throbber
~ Ch.eek for updates]
`
`Downloaded from http://ashpublications.org/blood/article-pdf/116/20/4099/1489743/zh804610004099.pdf by guest on 12 January 2022
`
`GENE THERAPY
`
`Brief report
`Eradication of B-lineage cells and regression of lymphoma in a patient treated
`with autologous T cells genetically engineered to recognize CD19
`James N. Kochenderfer,1 Wyndham H. Wilson,2 John E. Janik,2 Mark E. Dudley,1 Maryalice Stetler-Stevenson,3
`Steven A. Feldman,1 Irina Maric,4 Mark Raffeld,3 Debbie-Ann N. Nathan,1 Brock J. Lanier,1 Richard A. Morgan,1 and
`Steven A. Rosenberg1
`
`1Surgery Branch, 2Metabolism Branch, and 3Laboratory of Pathology, National Cancer Institute, Bethesda, MD; and 4Department of Laboratory Medicine,
`Clinical Center, National Institutes of Health, Bethesda, MD
`
`Adoptive transfer of genetically modified
`T cells is an attractive approach for gener-
`ating antitumor immune responses. We
`treated a patient with advanced follicular
`lymphoma by administering a preparative
`chemotherapy regimen followed by au-
`tologous T cells genetically engineered
`to express a chimeric antigen receptor
`(CAR) that recognized the B-cell antigen
`CD19. The patient’s lymphoma under-
`went a dramatic regression, and B-cell
`Introduction
`
`precursors were selectively eliminated
`from the patient’s bone marrow after infu-
`sion
`of
`anti–CD19-CAR-transduced
`T cells. Blood B cells were absent for at
`least 39 weeks after anti–CD19-CAR-
`transduced T-cell infusion despite prompt
`recovery of other blood cell counts. Con-
`sistent with eradication of B-lineage cells,
`serum immunoglobulins decreased to
`very low levels after treatment. The pro-
`longed and selective elimination of B-
`
`lineage cells could not be attributed to
`the chemotherapy that the patient re-
`ceived and indicated antigen-specific
`eradication of B-lineage cells. Adoptive
`transfer of anti–CD19-CAR-expressing
`T cells is a promising new approach for
`treating B-cell malignancies. This study
`is registered at www.clinicaltrials.gov as
`#NCT00924326.
`(Blood. 2010;116(20):
`4099-4102)
`
`T cells can be genetically modified to express chimeric antigen
`receptors (CARs).1-5 CARs consist of an antigen-recognition
`moiety, such as antibody-derived, single-chain variable fragments,
`coupled to T-cell activation domains.1-4 T cells have been geneti-
`cally engineered to express CARs that can recognize a variety of
`tumor-associated antigens, including the B-lineage antigen CD19,
`in a non-human leukocyte antigen-restricted manner.4-15 Expres-
`sion of the cell-surface protein CD19 is restricted to normal mature
`B cells, malignant B cells, B-cell precursors, and plasma cells.16-19
`We have designed a CAR that targets CD19 and initiated a clinical
`trial of autologous T cells expressing this CAR (www.clinicaltrials.
`gov; #NCT00924326).
`
`Methods
`
`This clinical trial was approved by the National Cancer Institute Institu-
`tional Review Board. Design and construction of the mouse stem cell
`virus-based splice-gag retroviral vector MSGV-FMC63-28Z encoding the
`anti-CD19 CAR used in our clinical trial have been described (GenBank
`HM852952).7 The anti-CD19 CAR contains an antigen-recognition
`moiety consisting of the variable regions of the FMC63 monoclonal
`antibody joined to part of the CD28 molecule and the signaling domains
`of the CD3␨ molecule.
`Peripheral blood mononuclear cells were transduced with retroviruses
`encoding the anti-CD19 CAR and cultured in an almost identical manner as
`previously described.20 As measured by flow cytometry, the CAR was
`expressed on 64% of the infused cells, which were 98% CD3⫹ T cells
`(supplemental Figure 1, available on the Blood Web site; see the Supplemen-
`
`tal Materials link at the top of the online article). The T cells were 66%
`CD8⫹ and 34% CD4⫹. The anti–CD19-CAR-transduced T cells specifi-
`cally recognized CD19⫹ target cells (supplemental Table 1). Methods of
`T-cell preparation, flow cytometry, polymerase chain reaction, and immuno-
`histochemistry are in the supplemental data. For the immunohistochemistry
`images in Figures 1 and 2, images were obtained via digital microscopy
`using an Olympus BX51 microscope (Olympus America) equipped with a
`UPlanFL 10⫻/0.3 numeric aperture and UPlanFL 40⫻/0.75 numeric aperture
`objectives. Images were captured using an Olympus DP70 digital camera system.
`Imaging software was Adobe Photoshop CS3 (Adobe Systems).
