throbber
© The American Society of Gene & Cell Therapy
`
`See page 666
`
`original article
`
`Case Report of a Serious Adverse Event Following
`the Administration of T Cells Transduced With
`a Chimeric Antigen Receptor Recognizing Erbb2
`Richard A Morgan1, James C Yang1, Mio Kitano1, Mark E Dudley1, Carolyn M Laurencot1
`and Steven A Rosenberg1
`
`1Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
`
`In an attempt to treat cancer patients with ERBB2
` overexpressing tumors, we developed a chimeric anti-
`gen receptor (CAR) based on the widely used humanized
`monoclonal antibody (mAb) Trastuzumab (Herceptin).
`An optimized CAR vector containing CD28, 4-1BB, and
`CD3ζ signaling moieties was assembled in a γ-retroviral
`vector and used to transduce autologous peripheral
`blood lymphocytes (PBLs) from a patient with colon
`cancer metastatic to the lungs and liver, refractory to
` multiple standard treatments. The gene transfer effi-
`ciency into autologous T cells was 79% CAR+ in CD3+
`cells and these cells demonstrated high-specific reactiv-
`ity in in vitro coculture assays. Following completion of
`nonmyeloablative conditioning, the patient received
`1010 cells intravenously. Within 15 minutes after cell infu-
`sion the patient experienced respiratory distress, and
`displayed a dramatic pulmonary infiltrate on chest X-ray.
`She was intubated and despite intensive medical inter-
`vention the patient died 5 days after treatment. Serum
`samples after cell infusion showed marked increases in
`interferon-γ (IFN-γ), granulocyte macrophage-colony
`stimulating factor (GM-CSF), tumor necrosis factor-α
`(TNF-α), interleukin-6 (IL-6), and IL-10, consistent with
`a cytokine storm. We speculate that the large number
`of administered cells localized to the lung immedi-
`ately following infusion and were triggered to release
`cytokine by the recognition of low levels of ERBB2 on
`lung epithelial cells.
`Received 14 January 2010; accepted 22 January 2010; published online
`23 February 2010. doi:10.1038/mt.2010.24
`
`IntroductIon
`ERBB2 (HER-2/neu) is a member of the epidermal growth factor
`receptor family. Epidermal growth factor receptor–ligand interac-
`tion induces the heterodimerization of receptors, which in turn
`results in the activation of intracellular tyrosine kinase domain
`signaling cascades that mediate cell growth, differentiation, and
`survival.1–3 Overexpression of ERBB2 can induce dimerization of
`ERBB2 and initiates signal transduction activities without ligand
`
`binding. ERBB2 overexpression/amplification occurs in ~15–25%
`of human breast cancer patients, and is associated with more
`aggressive disease.4 A proportion of other human cancers are also
`associated with ERBB2 gene amplification and protein overexpres-
`sion; including cancers of the colon, ovary, stomach, kidney, mela-
`noma, and others.5–7 Investigation of agents that target the ERBB2
`protein led to the development of Trastuzumab (Herceptin), a
`humanized monoclonal antibody (mAb) that binds to the extra-
`cellular domain of the receptor.8 Trastuzumab has been shown
`to be of clinical benefit for metastatic breast cancer patients with
`ERBB2 overexpression/amplification, either alone or in combina-
`tion with chemotherapy regimens.9,10 ERBB2 has also been the
`target of several cancer vaccine trials,11–13 as well as, adoptive cell
`therapy using anti-ERBB2 cytotoxic T lymphocyte lines.14
`Adoptive cell therapy has emerged as the most effective treat-
`ment for patients with metastatic melanoma. Adoptive cell therapy
`using tumor-reactive autologous tumor infiltrating lymphocytes
`(TIL) in combination with nonmyeloablative but lymphodeplet-
`ing conditioning resulted in 50% objective clinical regression in
`melanoma patients.15 Intensifying the lymphodepletion by adding
`total-body irradiation to the chemotherapy conditioning regi-
`men improved the objective response rate to 72%.16 This potent
`therapy, however, has been limited by the requisite surgery to pro-
`cure tumor-reactive TIL, by ex vivo identification and expansion
`of these cells, and by the failure to reproducibly isolate similar cells
`from common epithelial tumors.
`The transfer of genes into primary human lymphocytes per-
`mits the introduction of tumor antigen receptor molecules that
`can endow the engineered cell with antitumor specificity.17–19 We
`reported the first clinical trials using autologous peripheral blood
`lymphocytes (PBLs) modified to express a tumor antigen- reactive
`T-cell receptor in the treatment of patients with metastatic mela-
`noma that resulted in objective tumor regressions.20,21 These strat-
`egies, however, have a lower response rate than TIL, and only
`a minority of patients are eligible for current protocols, as they
`must express human leukocyte antigen-A*0201 in order to be
` recognized by the T-cell receptor-engineered cells.
