAR, i.e .. T cells with a 2nd or yd
`generation CARs should start at a lower dose than those with a I st generation CAR.
`Similarly, transfer of CAR-T cells into partially pre-conditioned patients should commence
`at a lower dose than transfer into non- myeloablated patients. Although one would expect
`that immediate toxicity due to transfer of CAR-T cells may directly correlate with numbers
`of injected cells, late adverse events may be independent of the injected dose due to
`construct-dependent dynamic changes in cell numbers.
`
`Providing reliable guidelines for starting doses ofCAR-T cells is currently not possible. To
`date, clinical trials have employed a fairly wide range of starting doses and some
`investigators dosed according to weight. others according to body surface area (BSA) and
`still others employed flat dosing schedules (Supplemental Table I). The use of unadjusted
`dosing should be avoided and a more uniform dosing scheme such as that based on cells/kg
`should be considered. An open question is whether cells/kg should be based on an ideal
`body weight or actual weight given that weight increases due to obesity may not justify an
`increase in dose (SR#l4).
`
`7. Ethical Considerations
`
`One of the ethical dilemmas facing investigators is the requirement in early phase research
`to design studies that arc safe while at the same time hoping to show biological activity or
`possibly even benefit to the individual subject. many of whom have few if any other
`therapeutic or even palliative options. In response to the unexpected death in the trial
`infusing HER2-specific CAR-T cells, initial starting doses in a number of trials
`
`Cancer Res. Author manuscript; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 6
`
`

`

`I
`
`z
`:r:::
`~
`)>
`C:
`~
`
`0 ...
`s: Q)
`
`::,
`C:
`C/J
`(")
`::l.
`
`"O --
`
`z
`:r:::
`I
`"'O
`)>
`)>
`
`C: --~
`0 ...
`s: Q)
`(") ... ;s· --
`
`::,
`C:
`C/J
`
`z
`:r:::
`I
`"'O
`)>
`)>
`C:
`~
`0 ...
`s: Q)
`
`::,
`C:
`C/J
`(")
`::l.
`
`"O --
`
`Ertl et al.
`
`Page 7
`
`Summary
`
`administering HERZ-specific CAR-T cells, were lowered to 104 cells/m2. There was general
`agreement among the RAC members that a lower starting dose of 104 cells/m2 would likely
`be safe but would unlikely have potential benefit to the subject. Therefore, the risk-benefit
`calculus was such that any benefit realized from the research would most likely be a societal
`benefit, i.e. an increase in generalizable knowledge with minimized risk but little potential
`benefit for the patient-subject. It will be a challenge to design early phase studies which
`appropriately balance accurate disclosure of the risks and benefits posed by this research and
`appropriate informed consent among a seriously ill patient-subject population with few
`therapeutic options. While the expectation of clinical benefit must be discouraged in clinical
`trials, especially those at the earliest stages of research, selecting a dose that would be
`relatively safe but have potential biologic activity is an appropriate goal. Whether the
`acceptable level of risk should be adjusted in relation to the disease prognosis for a given
`patient cohort remains a subject worthy of future debate.
`
`CAR-T cells have shown some benefit in cancer patients.( 19, 26, 30) The major challenge
`for achieving therapeutic benefit by CAR-T cell transfer remains lack of sustained
`engraftment and loss of T cell functions. These hurdles may be overcome in part by
`incorporation of costimulatory domains into CAR constructs or by modifying EB V-speci tic
`T cells. Additional studies are needed to assess the performance of EBY-specific CAR-T
`cells in cancer patients of diverse ages. Results to date ( excluding those based on EBV(cid:173)
`specific T cells) suggest that partial myeloablation is required for survival ofCAR-T cells.
`Mechanisms that cause loss of transferred 2nd and 3rd generation CAR-T cells remain poorly
`understood and may relate to the differentiation status of the transduced T cell subsets. It is
`unclear if and at what doses cytokines are needed to improve the clinical outcome of 2nd and
`3rd generation CAR-T cells. Until this issue is clarified, the upfront use of high doses of
`systemic cytokines should be carefully justified.
`
`The major immediate risk factor for CAR-T cells remains their activity against non-tumor
`cells rather than genotoxicity from the gene transfer event. Long term risk factors, e.g.
`sustained depletion of normal cell populations or exacerbated expansion of CAR-T cells
`cannot yet be assessed. While the risk of insertional mutagenesis remains a concern, this has
`neither been observed clinically nor in recent preclinical studies that have attempted to
`recreate insertional mutagenesis (SR# 15). However, as the field moves towards the use of
`less differentiated CAR-T cells, the risk of insertional mutagenesis may increase.
`
`Supplementary Material
`
`Refer to Web version on PubMed Ce ntral for supplementary material.
`
`Acknowledgments
`
`The authors acknowledge the following staff from the Otlice of Biotechnology Activities, Dr. R. Jambou , Dr. E.
`Rosenthal, Dr. M. O'Reilly. Ms . M. Montgomery and Ms. L. Gargiulo, for thei r contributions in bringing together
`this symposium and in assembling the data on trials and adverse events. The authors also thank Dr. Richard
`Junghans for helpful discussions.
`
`Cancer Res . Author manuscript; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 7
`
`

