throbber
Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`Plenary paper
`
`B-cell depletion and remissions of malignancy along with cytokine-associated
`toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor–transduced
`T cells
`James N. Kochenderfer,1 Mark E. Dudley,2 Steven A. Feldman,2 Wyndham H. Wilson,3 David E. Spaner,4 Irina Maric,5
`Maryalice Stetler-Stevenson,6 Giao Q. Phan,2 Marybeth S. Hughes,2 Richard M. Sherry,2 James C. Yang,2
`Udai S. Kammula,2 Laura Devillier,2 Robert Carpenter,1 Debbie-Ann N. Nathan,2 Richard A. Morgan,2 Carolyn Laurencot,2
`and Steven A. Rosenberg2
`
`1Experimental Transplantation and Immunology Branch, 2Surgery Branch, and 3Metabolism Branch, National Cancer Institute (NCI), Bethesda, MD;
`4Sunnybrook Health Sciences Center, Toronto, ON; 5Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD; and
`6Laboratory of Pathology, NCI, Bethesda, MD
`
`We conducted a clinical trial to assess
`adoptive transfer of T cells genetically
`modified to express an anti-CD19 chime-
`ric Ag receptor (CAR). Our clinical proto-
`col consisted of chemotherapy followed
`by an infusion of anti–CD19-CAR–
`transduced T cells and a course of IL-2.
`Six of the 8 patients treated on our proto-
`col obtained remissions of
`their ad-
`vanced, progressive B-cell malignancies.
`Four of the 8 patients treated on the
`protocol had long-term depletion of nor-
`mal polyclonal CD19ⴙ B-lineage cells.
`Introduction
`
`Cells containing the anti-CD19 CAR gene
`were detected in the blood of all patients.
`Four of the 8 treated patients had promi-
`nent elevations in serum levels of the
`inflammatory cytokines IFN␥ and TNF.
`The severity of acute toxicities experi-
`enced by the patients correlated with
`serum IFN␥ and TNF levels. The infused
`anti–CD19-CAR–transduced T cells were
`a possible source of these inflammatory
`cytokines because we demonstrated pe-
`ripheral blood T cells that produced TNF
`and IFN␥ ex vivo in a CD19-specific man-
`
`ner after anti–CD19-CAR–transduced
`T-cell
`infusions. Anti–CD19-CAR–trans-
`duced T cells have great promise to im-
`prove the treatment of B-cell malignan-
`cies because of a potent ability to
`eradicate CD19ⴙ cells in vivo; however,
`reversible cytokine-associated toxicities
`occurred after CAR–transduced T-cell in-
`fusions. This trial was registered with
`ClinicalTrials.gov
`as NCT00924326.
`(Blood. 2012;119(12):2709-2720)
`
`Chimeric Ag receptors (CARs) are fusion proteins that incorporate Ag
`recognition moieties and T-cell activation domains.1-3 The Ag recogni-
`tion moieties of CARs are usually variable regions of mAbs.1-3 T cells
`genetically modified to express CARs acquire the ability to
`specifically recognize targeted Ags.2-8 CD19 is a protein that is
`expressed on almost all B-lineage cells.9 Because expression of
`CD19 is limited to normal and malignant B-lineage cells, CD19
`is an attractive target for immunotherapies aimed at B-cell
`malignancies.9 Many groups have conducted preclinical experi-
`ments with T cells expressing anti-CD19 CARs, and these experi-
`ments have shown that anti–CD19-CAR–expressing T cells can
`recognize and destroy target cells in a CD19-specific manner.10-18
`The CARs used in these experiments have contained T-cell
`activation domains from molecules such as CD3␨ and a variety of
`costimulatory domains such as those from CD28 and 4-1BB.12-17
`Murine studies have shown that syngeneic T cells genetically
`modified to express anti-CD19 CARs can cure lymphoma and
`cause long-term eradication of normal B cells.19,20 Based on these
`preclinical experiments, clinical trials of anti-CD19 CARs have
`been initiated, and some early results from these trials have been
`reported.21-27 Similar to the murine studies, these early clinical
`reports have suggested an anti-malignancy effect of T cells express-
`ing anti-CD19 CARs, and Ag-specific eradication of normal B cells
`has been demonstrated.21,23,24,27
`
`Significant toxicities including hypotension, fevers, and renal
`insufficiency have occurred after infusions of anti–CD19-CAR–
`expressing T cells.22-24,27 Three patients with elevations in serum
`levels of inflammatory cytokines such as IFN␥ after anti–CD19-
`CAR–transduced T-cell infusions have been reported22-24; however,
`in one of these cases,
`the elevation in serum inflammatory
`cytokines was present before CAR-transduced T cells were in-
`fused.22 Determining the causes of elevated cytokine levels after
`anti–CD19-CAR–transduced T-cell infusions is not straightforward
`because only a small number of patients with elevated serum
`cytokine levels have been reported, and there are other possible
`causes of elevated serum cytokines such as sepsis.28 Inflammatory
`cytokines such as IFN␥ and TNF (formerly known as TNF␣) are
`produced by anti–CD19-CAR–transduced T cells in vitro.10,12,15
`IFN␥ and TNF can cause significant
`toxicity in humans29-32;
`however, an association between inflammatory cytokine produc-
`tion by anti–CD19-CAR–transduced T cells and clinical toxicity
`has not been demonstrated. A better understanding of the relation-
`ship between cytokine production by CAR-transduced T cells and
`clinical toxicity is necessary to rationally plan future research
`aimed at
`increasing the safety of anti–CD19-CAR–transduced
`T cells.