`An alternative to T-cell receptor gene therapy is the use of
`a chimeric antigen receptor (CAR) that is capable of relaying
`excitatory signals to T cells in a non-Major histocompatibility
`
`Correspondence: Richard A Morgan, Surgery Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive, CRC-3W5940, Bethesda,
`Maryland 20892, USA. E-mail: rmorgan@mail.nih.gov
`
`Molecular Therapy vol. 18 no. 4, 843–851 apr. 2010
`
`843
`
`UPenn Ex. 2033
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Serious Adverse Event With ERBB2-based CAR
`
`© The American Society of Gene & Cell Therapy
`
`complex-restricted manner. These hybrid proteins, composed of
`an extracellular antigen recognition domain fused to an intra-
`cellular T-cell activation domain,22,23 may therefore be used in
`patients regardless of their human leukocyte antigen genotype.
`The absence of human leukocyte antigen-restricted antigen rec-
`ognition is achieved by harnessing the antigen-binding properties
`of mAb; this recognition is also independent of antigen process-
`ing, thus bypassing a potential mechanism by which tumor cells
`can evade the immune system in vivo. Several clinical trials using
`CAR-transduced T cells have been reported.24–27 ERBB2-based
`CARs reported thus far are composed of single-chain Fv fragment
`from murine mAb, which have been shown to induce anti-CAR
`immune responses in humans.25,26 The anti-ERBB2 CAR used in
`this case report was a next generation CAR containing both the
`humanized Herceptin single-chain Fv fragment and optimized
`costimulatory signaling domains designed for increased cytokine
`secretion, lytic activity, and shown to display robust in vivo anti-
`tumor activity in a human breast cancer xenograft model.28
`
`results
`In vitro characteristics of the erbB2-cAr
`transduced t cells for patient treatment
`Leukophoresis was performed to obtain patient peripheral blood
`mononuclear cells (PBMCs), which were stimulated with an anti-
`CD3 mAb and interleukin-2 (IL-2) to initiate T-cell expansion
`followed by transduction with the 4D5-CD8-28BBZ ERBB2-
`
`CAR vector as described in Materials and Methods section. At
`4 days before infusion, cells were analyzed for the expression of
`the ERBB2 CAR using an ERBB2-Fc fusion protein as previously
`described.28 As shown in Figure 1, 79% of CD3+ T cells expressed
`the CAR with gene transfer into both CD4+ (17%) and CD8+
`(63%) T-cell subsets. To determine functional activity, transduced
`T cells were cocultured with ERBB2+ melanoma, breast cancer,
`and ovarian cancer cell lines, or ERBB2− breast cancer and T
`lymphoblastoid cell lines. ERBB2-specific reactivity was demon-
`strated by production of effector cytokine interferon-γ (IFN-γ)
`only in cell lines expressing ERBB2 (Table 1). Background
`cytokine production of ERBB2-CAR transduced cells, when
`cocultured with ERBB2-targets, was similar to untransduced
`control cells. To complete the certificate of analysis for patient
`treatment, transduced cells were also tested to be negative for the
`presence of replication competent retrovirus by PCR and passed
`sterility testing (data not shown). Retrospective testing using an
`amplification-based S+/L− assay was also negative for replication
`competent retrovirus.