`

`z
`I
`I -u
`)>
`)>
`
`C -:T
`
`0 -,
`
`)>
`
`z
`I
`I
`~
`C -:T
`0 -,
`s:: Q)
`-5· -
`
`:::,
`C
`C/l
`(') -,
`
`z
`I
`I -u
`)>
`)>
`
`C -:T
`0 -,
`s:: Q)
`-5· -
`
`:::,
`C
`C/l
`(') -,
`
`Ertl et al.
`
`References
`
`Page 8
`
`1. Morgan RA, Dudley ME, Wunderlich JR. Hughes MS, Yang JC, Sherry RM. et al. Cancer
`Regression in Patients After Transfer of Genetically Engineered Lymphocytes. Science. 2006;
`314:126- 9. [PubMed: 16946036]
`2. Hwu P, Shafer GE. Treisman J, Schindler DG, Gross G, Cowherd R, et al. Lysis of ovarian cancer
`cells by human lymphocytes redirected with a chimeric gene composed of an antibody variable
`region and the Fe receptor gamma chain. J Exp Med. 1993: 178:361 - 6. [PubMed: 8315392]
`3. Eshhar Z. Waks T. Gross G, Schindler DG. Specific activation and targeting of cytotoxic
`lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma
`or zeta subunits of the immunoglobulin and T-cell receptors. Proceedings of the National Academy
`of Sciences of the United States of America. 1993 ; 90:720---4. [PubMed: 8421711]
`4. Stancovski I, Schindler DG, Waks T , Yarden Y, Sela M, Eshhar Z. Targeting ofT lymphocytes to
`Neu/HER2-expressing cells using chimeric single chain Fv receptors. J lmmunol. 1993; 15 l :6577
`82. [PubMed: 7902379]
`5. Gross G, Waks T, Eshhar Z. Expression ofimmunoglobulin-T-cell receptor chimeric molecules as
`functional receptors with antibody-type specificity. Proceedings of the National Academy of
`Sciences of the United States of America. 1989; 86: l 0024-8. [Pub Med: 2513569]
`6. Moritz D, Weis W, Mattern J. Groner 8. Cytotoxic T lymphocytes with a grafted recognition
`specificity for ERBB2-expressing tumor cells. Proceedings of the National Academy of Sciences of
`the United States of America. 1994: 91 :43 l 8- 22. [PubMed: 79 I 0405]
`7. Pinthus JH, Waks T, Kaufman-Francis K, Schindler DG, Harmelin A, Kanety H, et al. lmmuno(cid:173)
`Gene Therapy of Established Prostate Tumors Using Chimeric Receptor-redirected Human
`Lymphocytes. Cancer Research. 2003: 63:2470- 6. [PubMed: 12750268]
`8. Kershaw MH, Westwood JA, Parker LL. Wang G, Eshhar Z, Mavroukakis SA, ct al. A Phase l
`Study on Adoptive lmmunotherapy Using Gene-Modified T Cells for Ovarian Cancer. Clinical
`Cancer Research. 2006: 12:6 l 06-15. [PubMed: 17062687]
`9. Park JR, DiGiusto DL, Slovak M. Wright C, Naranjo A, Wagner J, et al. Adoptive Transfer of
`Chimeric Antigen Receptor Re-directed Cytolytic T Lymphocyte Clones in Patients with
`Neuroblastoma. Mol Ther. 2007; 15:825 . [PubMed: 17299405]
`I 0. Lamers Cl IJ, Sleijfcr S, Yul to AG, Kruit WI IJ, Kliffen M. Dcbcts R. et al. Treatment of Metastatic
`Renal Cell Carcinoma With Autologous T-Lymphocytes Genetically Rctargeted Against Carbonic
`Anhydrase IX: First Clinical Experience. J Clin Oncol. 2006: 24:e20- 2. [PubMed: 16648493]
`11. Zhao Y, Wang QJ. Yang S, Kochenderfcr JN. Zheng Z. Zhong X. et al. A Herceptin-Based
`Chimeric Antigen Receptor with Modified Signaling Domains Leads to Enhanced Survival of
`Transduced T Lymphocytes and Antitumor Activity. J Immunol. 2009; 183:5563- 74. [PubMcd:
`19843940]
`12. Murphy A, Westwood JA. Teng MWL, Moeller M. Darcy PK, Kershaw MIi. Gene Modification
`Strategies to Induce Tumor Immunity. Immunity. 2005: 22:403. [PubMed: I 5845446]
`13. Finney HM, Akbar AN. Lawson ADO. Activation of Resting Human Primary T Cells with
`Chimeric Receptors: Costimulation from CD28, Inducible Costimulator. CD 134. and CD 13 7 in
`Series with Signals from the TCR: zeta: Chain. J lmmunol. 2004: 172: I 04 - I 3. [Pub Med:
`14688315]
`14. Maher J, Brcntjens RJ , Gunset G. Riviere I, Sadclain M. Human T-lymphocyte cytotoxicity and
`proliferation directed by a single chimeric TCR[zcta] /CD28 receptor. Nat Biotech. 2002: 20:70.
`15. Friedmann-Morvinski D, Bendavid A, Waks T, Schindler D. Eshhar Z. Redirected primary T cells
`harboring a chimeric receptor require costimulation for their antigen-specific activation. Blood.
`2005: 105:3087 -93. [PubMed: 15626734]
`16. Savoldo 8, Ramos C. Bollard C. Liu E, Mims M, Keating M, et al. Simultaneous Comparison ofT
`Cells Expressing I st or 2nd Generation CARs in Human Subjects with 8-Cell Malignancies:
`Contribution ofCostimulatory Endodomains. Abstract from the annual meeting of the American
`Sucietv uj"Gene and Cell Therapy. 20 I 0
`
`Cancer Res. Author manuscript; available in PMC 2014 August 11
`
`UPenn Ex. 2052
`Miltenyi v. UPenn
`IPR2022-00855
`Page 8
`
`