`We are conducting a clinical trial to assess the anti-malignancy
`efficacy, toxicity, and in vivo persistence of T cells transduced with
`
`Submitted October 6, 2011; accepted December 5, 2011. Prepublished online
`as Blood First Edition paper, December 8, 2011; DOI 10.1182/blood-2011-10-
`384388.
`
`There is an Inside Blood commentary on this article in this issue.
`
`The online version of this article contains a data supplement.
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`2709
`
`UPenn Ex. 2020
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`2710
`
`KOCHENDERFER et al
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`Figure 1. Anti–CD19-CAR–transduced T-cell produc-
`tion and clinical treatment protocols. (A) PBMCs were
`stimulated with the anti-CD3 mAb OKT3 on day 0. The
`cells were transduced with gammaretroviruses encoding
`the anti-CD19 CAR on days 2 and 3. On day 10, a rapid
`expansion protocol was started, and the cells were ready
`for infusion on day 24. (B) Patients received 60 mg/kg
`cyclophosphamide chemotherapy daily for 2 days. Next,
`patients received 25 mg/m2 fludarabine chemotherapy
`daily for 5 days. One day later, the patients received a
`single infusion of anti–CD19-CAR–transduced T cells.
`Starting on the same day as the T-cell
`infusion, the
`patients received IV IL-2 every 8 hours.
`
`Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`an anti-CD19 CAR. All of the patients on our clinical trial had
`advanced, progressive B-cell malignancies that were incurable by
`any standard treatment except allogeneic stem cell transplantation.
`Six of the 8 patients treated on our trial obtained objective
`remissions of their malignancies, and 4 of 8 patients had long-term
`elimination of CD19⫹ B-lineage cells. Significant toxicities that
`correlated with elevations in serum IFN␥ and TNF occurred after
`infusions of anti–CD19-CAR–transduced T cells. In addition, we
`demonstrated CD19-specific IFN␥ and TNF production by T cells
`from the blood of patients who had received infusions of anti–CD19-
`CAR–transduced T cells.
`
`Methods
`
`Clinical trial design
`
`The trial was reviewed by the US Food and Drug Administration and the
`Institutional Review Board of the National Cancer Institute and allowed to
`proceed. Patients provided written informed consent before participation in this
`study in accordance with the Declaration of Helsinki. Patients underwent an
`apheresis to obtain PBMCs for producing anti–CD19-CAR–transduced T cells.
`Cyclophosphamide was administered daily on days ⫺7 and ⫺6 at a dose of
`60 mg/kg. On days ⫺5 through ⫺1, patients received 25 mg/m2 fludarabine
`daily. On day 0, patients received a single infusion of CAR-transduced T cells.
`Three hours after CAR-transduced T cells were administered, IL-2 was initiated.
`IL-2 was administered intravenously at a dose of 720 000 international units/kg
`every 8 hours until toxicity precluded additional doses. Remissions of chronic
`lymphocytic leukemia (CLL) or lymphoma were defined according to standard
`international criteria.33,34
`
`In vitro and ex vivo assays
`
`Anti-Fab Ab staining and staining with labeled CD19 protein were used in
`flow cytometry to detect surface expression of the anti-CD19 CAR as
`described in the supplemental Methods. ELISAs, intracellular cytokine
`staining assays (ICCS), and CD107a degranulation assays were performed
`as described in supplemental Methods and as previously described.12,21
`Serum ELISAs were carried out as described in supplemental Methods.