`
`clinical course
`The patient was a 39-year-old female who 3 years earlier had
`undergone a sigmoid resection for colon cancer that on patho-
`logic ana lysis exhibited
`lymphatic
`invasion and vascular
`involvement, with spread to 6 of 21 lymph nodes and the pres-
`ence of synchronous liver metastases. She was treated with a
`
`5′ LTR
`
`sd
`

`
`sa
`
`4D5 scFv
`
`CD8 Hinge &TM
`
`CD28
`
`4-1BB
`
`CD3ζ
`
`3’ LTR
`
`0.2
`
`0.5
`
`0.4
`
`0.3
`
`UnTd
`
`21
`
`78
`
`102
`101
`100
`103
`APC-Cy7-A:: CD8 APC-Cy7-A
`
`103
`
`102
`
`101
`
`100
`
`PE-A:ErbB2 + GAMPE PE-A
`
`0.6
`
`0.1
`
`75
`
`24
`
`100
`
`102
`101
`103
`PE-Cy7-A:: CD4 PE-Cy7-A
`
`CD4
`
`CD8
`
`16
`
`6
`
`CAR
`
`63
`
`14
`
`102
`101
`100
`103
`APC-Cy7-A:: CD8 APC-Cy7-A
`
`CD8
`
`103
`
`102
`
`101
`
`100
`
`PE-A:ErbB2 + GAMPE PE-A
`
`62
`
`15
`
`17
`
`5.0
`
`100
`
`102
`101
`103
`PE-Cy7-A:: CD4 PE-Cy7-A
`
`CD4
`
`103
`
`102
`
`101
`
`100
`
`PE-A:ErbB2 + GAMPE PE-A
`
`103
`
`102
`
`101
`
`100
`
`PE-A:ErbB2 + GAMPE PE-A
`
`98
`
`79
`
`20
`
`103
`
`102
`
`101
`
`100
`
`PE-A:ErbB2 + GAMPE PE-A
`
`0.5
`
`100
`
`101
`103
`102
`FITC-A:: CD3 FITC-A
`
`CD3
`
`102
`101
`103
`FITC-A:: CD3 FITC-A
`
`0.4
`
`0.3
`
`100
`
`103
`
`102
`
`101
`
`100
`
`PE-A:ErbB2 + GAMPE PE-A
`
`CD3
`
`a
`
`b
`
`ErbB2
`CAR
`
`ErbB2
`CAR
`
`Figure 1 expression of the erBB2 cAr. Diagram of the ERBB2-CAR vector (MSGV1-4D5-CD8-28BBZ) used in this trial is as shown on the top of
`the figure. As described in Materials and Methods section, patient PBMC were stimulated to induced T-cell division and then transduced with the
`CAR vector. Four days before cell infusion samples were removed for analysis of ERBB2-CAR gene expression by FACS (along with the CD3, CD4, or
`CD8 T-cell markers). CAR, chimeric antigen receptor; LTR, long terminal repeats; PBMC, peripheral blood mononuclear cell; scFv, single-chain Fv
`fragment.
`
`844
`
`www.moleculartherapy.org vol. 18 no. 4 apr. 2010
`
`UPenn Ex. 2033
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`© The American Society of Gene & Cell Therapy
`
`Serious Adverse Event With ERBB2-based CAR
`
`table 1 erbB2-cAr certificate of analysis
`
`erbB2+ target
`MdA361
`Mel526
`Mel624
`Mel938
`Mel888
`Media
`effector
`57
`44
`66
`68
`49
`24
`None
`37
`3,700
`4,900
`35
`37
`26
`MART-1 TCR
`>26,815
`11,430
`>21,065
`13,210
`12,745
`46
`Patient CAR
`82
`93
`127
`81
`69
`46
`Mock Td PBL
`Abbreviations: CAR, chimeric antigen receptor; IFN-γ, interferon-γ; PBL, peripheral blood lymphocyte.
`Cell reactivity following coculture of 1e5 effectors with 1e5 targets. Data are IFN-γ (pg/ml) following overnight incubation. Mock Td PBL, untransduced PBL maintained
`under identical culture conditions; MART-1 TCR, MART-1 TCR-transduced PBL; patient CAR, ErbB2 CAR-transduced patient PBL. ErbB2 antigen expression was as
`indicated. Only melanoma lines 526 and 624 expressed both HLA-A2 and MART-1, required for TCR recognition.
`
`erbB2− target
`ceM
`MdA468
`37
`99
`29
`55
`114
`134
`117
`103
`
`sK-Br3
`143
`68
`>33,385
`144
`
`sK-oV3
`82
`47
`>58,480
`146
`
`chemotherapy regimen consisting of 5-fluorourocil, leucovorin
`and oxaliplatin plus the antivascular endothelial growth factor
`mAb, bevacizumab. The tumor progressed and the patient was
`then treated with an alternate chemotherapy regimen in which
` irinotecan was substituted for oxaliplatin (FOLFIRI). The patient
`again progressed and after desensitization to oxaliplatin for an
`allergic reaction, she received a third chemotherapy regimen con-
`sisting of capecitabine, oxaliplatin, and bevacizumab. The tumors
`in the lung and liver continued to progress and the patient was
`referred to the Surgery Branch, National Cancer Institute (NCI;
`Bethesda, MD) and signed an informed consent for our protocol.
`The protocol was reviewed and approved by the National Institutes
`of Health Institutional Biosafety Committee, the NCI Institutional
`Review Board, the National Institutes of Health Office of
`Biotechnology Activities, and the Food and Drug Administration
`(all Bethesda, MD). Patient inclusion criteria included metastatic
`cancer that expressed ERBB2 (Her-2/neu) at ≥2+ as assessed by
`immunohistochemistry.
