throbber
T-Cell Immunotherapy:
`Looking Forward
`
`T Cell Immunotherapy: Optimizing Trial Design
`Bethesda, Maryland
`10–11 September 2013
`
`Jacqueline Corrigan-Curay1, Hans-Peter Kiem2, David Baltimore3,
`Marina O’Reilly1, Renier J Brentjens4, Laurence Cooper5,
`Stephen Forman6, Stephen Gottschalk7, Philip Greenberg2,
`Richard Junghans8, Helen Heslop7, Michael Jensen9, Crystal Mackall10,
`Carl June11, Oliver Press2, Daniel Powell11, Antoni Ribas12,
`Steven Rosenberg10, Michel Sadelain4, Brian Till2, Amy P Patterson1,
`Robert C Jambou1, Eugene Rosenthal1, Linda Gargiulo1,
`Maureen Montgomery1 and Donald B Kohn12
`
`The rapidly expanding field of T-cell im-
`
`munotherapy has experienced clinical
`successes along with some serious toxici-
`ties. “T Cell Immunotherapy: Optimizing
`Trial Design,” a workshop sponsored by
`the National Institutes of Health’s (NIH’s)
`Office of Biotechnology Activities (OBA),
`brought together researchers to discuss the
`
`1Office of Science Policy, Office of the Director,
`National Institutes of Health, Bethesda, Mary-
`land, USA; 2Fred Hutchinson Cancer Research
`Center, Seattle, Washington, USA; 3California
`Institute of Technology, Pasadena, California,
`USA; 4Memorial Sloan-Kettering Cancer Center,
`New York, New York, USA; 5MD Anderson
`Cancer Center, Houston, Texas, USA; 6City of
`Hope, Duarte, California, USA; 7Baylor College
`of Medicine, Houston, Texas, USA; 8Roger
`Williams Medical Center, Providence, Rhode
`Island, USA; 9University of Washington, Seattle,
`Washington, USA; 10National Cancer Insti-
`tute, National Institutes of Health, Bethesda,
`Maryland, USA; 11Abramson Cancer Center,
`University of Pennsylvania, Philadelphia,
`Pennsylvania, USA; 12David Geffen School of
`Medicine, University of California, Los Angeles,
`Los Angeles, California, USA
`Correspondence: Jacqueline Corrigan-Curay,
`Office of Biotechnology Activities, National In-
`stitutes of Health, 6705 Rockledge Drive, Suite
`750, Bethesda, Maryland 20892, USA.
`E-mail: corrigaja@od.nih.gov
`
`scientific advances and share new data on
`key trial design issues, including the selec-
`tion of new targets, optimizing the T-cell
`population, preconditioning regimens,
`strategies to promote persistence of cells,
`and analysis and management of acute re-
`actions to T-cell infusions with the goal of
`identifying best practices and a research
`agenda that will facilitate further devel-
`opment and maximize the safety of this
`promising approach.
`
`Introduction
`T-cell immunotherapy for cancer is a
`rapidly growing field for gene therapy.
`Broadly, this field can be divided into two
`approaches—the use of gene-modified T-
`cell receptors (TCRs) in which recognition
`of the tumor antigen is in the context of
`human leukocyte antigens (HLAs) or use
`of chimeric antigen receptors (CARs) that
`typically link a single-chain variable re-
`gion domain of an antibody (scFv) to one
`or more signaling elements of a TCR com-
`plex to allow T-cell activation.1 The deci-
`sion to use one approach vs. the other may
`depend on several factors. For example,
`CARs offer the ability to bind antigens that
`are not restricted by HLA recognition, and
`the ability to modify the T-cell signaling
`moieties may offer “a broader functional
`
`doi:10.1038/mt.2014.148
`
`effect than transduced” TCRs.2 TCRs, how-
`ever, have the ability to recognize intracellular
`proteins, in addition to cell surface antigens,
`providing a broader array of target tumor-as-
`sociated targets.