`Immunohistochemistry and flow cytometry were carried out as detailed in
`the supplemental Methods and as previously described.21
`
`Real-time qPCR
`
`Real-time quantitative PCR (qPCR) was carried out to determine the
`percentage of peripheral blood mononuclear cells containing the CAR gene
`as described in supplemental Methods.
`
`Calculation of SOFA scores
`
`The sequential organ failure assessment (SOFA) score is an established
`method of quantifying the overall severity of illness.36 The SOFA score
`includes an assessment of hypotension, the platelet count, and measure-
`ments of respiratory, liver, renal, and central nervous system func-
`tion.36,37 We calculated daily SOFA scores for each patient by using
`clinical records from the day of CAR-tranduced T-cell infusion and each
`of the first 10 days after CAR-transduced T-cell infusion.36 For each
`patient, the sum of the SOFA scores from each day was calculated to
`give the total SOFA score.36
`
`Results
`
`Anti-CD19 CAR retroviral vector design
`
`Production of anti–CD19-CAR–transduced T cells
`
`We previously reported the design and construction of the murine stem cell
`virus–based splice-gag vector (MSGV)–FMC63-28Z that encoded the
`anti-CD19 CAR used in our clinical trial.12
`
`Anti–CD19-CAR–transduced T cell preparation
`
`Anti–CD19-CAR–transduced T cells were prepared as described in supple-
`mental Methods (available on the Blood Web site; see the Supplemental
`Materials link at the top of the online article) and as previously described.35
`
`Autologous PBMCs were stimulated with an anti-CD3 mAb and
`transduced with gammaretroviruses encoding an anti-CD19
`CAR (Figure 1A). The anti-CD19 CAR used in our clinical trial
`contained the variable regions of a murine anti–human-CD19
`Ab, a portion of the CD28 molecule, and the signaling domain of
`the CD3␨ molecule.12 The anti-CD19 CAR could be detected on
`the surface of transduced T cells by flow cytometry (Figure 2A).
`For the 8 treated patients,
`the mean percentage of T cells
`
`UPenn Ex. 2020
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`ANTI-CD19 CAR CLINICAL TRIAL
`
`2711
`
`Figure 2. Anti–CD19-CAR–transduced T cells pro-
`duced cytokines in a CD19-specific manner and
`recognized autologous leukemia cells. (A) Staining
`with an anti-Fab Ab revealed expression of the anti-
`CD19 CAR on the surface of T cells that were adminis-
`tered to patient 7. Staining with an isotype control Ab is
`also shown. Both plots were gated on CD3⫹ lympho-
`cytes, which made up 99% of the cells in the culture.
`(B) On the day of
`infusion, T cells of patient 7 up-
`regulated CD107a expression after a 4-hour culture with
`the CD19⫹ target cell CD19-K562 but not the negative
`control cell NGFR-K562 that does not express CD19.
`(C) On the day of infusion, anti–CD19-CAR–transduced
`T cells of patient 7 produced IFN␥, TNF, and IL-2 when
`cultured for 6 hours with the CD19⫹ target cell CD19-
`K562 but not the negative control cell NGFR-K562 that
`does not express CD19. The results shown in panels A
`through C are representative of the results obtained for
`all of the patients on the protocol. (D) Anti–CD19-CAR–
`transduced T cells of patient 3 were cultured with either
`autologous pretreatment lymphocytes or autologous re-
`mission lymphocytes overnight, and an IFN␥ ELISA was
`performed on the supernatant. Anti–CD19-CAR–
`transduced T cells of patient 3 specifically recognized
`pretreatment
`lymphocytes but not remission lympho-
`cytes obtained 7 weeks after CAR-transduced T-cell
`infusion. Sixty-four percent of the pretreatment lympho-
`cytes were CD19⫹ leukemia cells. The remission lympho-
`cytes contained only 0.1% CD19⫹ cells. (E) Anti–CD19-
`CAR–transduced T cells of patient 6 were cultured with
`either pretreatment autologous lymphocytes or autolo-
`gous remission lymphocytes overnight and an IFN␥
`ELISA was performed on the supernatant. Anti–CD19-
`CAR–transduced T cells of patient 6 specifically recog-
`nized pretreatment lymphocytes but not remission lym-
`phocytes obtained 2 weeks after CAR–transduced T-cell
`infusion. Seventy-six percent of the pretreatment lympho-
`cytes were CD19⫹ leukemia cells. The remission lympho-
`cytes contained only 0.1% CD19⫹ cells. In both panels D
`and E, pretreatment lymphocytes cultured alone did not
`produce detectable quantities of IFN␥.