`To facilitate homeostatic expansion of the transduced cells, the
`patient received a lymphodepleting regimen (60 mg/kg cyclophos-
`phamide daily for 2 days followed by flurodarabine 25 mg/m2 for
`the next 5 days). On the day following the last chemotherapy dose
`the patient received an intravenous infusion of 1010 cells trans-
`duced with the ERBB2 CAR in 125 ml over 30 minutes. This was
`the largest number of cells permitted in the first dose- escalation
`cohort. Within 15 minutes after completing the infusion, the
`patient developed respiratory distress with decreased blood oxy-
`gen saturation that worsened over the next hour. Chest X-ray
`obtained 40 minutes after completion of the infusion showed pul-
`monary edema, which appeared worse on chest X-rays repeated
`at 2 and 4 hours after the infusion. Because of decreasing respi-
`ratory function the patient was transferred to the intensive care
`unit and was intubated about 1 hour after the cell infusion. The
`patient then developed severe hypotension requiring vasopres-
`sors. Dexamethasone, 8 mg every 6 hours, was administered start-
`ing at about 5 hours after the cell infusion (it was continued for 2
`days, after which the dose was tapered). The patient experienced
`two cardiac arrests in the next 12 hours after cell infusion both
`requiring cardiopulmonary resuscitation. She was maximally sup-
`ported with vasopressors and ventilatory support. She remained
`severely ill with maximum intensive care unit support for the next
`5 days at which time progressive hypotension and bradycardia as
`well as gastrointestinal bleeding resulted in cardiac arrest from
`which she could not be resuscitated.
`
`Postmortem analysis
`At autopsy, multiple organs exhibited signs of systemic ischemia
`and hemorrhagic microangiopathic injury. The lungs also showed
`diffuse alveolar damage consistent with the clinical findings of
`acute respiratory distress syndrome. Autopsy also revealed a
` generalized rhabdomyolysis. Copious blood in the small intestine
`indicated that the patient succumbed to hemorrhage in the set-
`ting of multiple organ failure secondary to systematic microangio-
`pathic injury. The autopsy findings appeared to be a combination
`of the initial pulmonary injury followed by the sequela of several
`days of hypotension and organ ischemia.
`Beginning at about 4 hours after cell infusion serum samples
`were obtained and stored for analysis. Compared to pretreat-
`ment samples, the patient’s serum displayed a rapid and marked
`increase in the levels of five cytokines; IFN-γ, granulocyte mac-
`rophage-colony stimulating factor (GM-CSF), tumor necrosis
`factor-α (TNF-α), IL-6, and IL-10 (Figure 2). At 4 hours after
`infusion, four of the five cytokines displayed peak serum levels:
`IFN-γ,11, 456 pg/ml; TNF-α, 380 pg/ml; GM-CSF, 10,191 pg/ml;
`and IL-6, 34,467 pg/ml. Preinfusion levels for IFN-γ, GM-CSF,
`and IL-6 were undetectable, whereas TNF-α values varied from
`0 to 51 pg/ ml. The levels of these cytokines decreased over the
`next 3 days but remained above baseline values. The levels of
`seven additional cytokines (IL-1β, IL-2, IL-4, IL-7, IL-10, IL-12,
`and TRAIL) were determined by cytokine array (SearchLight
`assay) with only IL-10 showing increased levels after infusion,
`which unlike the other cytokines, was sustained through the study
`period (8 pg/ml preinfusion, 219 pg/ml at about 100 hours after
`infusion, Figure 2). An increased amount of IL-2 was observed
`at 4 hours only and was likely associated with the administra-
`tion of the cell product, which was given in saline with 300 IU/ ml
`IL-2 (the patient received no other IL-2). The possible involve-
`ment of an anaphylactic response to the infused cells was deemed
`unlikely because we measured only a modest twofold increase in
`serum tryptase levels at the 4 hours time point (from 7–9 pg/ml
` preinfusion to 15 pg/ml at 4 hours).