`In 2010, the OBA hosted a meeting to ex-
`amine the state of the science and key trial de-
`sign questions for this emerging field.3 At the
`time, some clinical benefit and unexpected
`toxicities highlighted both the therapeutic
`potential as well as the need to share data and
`expertise to optimize the safety of trial design.
`Since 2010, several promising and clinically
`successful developments have been reported
`in leading scientific and medical journals4–7
`as well as national media. Given these devel-
`opments, the OBA and the NIH Recombinant
`DNA Advisory Committee concluded that it
`was an opportune time to reconvene the lead-
`ing experts in the field from the United States
`to continue to foster sharing of data across pro-
`tocols and discuss the key issues in trial design,
`including optimal management of the cytokine
`release syndrome (CRS) seen in some research
`participants in response to the expansion of
`these active T cells.
`The following summary of the OBA work-
`shop represents the views of the individual
`authors and not the NIH. The full presentations
`and slides are available at the OBA’s website.8
`
`State of the science
`The number of CAR and TCR protocols reg-
`istered with the OBA has continued to in-
`crease rapidly (Figure 1); as of the meeting in
`September 2013 there were 111 protocols, 104
`of which targeted cancer, with more than 500
`subjects dosed. More than 40 protocols address
`hematological malignancies, with CD19 being
`the most common target in these protocols.
`Among protocols for solid tumors, the mela-
`noma antigens (gp100, MART-1) and cancer-
`testis antigens predominate for TCRs; for
`CARs there are multiple targets, with a slight
`predominance of Her2/neu, GD2, and meso-
`thelin (Figures 2 and 3). Approximately 90%
`of TCR trials have targeted solid malignancies;
`approximately 50% of CAR trials have targeted
`hematological malignancies.
`Steven Rosenberg reviewed the exten-
`sive portfolio of National Cancer Institute
`(NCI) research in this area, beginning with
`a summary of his research using unmodi-
`fied tumor-infiltrating
`lymphocytes (TILs)
`against melanoma in 1988. He began using
`
`1564
`
`www.moleculartherapy.org vol. 22 no. 9 september 2014
`
`meeting report
`
`© The American Society of Gene & Cell Therapy
`
`UPenn Ex. 2080
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`No. of protocols
`
`2007–2012
`
`2001–2006
`
`1995–2000
`
`1989–1994
`
`Figure 1 Number of chimeric antigen receptor protocols registered with the
`National Institutes of Health’s Office of Biotechnology Activities by year.
`
`Forman, Michael Jensen, Helen Heslop,
`and Crystal Mackall summarized their re-
`sults in ongoing trials using CD19-specific
`CARs in leukemia and lymphoma.4,6,14 Dr.
`Heslop noted that in a trial comparing first-
`and second-generation CARs, her group
`found that the second-generation CAR
`demonstrated both improved expansion
`and persistence.15 In addition, several pro-
`tocols have established that administration
`of CAR T cells after stem cell transplant
`does not interfere with engraftment of the
`transplant. The investigators presented ex-
`amples of clinical remissions, but, because
`the goal is often to establish remission so
`as to proceed with a curative transplant,
`the durability of remissions from CAR T
`cells without subsequent transplant has
`not yet been determined. However, even
`in the setting of multiple previous thera-
`pies, CD19-specific CARs have shown ef-
`ficacy. Dr. Brentjens reported that in his
`protocol with relapsed or refractory B-cell
`acute lymphoblastic leukemia (ALL), 14
`of 16 subjects achieved molecular chronic
`remissions as assessed by deep-sequencing
`PCR analysis to search for the malignant
`clone.16 Another emerging theme was the
`responsiveness of ALL to this approach,
`which was also highlighted in Dr. June’s
`and Dr. Mackall’s presentations. Dr. Coo-
`per presented data from ongoing trials
`infusing CD19-specific CAR+ T cells after
`autologous and allogeneic hematopoietic
`stem cell transplantation. The intent was
`to augment the graft-vs.-tumor effect, rec-
`
`ognizing that the current clinical practice
`for many patients with B-cell malignan-
`cies is to infuse tumor-specific T cells as
`a bridge to transplantation. These trials
`have advanced a new approach to hu-
`man gene therapy based on the electro-
`transfer of DNA plasmids encoding a
`second-generation CAR stably expressed
`following transposition from the Sleeping
`Beauty (SB) system.