`
`expressing the anti-CD19 CAR at the time of infusion was 55%
`(Table 1). Most of the infused CAR-transduced cells were
`CCR7-negative, CD45RA-negative effector memory cells, but
`variable numbers of CCR7⫹, CD45RA-negative central memory
`
`cells were also present (Table 2). The CAR-expressing T cells
`specifically up-regulated CD107a when cultured with CD19-
`expressing target cells but not when cultured with negative
`control target cells that lacked CD19 expression (Figure 2B).
`
`Table 1. Patient data
`
`Patient Age, y
`
`Malignancy
`
`Total no. of
`infused
`cells/kg,
`ⴛ107
`
`No. of prior
`therapies
`
`Percentage of
`infused cells
`CARⴙ
`
`No. of infused
`CARⴙ cells/kg
`ⴛ107
`
`Infused cells
`CD4/CD8
`ratio
`
`Doses of IL-2
`administered
`
`Response and time
`since treatment, mo*
`
`1a†
`1b†
`2
`3
`4
`
`5
`6
`7
`8
`
`47
`48
`48
`61
`55
`
`54
`57
`61
`63
`
`Follicular lymphoma
`Follicular lymphoma
`Follicular lymphoma
`CLL
`Splenic marginal zone
`lymphoma
`CLL
`CLL
`CLL
`Follicular lymphoma
`
`4
`5
`5
`3
`3
`
`4
`7
`4
`7
`
`0.5
`2.1
`0.5
`2.5
`2.0
`
`0.6
`5.5
`5.4
`4.2
`
`64
`63
`65
`45
`53
`
`50
`30
`51
`71
`
`0.3
`1.3
`0.3
`1.1
`1.1
`
`0.3
`1.7
`2.8
`3.0
`
`29/63
`19/71
`23/73
`35/53
`72/24
`
`87/12
`37/57
`58/41
`54/43
`
`8
`10
`9
`2
`4
`
`2
`1
`2
`5
`
`PR (7)
`PR (18⫹)
`NE (died with influenza)
`CR (15⫹)
`PR (12)
`
`SD (6)
`PR (7)
`PR (7⫹)
`PR (8⫹)
`
`PR indicates partial remission; NE, not evaluable; CR, complete remission; SD, stable disease; and CLL, chronic lymphocytic leukemia.
`*Time since treatment is in months; ⫹ indicates an ongoing response as of the time of writing. The body surface area of each patient is given in supplemental Table 2.
`†Patient 1 was treated twice. His first treatment has been previously reported (21).
`
`UPenn Ex. 2020
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`2712
`
`KOCHENDERFER et al
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`Table 2. Memory phenotype of infused anti-CD19 CAR-expressing T cells
`CCR7ⴙCD45RAⴙ
`CCR7ⴙCD45RAⴚ
`Patient
`CD62L
`
`CCR7ⴚCD45RAⴚ
`
`CCR7ⴚCD45RAⴙ
`
`1a*
`1b*
`2
`3
`4
`5
`6
`7
`8
`
`21
`20
`25
`27
`35
`26
`10
`4
`8
`
`7
`8
`5
`1
`0
`3
`6
`4
`1
`
`30
`20
`21
`8
`5
`13
`27
`27
`9
`
`53
`55
`61
`87
`90
`71
`57
`60
`81
`
`10
`18
`13
`5
`5
`13
`10
`9
`8
`
`The phenotype of the infused CD19 CAR-expressing T cells was determined by flow cytometry. Values are the percentage of the CAR⫹ CD3⫹ cells that expressed the
`indicated markers.
`*Patient 1 was treated twice.
`
`Up-regulation of CD107a indicated degranulation, which is a
`prerequisite for perforin-mediated cytotoxicity.38 The CAR-
`transduced T cells produced IFN␥, TNF, and IL-2 in a CD19-
`specific manner (Figure 2C, Table 3).