`To determine the relative tissue distribution of vector-containing
`cells, DNA was isolated from samples obtained at autopsy and
`subjected to quantitative-PCR using vector-specific primers and
`probe. As a reference for this analysis, DNA was extracted from
`the infusion sample (79% ERBB2-CAR+) and arbitrarily assigned
`a value of 100 for comparison to tissue samples. There was a wide
`variation in the presence of vector-containing cells found in mul-
`tiple tissues, though the highest levels were seen in the lung and
`
`Molecular Therapy vol. 18 no. 4 apr. 2010
`
`845
`
`UPenn Ex. 2033
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Serious Adverse Event With ERBB2-based CAR
`
`© The American Society of Gene & Cell Therapy
`
`Pre
`
`~4 hours
`
`~18 hours
`
`
`
`~26 hours~48 hours
`
`~72 hours
`
`400
`
`350
`
`300
`
`250
`
`200
`
`150
`
`100
`
`50
`
`0
`
`TNF−α (pg/ml)
`
`35,000
`
`30,000
`
`25,000
`
`20,000
`
`15,000
`
`10,000
`
`5,000
`
`IL-6 (pg/ml)
`
`12,000
`
`10,000
`
`8,000
`
`6,000
`
`4,000
`
`2,000
`
`IFN-γ (pg/ml)
`
`0
`
`Pre
`
`~4 hours
`
`~18 hours
`
`
`
`~26 hours~48 hours
`
`~72 hours
`
`12,000
`
`10,000
`
`8,000
`
`6,000
`
`4,000
`
`2,000
`
`GM-CSF (pg/ml)
`
`0
`
`Pre
`
`~4 hours
`
`~18 hours
`
`
`
`~26 hours~48 hours
`
`~72 hours
`
`0
`
`Pre
`
`~4 hours
`
`~18 hours
`
`
`
`~26 hours~48 hours
`
`~72 hours
`
`Pre
`
`~4 hours
`
`~18 hours
`
`~26 hours
`
`~100 hours
`
`250
`
`200
`
`150
`
`100
`
`50
`
`0
`
`IL-10 (pg/ml)
`
`Figure 2 serum cytokine levels. Serum samples obtained at the approximate times indicated after cell infusion were assayed for cytokine expression
`using commercial ELISA kits for cytokines IL-6, TNF-α, GM-CSF, and IFN-γ. The levels of cytokine IL-10 were independently determined by cytokine
`array (SearchLight) assay. All samples were diluted as necessary as to be in the linear range of the assay. ELISA, enzyme-linked immunosorbent assay;
`GM-CSF, granulocyte macrophage-colony stimulating factor; IFN-γ, interferon-γ; IL, interleukin; TNF-α, tumor necrosis factor-α.
`
`abdominal/mediastinal lymph nodes (Table 2). There did not
`appear to be a preferential accumulation of vector-containing
`cells in metastatic deposits in the liver or lungs. DNA from the
`pretreatment PBMC was also subjected to analysis of single-
` nucleotide polymorphisms associated with the activity/function
`of five cytokine genes (IL-6, IL-10, IFN-γ, TGF-β1, and TNF-α).
`PCR using sequence-specific primers (Figure 3) indicated that
`the patients’ genotype was: IL-6, heterozygous −741G/C; IL-10,
`homozygous −1082G, −819C, −592C; IFN-γ, homozygous 874A;
`transforming growth factor-β1, homozygous 10T, 25G; and TNF-α
`homozygous −308G. This genotype is consistent with a phenotype
`of increased synthesis of transforming growth factor-β, IL-10, and
`IL-6, but lower production of TNF-α and IFN-γ.
`Analysis of the ERBB2-CAR transduced T cells before infu-
`sion demonstrated specific recognition of ERBB2-expressing
`tumor cells (Table 1). To confirm and extend this analysis, an
`aliquot of the cryopreserved infusion sample was thawed and
`
`retested. In addition to cell lines used for the certificate of analy-
`sis, the patient’s PBMC were used to derive autologous dendritic
`cell and macrophage cultures and several cultures of allogeneic
`primary cells adapted for growth in culture by a commercial sup-
`plier were obtained. Coculture results presented in Figure 4 con-
`firm the data obtained in the certificate of analysis. There was no
`reactivity seen to the dendritic cell and macrophage autologous
`cell cultures. Cytokine release was observed in several cocultures
`using allogeneic primary cells adapted for growth in culture.