`In parallel to work on CD19-specific
`CARs, Brian Till highlighted the results of
`his trials targeting CD20, including a trial
`that used a third-generation CAR with
`CD28 and 4–1BB costimulatory domains.
`Unlike the other trials, which use retro-
`viral vectors or SB transposons, he used an
`electroporated DNA plasmid. In general,
`the T cells were well tolerated, with some
`immediate febrile reactions, and two of the
`three subjects had prolonged remissions
`with persistence of the T cells for up to a
`year.17 However, the DNA plasmid vector
`was not an efficient vector, and the IL-2
`used to promote persistence also led to an
`increase in T regulatory cells (Tregs).
`Philip Greenberg highlighted his
`group’s work using a TCR targeting an-
`other hematological malignancy antigen,
`Wilms tumor antigen 1 (WT1), which is
`highly expressed in leukemia and some
`solid tumors but is also expressed on some
`normal tissues. Their trial built on a previ-
`ous trial using naturally isolated, cloned T
`cells targeting WT1, which did not show
`toxicity but had limited efficacy. Using
`
`1565
`
`lymphodepletion before administration of
`TILs in 2002 and demonstrated increased
`efficacy.9 Dr. Rosenberg has continued to
`apply this approach to melanoma, includ-
`ing ocular melanoma, as well as metastatic
`gastrointestinal and human papillomavi-
`rus–induced cancers. These studies have
`demonstrated that in a subset of patients
`(about 20%), administration of T cells can
`result in prolonged remissions of five years
`or longer. The results led to a program of
`research dedicated to gene-modified T
`cells that accounts for almost 20% of T-cell
`immunotherapy protocols registered with
`the OBA to date. The results of the Rosen-
`berg group’s first trials with gene-modified
`TCRs for melanoma were published in
`2006 in Science.10 In a recent TCR study
`targeting the cancer-testis antigen NY-
`ESO-1, the overall response rate was 50%
`in the 19 subjects with melanoma, includ-
`ing 4 with complete remissions, and a 67%
`overall response for those with synovial
`sarcoma, including one complete remis-
`sion, in a population that had multiple
`prior chemotherapy regimens.11 These re-
`sults contrasted with the MAGE-A3 trial
`in which an unexpected off-target neu-
`rological toxicity was seen.12 Rosenberg’s
`group has also developed an extensive
`portfolio of CAR protocols, focusing pri-
`marily on solid tumors, with novel targets
`such as vascular endothelial growth factor
`receptor 2 (VEGFR2), epidermal growth
`factor receptor variant III (EGFRvIII),
`and mesothelin, as well as new targets in
`development, such as chondroitin sulfate
`proteoglycan 4 (CSP4).
`Antoni Ribas, who uses a vector de-
`veloped by Rosenberg’s lab, described his
`work on melanoma using a TCR-targeting
`MART-1 given with lymphodepletion. He
`has observed a high frequency of tumor
`responses (9 of 14 subjects with tumor-size
`reductions), but few responses were dura-
`ble. He has also recently started enrolling
`research participants into a trial using a
`TCR-targeting NY-ESO-1. He noted that
`one of the aspects being tested is whether
`fresh cells are potentially more active than
`cryopreserved cells.
`included clinical
`Other highlights
`results from several investigators target-
`ing CD19 in leukemia and lymphoma.
`In addition to Dr. Rosenberg’s summary
`of his work in this area,13 Carl June, Re-
`nier Brentjens, Laurence Cooper, Stephen
`
`Molecular Therapy vol. 22 no. 9 september 2014
`
`© The American Society of Gene & Cell Therapy
`
`meeting report
`
`UPenn Ex. 2080
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`virus-specific T cells, they have recently
`initiated a trial to test a TCR based on a
`high-avidity, natural clone.