`
`Anti–CD19-CAR–transduced T cells can specifically recognize
`autologous leukemia cells
`
`The blood of some of the patients on our trial contained large
`numbers of CD19⫹ CLL cells. Access to pretreatment blood
`samples from these patients allowed us to determine whether
`anti–CD19-CAR–transduced T cells could specifically recognize
`unmanipulated autologous CLL cells. The blood lymphocytes of
`patient 3 contained 64% CD19⫹ CLL cells before treatment on our
`protocol, and the blood lymphocytes of patient 6 contained 76%
`CD19⫹ CLL cells before treatment on our protocol. After treatment
`when the patients were in remission, the blood lymphocytes of both
`patient 3 and patient 6 contained only 0.1% CD19⫹ cells. As shown
`in Figure 2D and E, the anti–CD19-CAR–transduced T cells from
`each of these patients produced large amounts of IFN␥ when cultured
`with the pretreatment lymphocytes that were mostly leukemia cells, but
`the CAR-transduced T cells produced only background levels of IFN␥
`when cultured with the lymphocytes that were obtained after treatment
`when the patients were in remission. IFN␥ was not produced by the
`leukemia cells of either patient (data not shown).
`
`Six of the 8 treated patients obtained objective remissions
`
`The 8 patients treated on our protocol had either B-cell lymphoma or
`CLL (Table 1). The patients all had progressive malignancy at the time
`of enrollment on our protocol despite a median of 4 prior therapies. All
`patients treated on our protocol received cyclophosphamide daily for
`
`2 days followed by fludarabine daily for 5 days. One day after the last
`dose of fludarabine, the patients received a single IV infusion of
`anti–CD19-CAR–transduced T cells (Figure 1B). Three hours after the
`CAR-transduced T-cell infusion, a course of IV IL-2 was initiated
`(Figure 1B). The patients received doses of CAR-expressing T cells that
`ranged from 0.3 ⫻ 107 to 3.0 ⫻ 107 CAR⫹ T cells/kg bodyweight.
`Patient 1 was treated twice. His first treatment course was previously
`reported.21 Patient 1 developed progressive CD19⫹ lymphoma 7 months
`after his first infusion of anti–CD19-CAR–transduced T cells. After his
`lymphoma progressed, patient 1 was treated a second time with the
`same regimen. He remains in a partial remission (PR) 18 months after
`the second treatment. Patient 2 died 18 days after CAR-transduced
`T-cell infusion with culture-proven influenza A pneumonia, nonbacterial
`thrombotic endocarditis, and cerebral infarction, so he is not evaluable
`for lymphoma response. Overall, 6 of the 7 evaluable patients treated on
`our trial obtained objective remissions (Table 1). Because the patients
`received chemotherapy with activity against B-cell malignancies imme-
`diately before the anti–CD19-CAR–transduced T-cell infusion, the
`contribution that the CAR-transduced T cells made to the remissions is
`unclear.
`
`Patient 3 had a prolonged complete remission and depletion of
`normal B cells
`
`At the time of enrollment on our protocol, patient 3 had progressive
`CLL after receiving 3 prior therapies. Just before treatment on our
`protocol, he had a blood B-cell count of 1544 B cells/␮L (Figure
`3A). Ninety-six percent of the blood B cells were CLL cells. In
`addition, 50%-60% of his cellular BM was CLL (Figure 3B-C). A
`prominent clonal leukemia population that expressed the character-
`istic CD19⫹ and CD5⫹ phenotype of CLL was detected by flow
`cytometry of BM cells before treatment on our protocol (Figure
`
`Table 3. Cytokine production by infused cells
`IFN␥, pg/mL
`
`TNF, pg/mL
`
`IL-2, pg/mL
`
`Patient
`
`CD19-positive target
`
`CD19-negative target
`
`CD19-positive target
`
`CD19-negative target
`
`CD19-positive target
`
`CD19-negative target
`
`1*
`2
`3
`4
`5
`6
`7
`8
`
`8190
`9850
`19 000
`27 900
`36 700
`14 800
`29 300
`9960
`
`411
`506
`916
`944
`734
`130
`341
`240
`
`448
`6250
`9312
`21 895
`21 515
`8288
`21 980
`9830
`
`⬍ 31
`⬍ 31
`⬍ 31
`⬍ 31
`⬍ 31
`⬍ 31
`⬍ 31
`73
`
`1156
`2002
`1683
`2768
`2421
`798
`1661
`1697
`
`48
`139
`51
`40
`32
`44
`36
`46
`
`Levels of the indicated cytokines were determined by standard ELISAs after an overnight culture of T cells from the time of infusion with either CD19-positive target cells or
`CD19-negative target cells. NGFR-K562 cells were used as CD19-negative target cells for all cultures. CD19-K562 cells were used as CD19-positive targets for the cultures
`preceding the TNF ELISAs. NALM6 cells were used as the CD19-positive target cells for the cultures preceding the IFN␥ and IL-2 ELISAs.