`
`dIscussIon
`The ERBB2 gene has been extensively studied as a target for
`both chemotherapy and immunotherapy. In breast cancer,
`overexpression of ERBB2 is correlated with a poor clinical out-
`come and at the same time, is a positive predictive factor for
`those women who are most likely to respond to therapy with
`the anti-ERBB2 antibody, trastuzumab.10 In randomized clinical
`
`846
`
`www.moleculartherapy.org vol. 18 no. 4 apr. 2010
`
`UPenn Ex. 2033
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`© The American Society of Gene & Cell Therapy
`
`Serious Adverse Event With ERBB2-based CAR
`
`table 2 Vector tissue distribution
`
`signal
`normal tissue
`0.00
`Brain (left frontal lobe)
`0.00
`Pectoralis muscle (left)
`0.01
`Aorta
`0.04
`Atrium (right)
`0.05
`Neck LN (left)
`0.08
`Ventricle (left)
`0.09
`Small bowel
`0.11
`Liver (left lobe)
`0.13
`Liver (right lobe)
`0.14
`Abdominal para-aortic LN
`0.15
`Liver
`0.15
`Auxiliary LN
`0.16
`Kidney (left)
`0.17
`Adrenal gland (left)
`0.25
`Kidney (right)
`0.30
`Spleen
`0.44
`Lung left—A
`0.49
`Adrenal gland (right)
`0.56
`Lung left—B
`0.70
`Mediastinal
`0.78
`Lung right—A
`1.59
`Lung right—B
`1.99
`Para-aortic/mediastinal LN
`2.24
`Abdominal para-aortic LN
`2.58
`Abdominal LN
`2.64
`Mediastinal LN
`signal
`Metastasis
`0.03
`Liver metastasis
`0.08
`Lung metastasis (left)
`0.09
`Liver metastasis
`0.11
`Liver metastasis (left lateral)
`0.29
`Lung metastasis (left)
`0.34
`Liver metastasis (left anterior)
`Abbreviations: CAR, chimeric antigen receptor; FACS, fluorescence-activated cell
`sorting; LN, lymph node; PBL, peripheral blood lymphocyte; Q-PCR, quantitative-
`PCR.
`DNA extracted from tissues and metastasizes at autopsy were subjected to Q-PCR
`using ErbB2 CAR-vector specific primers/probe. Values were normalized to the
`infusion PBL (arbitrarily assigned a value of 100). Gene transfer efficiency of the
`infusion PBL was 79% as measured by FACS.
`
`trials, trastuzumab in combination with chemotherapy lead
`to increased disease-free and overall survival in breast cancer
`patients.9,29,30 The mechanism of action of trastuzumab appears
`to be multifactorial, but there are several studies that indicated
`the involvement of cell- mediated immunity (natural killer
`cell–based antibody-dependent cellular cytotoxicity) in patient
`responses.31–33 Furthermore, patient immune responses (anti-
`body-dependent cellular cytotoxicity) can be increased when
`trastuzumab is combined with concomitant cytokine (IL-2,
`
`IL-12) administration,34,35 or in vaccine trials where anti-ERBB2
`T cells responses have been reported.11,13
`Safety considerations that preceded our clinical trial included
`the use of trastuzumab in thousands of cancer patients, the lack
`of toxicity seen in multiple studies immunizing against epitopes
`of ERBB2, and the lack of toxicity seen in a report of the adop-
`tive transfer of autologous anti-ERBB2 cytotoxic T lymphocyte
`clones in the setting of breast cancer.14 In this report, three dif-
`ferent cytotoxic T lymphocyte clones were administered in five
`transfers given 2 weeks apart. A total of 2.65 × 109 total cells
`were administered along with low-dose IL-2. With the excep-
`tion of low-grade fever and chills following the third and fourth
`infusions, no side effects were noted. The therapy was associated
`with a decrease in tumor cells within the patient’s bone mar-
`row, but larger metastatic sites (such as liver) were not impacted.
`Radioimaging was performed using 111In-labeled cytotoxic T
`lymphocyte that demon strated an immediate accumulation of
`cells in the lung that decreased over 72 hours, whereas uptake
`to the liver and spleen increased over the first 24 hours then
`remained stable for the 72-hour study period. We have demon-
`strated similar uptake of 111In-labeled TIL36 and have observed
`that lung uptake of TIL happens at the first pass (at the 90%
`retention level) after which the TIL then leak out of the lungs
`to the liver and other organs (ref. 36 and J.C. Yang, unpublished
`results).
`The γ-retroviral vector construct used in this cancer gene
`therapy trial was designed for optimal ERBB2-CAR gene expres-
`sion and anti-ERBB2 reactivity. It was demonstrated to be highly
`specific, was able to recognize a wide range of tumor histologies,
`and was able to significantly prevent the growth of human breast
`cancer cells orthotopically implanted into the mammary fat pad of
`severe combined immunodeficiency mice.28 Part of the process of
`optimizing this anti-ERBB2-CAR vector was the inclusion of two
`T-cell costimulatory domains from CD28 and 4-1BB (CD137)
`that were linked to the CD3z signaling element. 4-1BB is essential
`for the optimal activity of CD8+ T cells37,38 and inclusion of 4-1BB
`signaling domains was shown to enhance the in vivo antitumor
`activity of CARs in tumor xenograft models.28,39–41 To date, the
`reported clinical application of CAR-engineered T cells has been
`limited to constructs containing CD3z alone.24–27,42 In one report
`targeting carbonic anhydrase IX, on-target toxicity was observed
`most likely due to recognition of antigen expression on biliary epi-
`thelium. The results with the carbonic anhydrase IX directed CAR
`suggest that on-target toxicity may be antigen dependant and does
`not require the presence of costimulatory signals (such as CD28
`and 4-1BB) in the CAR construct.