`

`z
`I
`I
`~
`C --:::J'
`)>
`0 -,
`s:: Q)
`:::,
`C en
`0 ..,
`"§:
`
`z
`I
`I
`~
`)>
`
`C --:::J'
`0 -,
`s:: Q)
`:::,
`C en
`0
`::!.
`
`"'C -
`
`z
`I
`I
`~
`)>
`
`C --:::J'
`0 ..,
`s:: Cl)
`:::,
`C en
`
`0 -, ;s· -
`
`Ertl et al.
`
`Page 9
`
`17. Wang J, Jensen M, Lin Y, Sui X, Chen E, Lindgren CG, et al. Optimizing Adoptive Polyclonal T
`Cell lmmunotherapy of Lymphomas, Using a Chimeric T Cell Receptor Possessing CD28 and
`CDI 37 Costimulatory Domains. Human Gene Therapy. 2007; 18:712 - 25. [Pub Med: 17685852]
`18. Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM , et al. Control of large,
`established tumor xenografts with genetically retargeted human T cells containing CD28 and
`CD 13 7 domains. Proceedings of the National Academy of Sciences. 2009; I 06:3360-65.
`19. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HY, Dotti G, et al. Virus-specific T cells
`engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals
`with neuroblastoma. Nat Med. 2008; 14: I 264. [PubMed: I 8978797)
`20. Jena B, Dotti G , Cooper LJN. Redirecting T-cell specificity by introducing a tumor-specific
`chimeric antigen receptor. Blood. blood-20I0-2001-04373 7.
`21. Brentjens R, Yeh R, Bernal Y, Riviere I, Sade lain M. Treatment of Chronic Lymphocytic
`Leukemia With Genetically Targeted Autologous T Cells: Case Report of an Unforeseen Adverse
`Event in a Phase I Clinical Trial. Mol Ther. 18:666. [PubMed: 20357779]
`22. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case Report ofa
`Serious Adverse Event Following the Administration ofT Cells Transduced With a Chimeric
`Antigen Receptor Recognizing ERBB2. Mol Ther. 18:843. [PubMed: 20179677]
`23. Junghans R. Strategy escalation: an emerging paradigm for safe clinical development ofT cell
`gene therapies. J Transl Med. 201 O; 8:55. [PubMed: 20537174)
`24. Straathof KC, Pule MA, Yotnda P, Dotti G, Van in EF, Brenner MK, et al. An inducible caspase 9
`safety switch for T-cell therapy. Blood. 2005; I 05:4247- 54. (Pub Med: I 5728 I 25]
`25. Bryder D, Sigvardsson M. Shaping up a lineage--less

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