`*Results are from the second treatment of patient 1.
`
`UPenn Ex. 2020
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`ANTI-CD19 CAR CLINICAL TRIAL
`
`2713
`
`Figure 3. Normal and malignant B-lineage cells were eliminated from
`the blood and BM of patient 3. (A) Before treatment, the blood of patient
`3 contained an elevated number of B cells, 96% of which were leukemia
`cells. After treatment, the blood B-cell count has remained below normal
`and patient 3 has been in complete remission for 67 weeks. B cells were
`quantitated by flow cytometry staining of CD19⫹ cells. (B) CD19 IHC
`staining of the BM of patient 3 is shown before treatment and 13 weeks
`after treatment. The BM contained large numbers of CD19⫹ cells before
`treatment. Thirteen weeks after treatment, CD19⫹ cells were nearly
`absent. (C) CD20 IHC staining of the BM of patient 3 is shown before
`treatment and 13 weeks after treatment. The BM contained large numbers
`of CD20⫹ cells before treatment. Thirteen weeks after treatment, CD20⫹
`cells were nearly absent. (D) Flow cytometric results of a BM aspirate from
`patient 3 are shown. Plots are gated on lymphoid cells by forward and side
`scatter. A monoclonal population of B cells expressing the characteristic
`CD19⫹ and CD5⫹ phenotype of CLL (circled) was present before treat-
`ment but not 13 weeks after treatment.
`
`3D). After treatment, CLL was completely eradicated from the
`blood of patient 3, and the number of polyclonal blood B cells has
`levels of 17 to 40 B cells/␮L for
`stayed at below-normal
`⬎ 15 months (lower limit of the normal 61 B cells/␮L; Figure 3A).
`Patient 3’s blood B cells that have returned after treatment were
`determined to be polyclonal by flow cytometry staining for Ig ␬ and
`␭ proteins (data not shown). CLL was eliminated from the BM of
`patient 3 after treatment as shown by BM IHC staining (Figure
`3B-C) and by multicolor flow cytometry analysis of the BM cells
`(Figure 3D). B-lineage cells were nearly absent from the BM, but
`other hematopoietic cells had recovered (Figure 3B-C). Overall,
`the cellularity of the BM varied between 20% and 70%. A normal
`BM cellularity for a 61-year-old patient is ⬃ 40%. Patient 3
`continues in complete remission ⬎ 15 months after treatment on
`our protocol.
`
`adenopathy occurred during the time between a pretreatment
`computed tomography (CT) scan and a second CT scan that was
`performed 32 days after treatment (Figure 4A). Regression of the
`adenopathy continued between day 32 and day 132 after the
`CAR-transduced T-cell infusion. This continued regression, which
`occurred ⬎ 33 days after the last dose of chemotherapy suggested
`that the CAR-transduced T cells contributed to the regression of the
`adenopathy. In accordance with this possibility, anti–CD19-CAR–
`transduced T cells persisted in the blood of patient 7 until at least
`day 132 after infusion (Figure 5B). Because the patient received
`chemotherapy before the CAR-transduced T cells, an alternative
`explanation for the decreasing adenopathy between day 32 and
`132 after CAR-transduced T-cell infusion is continued resolution
`of adenopathy because of clearance of leukemia cells that were
`killed by the chemotherapy.