`In our initial report we observed, in some transductions, T-cell
`recognition of the ERBB2−-tumor line MDA468 (this line is nega-
`tive for ERBB2 expression by fluorescence-activated cell sorting,
`but ERBB2 mRNA can be detected by PCR). Transduction of the
`current patient’s T cells with the identical ERBB2-CAR vector
`did not result in recognition of the MDA468 cell line (Table 1,
`Figure 4). The significance of the recognition of allogenic primary
`cells adapted for growth in culture (Figure 4) is not clear, as it is
`not known how the adaptation of these cells for growth in ex vivo
`culture influences the expression of ERBB2. For example, it was
`reported that while normal rat liver hepatocytes do not express
`
`Molecular Therapy vol. 18 no. 4 apr. 2010
`
`847
`
`UPenn Ex. 2033
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Serious Adverse Event With ERBB2-based CAR
`
`© The American Society of Gene & Cell Therapy
`
`IL-10
`−1082A
`−819T
`
`TGFβ1
`25G
`
`TGFβ1
`25C
`
`TGFβ1
`10C
`
`TGFβ1
`10T
`
`TNFα
`−308G
`
`TNFα
`−308A
`
`Neg
`cntl
`
`IFNγ
`874A
`
`IFNγ
`874T
`
`IL-6
`−174G
`
`IL-6
`−174C
`
`IL-10
`−819C
`−592C
`
`IL-10
`−819T
`−592A
`
`IL-10
`−1082A
`−819C
`
`IL-10
`−1082G
`−819C
`
`β-Globin control
`
`Cytokine SNP
`
`Primers
`
`Figure 3 cytokine genotype. DNA extracted for patient PBMC was subject to PCR with sequence-specific primers (PCR-SSP) as described in
`Materials and Methods section. Primer pairs are designed to have perfect matches only with a single allele or group of alleles. Matched primer pairs
`result in the amplification of target sequences (i.e., a positive amplification band), whereas mismatched primer pairs do not result in amplification
`(i.e., a negative result). The specific genotypes detected by the SSP are shown above each lane (Neg cntl, reaction without cytokine primers). Shown
`is one of duplicate determinations. IFN-γ, interferon-γ; IL, interleukin; SNP, single-nucleotide polymorphism; TNF-α, tumor necrosis factor-α.
`
`with anti-CD28 mAb TGN1412.48–50 In that report,50 TNF-α lev-
`els peaked within 1 hour after infusion and IL-2, IL-6, IL-10, and
`IFN-γ reached maximum levels at the next time point, 4 hours
`after infusion (elevation in other cytokines included IL-4, IL-8,
`IL-12, and IL-1β). All six patients in that study required sup-
`portive care in an intensive care unit and two of the six required
`extensive intensive care unit stays of 11 and 21 days. Similar to
`patients suffering serious adverse events from other mAb infu-
`sions, these patients showed signs of cardiovascular instability
`and disseminated intravascular coagulation, but unlike other
`treatments, these patients manifested additional sequelae includ-
`ing early and acute lung injury. Of interest was the lack of toxic-
`ity seen with this agent in preclinical tests of the anti-CD28 mAb
`TGN1412 in nonhuman primate studies.48,49
`Perhaps the most relevant to our patient, were results observed
`in a phase I trial of a bispecific antibody that targeted both ERBB2
`and FcγRIII.51 In this study, attempting to target FcγRIII-expressing
`cells (e.g., natural killer cells) to ERBB2-expressing tumors, some
`patients experienced side effects including dyspnea with arterial
`oxygen desaturation and hypotension. Analysis of serum cytokine
`levels demonstrated an increase in TNF-α within 30 minutes that
`peaked at 2.3 hours before declining over the next 24 hours with
`slightly delayed increases in IL-6, IL-8, IL-2, IL-1β, GM-CSF, and
`IFN-γ. A major difference between therapeutic antibody admin-
`istration and CAR-engineered T cells, is that while antibodies are
`subject to clearance by the body (e.g., the bispecific mAb used in
`the cited trial had a t1/2 of 20 hours), T cells can continuously pro-
`duce effector cytokines and can expand in cell numbers following
`antigen stimulation. The severity of this patients’ response lead
`us to investigate her cytokine genotype (Figure 3), as poly-
`morphisms in cytokine genes are associated with both levels of
`cytokine production and immunological responses, such as organ
`graft rejection.52–54 Although her genotype for IFN-γ (−874A) and
`TNF-α (−308G) are associated with lower levels of cytokine pro-
`duction, her IL-6, IL-10, and transforming growth factor-β1 geno-
`types are associated with higher levels of cytokine production, and
`the specific IL-6 (−174G/C) and the IL-10 (−1082G) genotypes
`are associated with shock in patients with sepsis55 and increased
`mortality in severe sepsis,56 respectively.