`In the solid-tumor area, Dr. Heslop
`presented a summary of her group’s trials
`for neuroblastoma, targeting GD2 using
`both virus-specific and non-virus-specific
`T cells.18,19 Their data have demonstrated
`an association between persistence of T
`cells and reduced tumor progression. In
`addition, in research participants with
`prolonged detection of activated T cells,
`the presence of central memory T cells was
`important, raising the question of what the
`optimal T-cell product is.
`Other solid-tumor trials discussed in-
`cluded CARs targeting HER2/neu for sar-
`coma and glioblastoma, including a trial
`using tri-virus-specific T cells and another
`trial that combines the CAR with a domi-
`nant-negative TGF-b receptor. Data were
`also presented on first- and second-gen-
`eration CARs targeting carcinoembryonic
`antigen (CEA) and prostate-specific mem-
`brane antigen. Again, some early indica-
`tions of clinical efficacy were promising,
`but an ongoing challenge will be to refine
`strategies to improve T-cell persistence
`and efficacy. In some cases, on-target, off-
`tissue toxicities may ultimately limit the
`use of certain targets; for example, colitis
`developed in protocols using CEA-specific
`TCR and CAR T cells.20
`Finally, Dr. Jensen reported his work
`in glioblastoma using a novel CAR called
`a zetakine. Instead of an antibody, single-
`chain target domain, he used a human
`cytokine, IL-13, with a mutation in the
`sequence that gave high affinity for IL-13
`receptor a2. These cells were infused
`intracranially, establishing the safety of
`intracranial administration with some
`antitumor responses.
`These talks provided an overview of a
`field that continues to expand rapidly, in
`terms of both targets and diseases. Most
`protocols involve administration of the cells
`in the setting of lymphodepletion, and some
`groups, predominantly in protocols for
`solid tumors, use IL-2 to promote cell per-
`sistence. In addition to identifying effective
`targets that have minimal off-tumor effects,
`finding the ideal balance between persis-
`tence and expansion of T cells without
`triggering systemic cytokine reactions is a
`key issue for the field. This may be achieved
`by such strategies as including the design
`
`of the cells, the type of T cells infused, the
`dose, the immune status of the recipient,
`and the use of cytokine support. Finally, as
`with many cancer therapies, some toxic-
`ity is likely. Establishing protocols to limit
`toxicity so that the risk-to-benefit ratio re-
`mains favorable is a high priority.
`
`Promoting T-cell persistence
`Persistence of the gene-modified T cells
`is associated with prolonged remission
`in subjects,18 and the field has developed
`strategies to promote persistence. One ap-
`proach is to create a host environment that
`is conducive to expansion of the T cells.
`Expansion should not only promote a rig-
`orous antitumor effect but also lead to the
`development of a stable population of tu-
`mor-specific T cells that can be reactivated
`in case of recurrence of tumor antigen. Use
`of selected central memory T cells may be
`another strategy to promote an enduring
`T-cell population.
`The majority of T-cell protocols reg-
`istered with the OBA to date involve ad-
`ministration of the cells to subjects when
`they are lymphopenic. For solid-tumor
`protocols, this involves administration of
`the T cells after administration of lym-
`phodepleting chemotherapy, such as cy-
`clophosphamide, whereas the protocols
`for hematological malignancies have most
`commonly called for administering cells
`in the posttransplant setting or the use of
`disease-specific chemotherapy regimens.