`
`Patient 7 had a substantial reduction in adenopathy after
`CAR-transduced T-cell infusion
`
`Before enrollment on our protocol, patient 7 had CLL that was
`progressive despite 4 prior therapies. Extensive regression of
`
`Patient 8 had a prolonged specific eradication of normal B cells
`
`Patient 8 had a normal level of polyclonal blood B cells before
`treatment on our protocol. After treatment, his blood B cells
`have been eliminated for 26 weeks as of his last follow-up
`
`UPenn Ex. 2020
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`2714
`
`KOCHENDERFER et al
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`Figure 4. Patients receiving infusions of anti–CD19-CAR–transduced
`T cells had reductions in adenopathy and elimination of normal
`B cells. CT scans of patient 7 showed extensive adenopathy before
`treatment. This adenopathy regressed by day 32 after CAR-transduced
`T-cell infusion (arrow). The enlarged lymph nodes continued to substan-
`tially regress between 32 and 132 days after CAR-transduced T-cell
`infusion. (B) Patient 8 had a normal number of polyclonal blood B cells
`before treatment. B cells were eliminated from the blood after treatment
`and had not recovered 26 weeks after anti–CD19-CAR–transduced T-cell
`infusion. B cell counts were determined by flow cytometry for CD19 and
`confirmed by flow cytometry for CD20. In contrast to the B cells, blood
`T-cell counts (C) and NK-cell counts (D) rapidly recovered after treatment.
`
`Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`(Figure 4B). The B-cell depletion in patient 8 was specific
`because blood T cells (Figure 4C) and NK cells (Figure 4D)
`recovered shortly after
`treatment. This specific,
`long-term
`elimination of normal CD19⫹ B cells indicates that anti–CD19-
`CAR–transduced T cells are destroying cells expressing the
`targeted CD19 Ag in vivo. Notably, this long-term elimination
`of B cells cannot be attributed to the chemotherapy that the
`patient received because we have previously shown that patients
`receiving the same chemotherapy regimen plus infusions of
`T cells targeting Ags that are not expressed on B cells recover
`normal blood B cell numbers in 8 to 19 weeks.21 Overall, we
`have noted B-cell depletion lasting at least 6 months in 4 of the
`8 patients treated on our protocol. As previously reported,
`patient 1 had a complete eradication of blood and BM B-lineage
`cells for 36 weeks after his first infusion of anti–CD19-CAR–
`transduced T cells.21 Nine weeks after his second treatment,
`patient 1 was found to have a normal number of polyclonal
`blood B cells. The reason for this recovery of normal B cells is
`unknown. We have noted prolonged B-cell depletion that lasted
`at least 6 months in 3 additional patients. Patient 7 had no
`detectable blood B cells at his last follow-up 7 months after
`infusion of anti–CD19-CAR–transduced T cells. The prolonged
`depressions of blood B cell numbers in patient 3 and patient 8
`are shown in Figures 3A and 4B, respectively. For all patients,
`the blood B-cells were quantitated 4 to 5 months after CAR-
`transduced T-cell infusion (supplemental Table 1).
`
`Varied levels of anti–CD19-CAR–transduced T cells could be
`detected in the blood of all patients
`
`To quantitate anti–CD19-CAR–transduced T cells in the blood of
`patients, we developed a qPCR assay that specifically detected the DNA
`sequence of the anti-CD19 CAR. Data from these quantitative PCR
`
`experiments were expressed as a percentage of total PBMCs that
`contained the anti-CD19 CAR gene. The persistence of cells containing
`the CAR gene varied widely, but in patient 1 through patient 6, cells
`containing the CAR gene could be detected at levels ⬎ 0.01% of total
`PBMCs for ⬍ 20 days (Figure 5A). In contrast, patient 7 and patient 8
`had high peak percentages and long-term persistence of PBMCs
`containing the CAR gene (Figure 5B). We confirmed the high levels of
`CAR-expressing cells in the blood of patient 7 and patient 8 by detecting
`CAR-expressing T cells with flow cytometry after staining with labeled
`CD19 protein (Figure 5C and D). We also detected cells containing the
`CAR gene in the BM of patient 7 and patient 8. As measured by qPCR,
`0.03% of the BM cells of patient 7 contained the CAR gene 14 weeks
`after CAR-transduced T-cell infusion, and 0.13% of the BM cells of
`patient 8 contained the CAR gene 8 weeks after CAR-transduced T-cell
`infusion.
`
`T cells that degranulated in a CD19-specific manner were
`detected in the blood of patient 7 ex vivo
`
`We demonstrated the presence of functional CD19-specific
`T cells in the blood of patient 7 by detecting T cells that
`up-regulated CD107a in a CD19-specific manner after anti–
`CD19-CAR–transduced T-cell infusion but not before CAR-
`transduced T-cell infusion (Figure 5E).