`
`Results and discussion
`
`The patient was diagnosed with grade 1, stage IVB follicular
`lymphoma in 2002. Before enrollment on our protocol, he had
`received the following treatments for his lymphoma: PACE (pred-
`nisone, doxorubicin, cyclophosphamide, and etoposide), an idio-
`type vaccine, the anti–CTLA-4 monoclonal antibody ipilimumab,
`and EPOCH-R (etoposide, prednisone, vincristine, cyclophospha-
`mide, doxorubicin, and rituximab). The last cycle of EPOCH-R
`was administered in January 2008. The EPOCH-R caused a partial
`remission; however, progressive disease was noted in July 2008.
`The patient received no further treatment before he was evaluated
`for enrollment on our trial of anti–CD19-CAR-transduced T cells.
`When we evaluated the patient in May 2009, he had progressive
`lymphoma that involved all major lymph node areas (Figure 1A).
`He had bilateral pleural effusions, night sweats, and a recent weight
`loss of 10 pounds. Flow cytometry of a fine needle aspirate from an
`
`Submitted April 23, 2010; accepted July 20, 2010. Prepublished online as
`Blood First Edition paper, July 28, 2010; DOI 10.1182/blood-2010-04-281931.
`
`The online version of this article contains a data supplement.
`
`The publication costs of this article were defrayed in part by page charge
`payment. Therefore, and solely to indicate this fact, this article is hereby
`marked ‘‘advertisement’’ in accordance with 18 USC section 1734.
`
`BLOOD, 18 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 20
`
`4099
`
`Miltenyi Ex. 1045 Page 1
`
`

`

`4100
`
`KOCHENDERFER et al
`
`BLOOD, 18 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 20
`
`A
`
`18 weeks after treatment
`
`Downloaded from http://ashpublications.org/blood/article-pdf/116/20/4099/1489743/zh804610004099.pdf by guest on 12 January 2022
`
`Figure 1. B-lineage cells, including B-cell precur-
`sors, were eradicated from the bone marrow after
`treatment with anti–CD19-CAR-transduced T cells.
`(A) Representative pretreatment computed tomography
`scan images and images from 18 weeks after treatment
`demonstrate regression of
`lymphoma masses in the
`chest and abdomen after treatment with chemotherapy
`followed by anti–CD19-CAR-transduced T cells plus IL-2.
`(B) Flow cytometric evaluation of a pretreatment bone
`marrow aspirate was conducted with a forward versus
`side light scatter analysis gate of lymphoid cells. The left
`upper quadrant contains CD19⫹ B-lineage cells (35% of
`lymphoid cells), and the right lower quadrant contains
`CD3⫹ T cells. (C) Flow cytometric evaluation of a pretreat-
`ment bone marrow aspirate with a CD19⫹ analysis gate
`is shown. ␬- and ␭-negative, CD19⫹, mostly immature
`B-lineage cells that are not part of the malignant lym-
`phoma clone are in the rectangle. The cells outside the
`rectangle are mostly lymphoma cells. (D) Flow cytomet-
`ric evaluation of a pretreatment bone marrow aspirate
`with a forward versus side light scatter analysis gate of
`lymphoid cells. Immature B-cell precursors in the oval
`are CD22⫹ and CD20⫺. (E) Flow cytometric evaluation
`of a pretreatment bone marrow aspirate with a forward
`versus side light scatter analysis gate of lymphoid cells.
`Immature B-cell precursors in the polyhedral demon-
`strate decreasing CD10 correlating with increasing CD20
`expression. (F) Flow cytometric evaluation of a bone
`marrow aspirate from 36 weeks after treatment with a
`forward versus side light scatter analysis gate of lym-
`phoid cells. CD19⫹ B-lineage cells are absent.
`(G) Immunohistochemistry staining of a pretreatment
`bone marrow biopsy reveals a large population of CD19⫹
`cells that includes lymphoma cells as well as nonmalig-
`nant B-lineage cells. (H) Immunohistochemistry staining
`of a bone marrow biopsy from 36 weeks after infusion of
`anti–CD19-CAR-transduced T cells demonstrates a com-
`plete absence of CD19⫹ cells. (I) High-power view of the
`same anti-CD19 staining shown in panel H.
`
`B
`
`a,
`,-
`C
`
`(.) -
`
`..
`~!· ·:
`CD3
`
`,..,• • ·
`
`E
`
`C
`,-
`C
`(.)
`
`~·: :
`
`H CD19
`
`,..
`
`. . :,,:9f.;:
`•
`'l(:· ·-,i~~1
`Kappa
`
`CD20
`
`C
`
`I'll
`'tJ
`.0
`E
`I'll
`.J
`
`F
`
`O>
`,-
`C
`(.)