`Since 2004, the Surgery Branch, NCI has conducted clini-
`cal trials involving the transfer of T-cell receptor genes into
`auto logous lymphocytes (either PBL or TIL) using the identical
`
`218,202
`
`UnTd
`CAR
`
`50,000
`
`40,000
`
`30,000
`
`20,000
`
`10,000
`
`0
`
`IFN-γ (pg/ml)
`
`SK-OV-3
`SK-BR-3
`MDA-361
`MDA-468
`CEM
`
`Auto-Mac
`Auto-DC
`
`HRE
`HMEC
`Keratinocyte
`HSMM
`PrEC
`NHBE
`EBV-B
`HUVEC
`HDF
`Media
`
`Figure 4 In vitro cytokine production. An aliquot of the ErbB2-CAR
`transduced (CAR) T lymphocytes infused into the patient or untrans-
`duced control T cells (UnTd) where assayed for IFN-γ cytokine produc-
`tion following overnight coculture with the indicated cell lines. ErbB2+
`target cells were SK-OV3, SK-BR3, and MDA361. ErbB2− tumor lines
`were MDA468 and CCRF-CEM (CEM). Primary cells adapted for growth
`in culture were HDF, human diploid fibroblast, HUVEC, human umbilical
`vein endothelial cells; EBV-B-EBV transformed B cell line, NHBE, normal
`human bronchial/tracheal epithelial cells; PrEC, human prostate epithe-
`lial cells, HSMM, human skeletal muscle myoblasts, keratinocytes-human
`keratinocytes, HMEC, human mammary epithelial cells, and HRE, human
`renal epithelial cells. Autologous patient cells were auto-DC-patient 6-day
`dendritic cell culture, and auto-mac-patient 6-day macrophage culture.
`All samples were diluted as necessary as to be in the linear range of the
`assay (sample SK-OV3 was off-scale in this assay and the value from a
`repeat determination was as indicated). CAR, chimeric antigen receptor;
`IFN-γ, interferon-γ.
`
`ERBB2, that upon ex vivo culture, ERBB2 expression was rapidly
`induced.43
`The most compelling finding in this case was the rapid rise
`in serum cytokine levels that has been associated with a multiple
`organ dysfunction syndrome.44–46 The administration of biolog-
`ics resulting in cytokine release syndrome was first reported to
`be associated with the administration of anti-CD3 mAb OTK3,
`which was administered as a systemic immunosuppressive agent
`during organ transplantation.47,48 Within 1–4 hours after OKT3
`injection, serum levels of proinflammatory cytokines such as
`TNF-α, IFN-γ, and IL-6 were markedly elevated. Most recently a
`cytokine storm was reported in six of six patients that were treated
`
`848
`
`www.moleculartherapy.org vol. 18 no. 4 apr. 2010
`
`UPenn Ex. 2033
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`© The American Society of Gene & Cell Therapy
`
`Serious Adverse Event With ERBB2-based CAR
`
`nonmyeloablative conditioning regimen used in the present study
`(in some cases total body irradiation was added to further reduce
`endogenous lymphocytes). These treatments were undertaken in a
`variety of cancers ( melanoma, synovial cell sarcoma, and cancers of
`the breast, colon, and kidney) and there was no indication that this
`conditioning regime was associated with the tumor lysis syndrome
`most frequently observed in cancers of hematopoietic origin.57
`Similar numbers of cells were administered in these trials as well as
`the present trial, and review of the certificates of analysis of these
`products demonstrated similar levels of background cytokine pro-
`duction. A total of 143 patients were treated during this period
`without any treatment related mortality. These trials, included 11
`patients treated with PBL engineered to target the wild-type p53
`tumor suppressor protein,58,59 and no significant treatment related
`toxicity was seen (data not shown). In our recent report describing
`the use of high-avidity T-cell receptors targeting melanocyte dif-
`ferentiation antigens, on-target toxicity was

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