`
`Patients experienced significant toxicity that correlated with
`elevated levels of IFN␥ and TNF
`
`As mentioned previously, 4 of the 8 patients on our clinical trial
`had long-term depletion of normal B cells. In accordance with
`an eradication of B-lineage cells, all of these 4 patients have
`developed hypogammaglobulinemia and have subsequently been treated
`
`UPenn Ex. 2020
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/119/12/2709/1350836/zh801212002709.pdf by guest on 19 July 2022
`
`BLOOD, 22 MARCH 2012 䡠 VOLUME 119, NUMBER 12
`
`ANTI-CD19 CAR CLINICAL TRIAL
`
`2715
`
`Figure 5. Anti–CD19-CAR–transduced T cells can be
`detected in the blood of patients for up to 181 days
`after infusion. (A) The percentage of total PBMCs that
`contained the anti-CD19 CAR gene was determined by
`quantitative PCR. In patients 1 through 6, the peak levels
`and persistence of cells containing the CAR gene varied.
`(B) The percentage of total PBMCs from patient 7 and
`patient 8 that contained the anti-CD19 CAR gene was
`determined by quantitative PCR. Compared with the
`other patients, these patients had substantially greater
`persistence of cells that contained the CAR gene.
`(C) Anti-CD19 CAR expression was detected ex vivo on
`PBMCs of patient 7 by flow cytometry staining with
`labeled CD19 protein (CD19-DDK) 13 days after CAR-
`transduced T-cell
`infusion. Only background levels of
`cells expressing the anti-CD19 CAR were detected
`before treatment. (D) In patient 8, T cells expressing
`the anti-CD19 CAR were detected by flow cytometry
`staining with labeled CD19 protein 13 days after CAR-
`transduced T-cell infusion. Only background levels of
`cells expressing the anti-CD19 CAR were detected
`before treatment. (C-D) The plots are gated on CD3⫹
`lymphocytes. (E) PBMCs from patient 7 were cultured
`for 4 hours with either CD19⫹ CD19-K562 cells or
`NGFR-K562 control cells that do not express CD19.
`Before treatment, CD107a was not up-regulated on
`T cells after culture with either CD19-K562 cells or
`NGFR-K562 cells. In contrast, T cells from 8 days after
`infusion up-regulated CD107a after a 4-hour culture with
`CD19-K562 cells but not NGFR-K562 cells. All plots are
`gated on CD3⫹ lymphocytes.
`
`with infusions of IV Igs. B-cell depletion is a manageable form of
`autoimmunity, and it is not unexpected because autoimmunity has been
`observed after other immunotherapies.39 Except for B-cell depletion, the
`most prominent toxicities experienced by patients on our trial included
`hypotension, fevers, fatigue, renal failure, and obtundation (Table 4).
`These toxicities generally peaked during the first 8 days after CAR-
`transduced T-cell infusion and resolved completely over time. Many of
`the toxicities that were observed in our patients can be caused by
`cytokines such as IFN␥ and TNF. For example, TNF is a potent inducer
`of hypotension.30,31 IFN␥ can cause fever, fatigue, and myalgias.29,32 We
`have previously reported a patient who received an infusion of T cells
`transduced with an anti-ERBB2 (human epidermal growth factor
`receptor-2)–specific CAR without administration of exogenous IL-2.40
`This patient developed very high levels of serum inflammatory cyto-
`kines shortly after CAR-transduced T-cell infusion and later died.40 The
`course of this patient demonstrates that CAR-transduced T cells can
`cause large elevations in serum inflammatory cytokines.40 We have
`shown that the CAR-transduced T cells administered to our patients
`could produce IFN␥ and TNF in a CD19-specific manner in vitro
`(Figure 2, Table 3). To determine whether elevated levels of the
`inflammatory cytokines IFN␥ and TNF occurred in our patients after
`anti–CD19-CAR–transduced T-cell infusions, we performed ELISAs
`for IFN␥ and TNF on serum samples obtained before treatment and at
`multiple time-points after treatment (Figure 6). We found that the
`patients could be divided into 2 groups. One group, which included
`
`patients 1, 2, 4, and 5, did not have prominent elevations of serum IFN␥
`or TNF (Figure 6A and C). The other group, which included patients 3,
`6, 7, and 8, had prominent elevations in both IFN␥ and TNF during the
`first 10 days after anti–CD19-CAR–transduced T-cell infusion (Figure
`6B and D). We hypothesized that much of the toxicity that occurred in
`our patients was because of elevations in inflammatory cytokines such
`as IFN␥ and TNF. To quantify the toxicity experienced by our patients,
`we calculated SOFA scores for each patient.36,37 The SOFA score is an
`est

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