`However, it is important to note that lym-
`phodepletion has not been universally
`applied, notable exceptions being studies
`administering virus-specific T cells, or the
`successful neuroblastoma protocols tar-
`geting GD2, which used both virus-specif-
`ic and non-virus-specific T cells.18
`Dr. Rosenberg reviewed his group’s
`clinical data, as well as the animal data
`that support lymphodepletion for pro-
`moting antitumor efficacy. As stated ear-
`lier, in the TIL melanoma studies, despite
`administration of 109 to 1010 T cells, the
`cells did not persist and there were mini-
`mal objective responses.21 However, when
`nonmyeloablative (NMA) chemotherapy
`using cyclophosphamide and fludarabine
`was added, and the TIL product was gen-
`erated with a shorter culture time, provid-
`ing a more diverse TIL population that
`contained both CD4+ and CD8+ T cells,
`T-cell persistence was enhanced and 6 of
`
`13 subjects showed objective cancer re-
`sponses.22 Dr. Rosenberg’s group went on
`to investigate whether the addition of 2
`or 12 Gy of total-body irradiation (TBI)
`to the NMA chemotherapy would further
`increase efficacy of TIL transfer in mela-
`noma patients. The response rate for those
`who received chemotherapy alone was
`about 49%; the addition of 2 Gy resulted
`in objective response in 52% of subjects,
`and 12 Gy of TBI resulted in a 72% ob-
`jective response rate, with a complete
`response rate of 40%.23 The addition of
`TBI to NMA chemotherapy was generally
`well tolerated, with the exception of one
`death in a subject with an undetected di-
`verticular abscess in the 12-Gy group. A
`drawback of escalation to 12 Gy of radia-
`tion is the need for autologous peripheral
`blood stem cell support. An ongoing ran-
`domized trial is comparing NMA chemo-
`therapy against NMA and TBI, although
`preliminary results indicate that the chal-
`lenges of adding TBI may not be balanced
`by the improved response.
`A significant amount of animal work
`has been done to elucidate the mechanisms
`that underlie the improved antitumor re-
`sponses observed with lymphodepletion.
`These data indicate that lymphodepletion
`augments the antitumor response by elim-
`inating Tregs, cellular “sinks” for cytokines
`such as IL-7 and IL-15, and by enhanc-
`ing antigen-presenting cell activation and
`availability.24–26 This activation of the im-
`mune system may be due in part to trans-
`location of bacteria from the gut. It was
`shown in a mouse model that adminis-
`tration of ciprofloxacin, which is effective
`against Gram-negative bacteria commonly
`found in the gut, to an irradiated animal
`reduced the activated dendritic cells in the
`spleen and reduced the effectiveness of
`adoptive cell transfer. Of note, it has been
`demonstrated that the effect of lymphode-
`pletion is on the host rather than on the tu-
`mor. Thus, if one shields the host—in this
`case, the mouse—and treats the tumor, no
`effect is seen in these melanoma models.
`One dilemma is that Tregs are the first
`T cells to recover after lymphodepletion,
`and therefore lymphodepletion may foster
`an environment that works against the an-
`titumor effect. Dr. Rosenberg noted that
`the NCI group has some data demonstrat-
`ing an inverse relationship between the
`recovery of Tregs and objective antitumor
`
`1566
`
`www.moleculartherapy.org vol. 22 no. 9 september 2014
`
`meeting report
`
`© The American Society of Gene & Cell Therapy
`
`UPenn Ex. 2080
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`response, supporting the importance of
`eliminating Tregs. However, others ques-
`tioned whether we clearly understand the
`role of Tregs, because suppression of a tu-
`mor response may depend on whether the
`Tregs are actually activated and tumor-
`specific. Therefore, the presence of Tregs
`may not be absolutely undesirable, as they
`may also organize the immune response.
`
`Gene delivery and design of T cells
`In addition to host preparation, the design
`of the T-cell vectors is a critical area of re-
`search. Dr. Cooper noted that the ability to
`stably express transgenes, such as CARs,
`in T cells has revolutionized adoptive im-
`munotherapy for certain malignancies.
`Recombinant fusion genes constructed to
`recognize tumor-associated antigens (e.g.,
`TCR and CAR) have been constitutively
`expressed in T cells using Moloney mu-
`rine leukemia virus (MMLV)-based retro-
`viruses, HIV-based lentiviruses, and DNA
`plasmids, including the SB transposon/
`transposase system.
`Until recently, retroviral transduc-
`tion by recombinant MMLV-derived vec-
`tors has been the most common method
`for delivery of transgenes intended to be
`integrated into the T-cell genome. Lenti-
`viral vectors have also been successfully
`used in the clinic. Both approaches are
`appealing, and at this time there appears
`to be equipoise regarding the therapeutic
`potential of these two viral systems for
`genetic modification of T cells to express
`CARs. Transduction using retroviral and
`lentiviral vectors can be highly efficient,
`and it is possible to integrate multiple cop-
`ies of a transgene in a given T cell, which
`provides for a high level of expression of
`the transduced gene product. The manu-
`facture of clinical-grade retroviral and
`lentiviral vector virions is quite similar, al-
`though retroviral vectors may be produced
`from stable packaging cell lines, whereas
`to date most lentiviral vectors have been
`produced by transient transfection.
`Overall, transduction of T cells with
`recombinant retrovirus and lentivirus in-
`volve similar packaging protocols, utilize
`similar integration mechanisms, and lead
`to similar transduction efficiencies. Thus,
`both viral-based approaches to gene trans-
`fer are appealing for the human application
`of CAR+ T cells, although some individual
`investigators have strong preferences.
`
`Molecular Therapy vol. 22 no. 9 september 2014
`
`transposons now offer an
`DNA
`to viral-based gene
`trans-
`alternative
`fer. Supercoiled plasmids can be directly
`electroporated into T cells using commer-
`cial devices, thus eliminating much of the
`labor and safety concerns associated with
`generating recombinant viral particles.
`DNA transposons, such as those derived
`from the SB system, insert into the genome
`via a copy-and-paste mechanism when a
`transposase is (transiently) available to cat-
`alyze the reaction. Dr. Cooper’s group has
`successfully used SB to integrate a CD19-
`specific CAR into human T cells in four
`human trials under investigational new
`drug applications. Unlike retroviral/lentivi-
`ral integration into transcriptionally active
`sites, the SB transposon appears to ran-
`domly integrate at TA dinucleotide repeats
`and is typically present at one or two copies
`per T-cell genome. As with viral-based gene
`transfer, there is the possibility that a trans-
`poson may cause genotoxicity resulting in
`oncogenesis. However, because the SB sys-
`tem does not readily target transcriptional
`or promoter elements, it appears suitable
`for human application. Furthermore, the
`relatively low cost of generating DNA plas-
`mids for use in compliance with current
`good manufacturing practice (GMP), in
`contrast to the cost and complexity of pro-
`ducing clinical-grade virus, renders the SB
`system an attractive and nimble approach
`to generate and modify vectors for delivery
`of therapeutic genes.
`investigator has
`In summary, the
`available multiple approaches to geneti-
`cally modify T cells. The use of a par-
`ticular approach will depend on resident
`expertise and the desired T-cell product.
`
`Design of CARs. CARs are recombinant
`receptors for antigens that retarget and
`eventually reprogram T-cell function. Un-
`like the physiological TCR for antigens,
`which signals T-cell activation through the
`associated CD3 complex, CARs possess
`in a single molecule the ability to trigger
`multiple antigen-specific T-cell
`func-
`tions. The CARs that have recently shown
`impressive clinical outcomes in research
`participants with B-cell malignancies are
`“second-generation CARs,” to distinguish
`them from earlier forms of activating
`fusion receptors, which only initiate T-cell
`activation and are now referred to as “first-
`generation CARs.”27
`
`Michel Sadelain described how the
`incorporation of co-stimulatory receptor
`signaling domains into the cytoplasmic
`tails of CAR (“embedded costimulation”)
`greatly increased the potency of CAR-
`modified T cells in preclinical models.4,28,29
`Several costimulatory domains have been
`incorporated in CARs over the past decade,
`including CD28 (ref. 28), 4–1BB (ref. 30),
`OX40 (ref. 31), and others (ref. 2). Different
`costimulatory molecules play roles in T-cell
`activation, proliferation, survival, cytokine
`secretion, antitumor cytolytic activity, and
`reactivation upon secondary stimulation.
`The second- and third-generation CARs
`have varying activities by recruiting multi-
`ple T-cell signaling pathways.2 Dr. Sadelain
`emphasized that small nuances in struc-
`tural design of different CAR molecules can
`eventually exert a significant effect on the
`relative activity of CARs encoding the same
`signaling domains, depending on epitope
`position, CAR affinity, physical parameters
`of the extracellular domains, and trans-
`membrane elements. Levels of CAR expres-
`sion also affect overall function, making it
`an important parameter to consider when
`comparing different CARs. Forced expres-
`sion of co-stimulatory ligands in the CAR T
`cells themselves can produce auto- or trans-
`costimulation and increase T-cell potency.32
`Clinical efficacy has been reported in
`trials from several institutions for B-lin-
`eage malignancies using CAR-modified
`T cells.4–6,14,16,33,34 Many features of the tri-
`als differ, including CARs (origin of scFv,
`epitope of CD19 targeted, antigen affinity,
`signaling domains), enhancer/promoters
`(varied expression levels, propensity to si-
`lencing), T-cell manufacturing techniques
`(activation of T cells with antibodies to CD3
`with or without anti-CD28, different culture
`media, duration of culture), cell products
`(cell dose, CD4/CD8 ratio, central memory
`T cells), lymphodepletion conditioning reg-
`imens (cyclophosphamide vs. cyclophos-
`phamide/fludarabine vs. bendamustine),
`and patient selection (chemosensitive vs.
`chemoresistant disease). Future trials will
`need to define the relative importance of
`these differences to improve response rates.
`It is noteworthy that the outcomes of CD19
`CAR therapy may vary depending on the
`disorder. Thus, results reported to date show
`greater efficacy in ALL than in chronic lym-
`phocytic leukemia (CLL), for reasons that
`remain to be elucidated.
`
`1567
`
`© The American Society of Gene & Cell Therapy
`
`meeting report
`
`UPenn Ex. 2080
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`(HSCs), which would
`cells
`stem
`continually produce transduced T cells.
`David Baltimore listed potential advan-
`tages of targeting HSCs. Because of the re-
`quirement for coexpression of CD3, trans-
`genic TCRs can be expressed only on the
`surface of T cells derived from the trans-
`duced HSCs. The TCRs introduced by the
`vector should allelically exclude the rear-
`rangement of endogenous TCR genes to
`yield monoclonal cells. However, one po-
`tential limitation of this approach may be
`that highly active T cells from HSCs that
`contain highly avid TCRs for self-antigens
`may be selected out by the thymus. In the
`trials using a MART-specific TCR, clini-
`cal effect was observed when the avidity
`of the natural TCR was increased several-
`fold, but such highly active T cells may be
`negatively selected by the thymus.
`HSCs transduced with CAR vectors
`produce CAR-expressing myeloid and
`natural killer cells in addition to T cells,
`and thus may provide more rapid and
`broader antitumor activity.36 In a mouse
`model with an EL4 tumor expressing the
`ovalbumin gene, an antitumor effect was
`observed using HSCs transduced with
`lentiviral vectors expressing TCR reactive
`to ovalbumin. A clinical trial involving
`autologous CD34+ cells transduced with a
`lentiviral vector expressing a CD19+ CAR
`in subjects with non-Hodgkin’s lymphoma
`is being developed at UCLA and the City
`of Hope Medical Center.
`
`Target selection
`Dr. Rosenberg reviewed the status of tar-
`get selection, which he viewed as the criti-
`cal challenge confronting immunotherapy.
`He considered the targets identified thus
`far to fall into five categories. The category
`that has been most extensively studied with
`TCRs is differentiation antigens that are
`overexpressed on cancers compared with
`normal tissues (e.g., MART-1, gp100, CEA,
`HER-2). As with conventional chemother-
`apy, this approach requires identifying a
`window of toxicity against the tumor cells
`without unacceptable damage to normal
`tissue. In the studies using the melanocyte
`differentiation antigens, an approximately
`25% objective response rate was obtained;
`however, normal melanocytes were also at-
`tacked, causing skin rashes, uveitis, and au-
`ditory and vestibular problems, all of which
`could be reversed by steroid treatment.10,37
`
`Design of T-cell receptors. TCRs are the
`physiological recognition system of T
`cells and react to a major histocompatibil-
`ity complex–antigen complex. Their two
`chains, a and b, are necessary and sufficient
`for T cells to recognize their targets, includ-
`ing cancer cells. Engineering of T cells with
`genetically modified TCR a- and b-chains
`redirects their antigen specificity and has
`been used in the clinic in adoptive cell
`transfer strategies. Clinical trials expressing
`TCRs for MART-1, gp100, and NY ESO-1
`have demonstrated antitumor activity in
`subjects with metastatic melanoma and
`sarcoma. However, these early clinical trials
`suggest that durable tumor responses seem
`to occur at lower frequency than with TILs
`or with CAR-engineered T cells.
`The clinical trials thus far have used
`TCRs with physiological peptide affinities,
`and most have used intact TCRs. However,
`studies with NY ESO-1 and MAGE-A3 as
`targets used TCRs with altered affinities due
`to targeted mutations in their complemen-
`tarity-determining region 2 or 3 (CDR2 or
`CDR3), the variable regions of the TCR that
`interact with the major histocompatibility
`complex–antigen complex. However, care
`must be taken because a CDR2-modified
`MAGE-A3 TCR led to cardiac toxicities,
`due to loss of specificity with cross-reaction
`to an off-target peptide.35
`Other means to increase antitumor
`activity of TCR-modified T cells are being
`developed preclinically, such as additional
`genetic engineering of the T cells to express
`
`other immune-activating genes, engineer-
`ing the signaling pathways downstream of
`the TCR, or blocking negative regulatory
`receptors. These approaches would pro-
`vide simultaneous genetic redirection of
`T cells with increased T-cell functionality
`that may no longer be blocked by physi-
`ological immune regulatory processes.
`A problem with some transgenic TCRs
`is that, when expressed in T cells that have
`their own endogenous TCR a- and b-
`chains, there can be heterologous pairing
`between the transgenic and endogenous
`TCR chains. This may decrease the ex-
`pression of the transgenic TCR and even
`lead to altered specificities that may po-
`tentially result in autoimmune toxicities.
`Several means to improve self-pairing of
`the transgenic TCR chains include the use
`of picornavirus-derived highly efficient
`self-cleaving 2A-like sequences to allow
`stoichiometric protein expression, includ-
`ing additional cysteine motifs allowing
`formation of an increased number of di-
`sulfide bonds between the a- and b-chains,
`partially murinizing the constant region of
`both TCR chains for preferential pairing,
`and the use of leucine zippers at the 3ʹ ends
`of both a- and b-chains for forced trans-
`genic TCR pairing. As these approaches
`move into the clinic, it will be important
`to test them in carefully designed clinical
`trials to minimize risks but also foster con-
`tinued improvements in treatment options.
`Longer-term antitumor activity may
`be achievable by targeting hematopoietic
`
`40
`
`35
`
`30
`
`25
`
`20
`
`15
`
`10
`
`5
`
`0
`
`No. of protocols
`
`CD19
`
`CD20
`
`CD30
`
`ROR1
`
`Kappa1
`
`Figure 2 Chimeric antigen receptor targets for hematological-malignancy protocols
`registered with the National Institutes of Health’s Office of Biotechnology Activities.
`
`1568
`
`www.moleculartherapy.org vol. 22 no. 9 september 2014
`
`meeting report
`
`© The American Society of Gene & Cell Therapy
`
`UPenn Ex. 2080
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Similar problems occurred with targeting
`of CEA, which is expressed at low levels
`on colonic epithelium, resulting in tem-
`porary but almost complete destruction
`of that tissue,20 and with HER-2 targeting,
`which results in severe adverse effects on
`the pulmonary epithelium and death of the
`subject.38 T-cell therapy is highly potent but
`also so sensitive that the T cells can recog-
`nize even extremely low target expression
`in normal tissues. This potential for on-tar-
`get, off-tumor toxicity has limited the de-
`velopment of certain targets as an effective
`cancer treatment.
`The second class of targets includes
`antigens expressed on tumor cells and rel-
`atively nonessential normal tissues. This
`includes CD19 as targeted by CARs and
`thyroglobulin, targeted by TCRs for thy-
`roid cancer. This approach is promising
`but requires identification of additional
`tissue-specific

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