`
`CD3
`
`' -
`
`enlarged cervical lymph node demonstrated a monoclonal B-cell
`process consistent with follicular lymphoma that uniformly ex-
`pressed CD19, CD20, CD22, CD10, and IgM-kappa. Flow cytom-
`etry showed that 14.5% of the blood lymphoid cells had a
`phenotype that was consistent with the lymphoma and 0.7% of the
`blood lymphoid cells were normal polyclonal B cells (data not
`shown). Before treatment, 35% of bone marrow lymphoid cells
`expressed CD19 (Figure 1B). A total of 55% of these CD19⫹ cells
`were monoclonal ␬-positive and ␭-negative lymphoma cells; 45%
`of the bone marrow CD19⫹ cells were normal surface-immuno-
`globulin (Ig)–negative immature B-cell precursors (Figure 1C).
`The immature B-cell precursors demonstrated a pattern of antigen
`expression consistent with normal maturation, namely, CD22⫹
`B cells with decreasing CD10 expression correlating with increas-
`ing CD20 expression (Figure 1D-E).21,22 Large numbers of bone
`marrow CD19⫹ cells and CD79a⫹ cells were detected by immuno-
`histochemistry before treatment (Figures 1G, 2A).
`The patient underwent apheresis, and peripheral blood
`mononuclear cells were used to prepare anti–CD19-CAR-
`transduced T cells. The patient received a lymphocyte-depleting
`regimen consisting of 60 mg/kg cyclophosphamide daily for
`2 days followed by 5 daily doses of 25 mg/m2 fludarabine. The
`
`day after the last fludarabine dose, the patient received 1 ⫻ 108
`anti–CD19-CAR-transduced T cells intravenously. The next
`day, he received 3 ⫻ 108 anti–CD19-CAR-transduced T cells intra-
`venously. After the second anti–CD19-CAR-transduced T-cell
`infusion, the patient received 720 000 IU/kg interleukin-2 (IL-2)
`intravenously every 8 hours. Eight doses of IL-2 were adminis-
`tered. The only acute toxicities that the patient experienced were
`cytopenias that were attributable to chemotherapy and a fever that
`lasted 2 days (maximum temperature, 38.5°C). The patient was
`discharged 11 days after his second anti–CD19-CAR-transduced
`T-cell infusion, and he resumed full-time employment.
`After therapy, computed tomography scans revealed an impres-
`sive partial remission of the lymphoma that lasted 32 weeks (Figure
`1A); 32 weeks after treatment, progressive CD19⫹ lymphoma was
`noted in right cervical and retroperitoneal lymph nodes.
`Blood B cells were absent from 9 weeks after anti–CD19-CAR-
`transduced T-cell infusion until at least 39 weeks after anti–CD19-CAR-
`transduced T-cell infusion (Figure 2C; supplemental Figure 2). This
`prolonged B-cell depletion cannot be attributed to the chemotherapy that
`the patient received. Neither the New York esophageal squamous cell
`carcinoma antigen-1 (NY-ESO) nor the melanoma antigen gp100 is
`expressed by B cells.23,24 In prior clinical trials, patients treated with the
`
`Miltenyi Ex. 1045 Page 2
`
`

`

`BLOOD, 18 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 20
`
`ERADICATION OF B-LINEAGE CELLS
`
`4101
`
`A
`
`C1'79a
`
`B
`
`D
`
`Figure 2. Prolonged B-cell depletion after anti–CD19-
`CAR-transduced T-cell infusion. (A) Immunohistochem-
`istry staining of a pretreatment bone marrow biopsy
`shows a large population of CD79a⫹ cells. (B) Thirty-six
`weeks after anti–CD19-CAR-transduced T-cell infusion,
`rare CD79a⫹ cells were detected by immunohistochem-
`isty staining of a bone marrow biopsy. The cells did not
`appear to be plasma cells morphologically. The number
`of CD79a⫹ cells was substantially below normal limits.
`the rare CD79a⫹ cells.
`The arrow indicates one of
`(C) The blood B-cell count of the patient treated with
`anti–CD19-CAR-transduced T cells is shown before treat-
`ment and at multiple time points after treatment. B cells
`were measured by flow cytometry for CD19. The dashed
`line indicates the lower limit of normal. Day 0 is the day of
`the second anti–CD19-CAR-transduced T-cell infusion.
`(D) The mean ⫾ SEM blood B-cell count is shown for
`patients who received infusions of T cells targeted to
`either the NY-ESO antigen or the gp100 antigen. The
`patients all received the same chemotherapy and IL-2
`regimen as the patient who received anti–CD19-CAR-
`transduced T cells. NY-ESO and gp100 are not ex-
`pressed by B cells. Day 0 is the day of T-cell infusion. All
`available B-cell counts were included for each time point
`(pretreatment, n ⫽ 28; 4-5 weeks after T-cell
`infusion,
`n ⫽ 29; 8-11 weeks after T-cell
`infusion, n ⫽ 31;
`14-19 weeks after T-cell infusion, n ⫽ 20). All patients
`with available samples had a B-cell count in the normal
`range by 14 to 19 weeks after T-cell infusion. (E) The
`blood CD3⫹ T-cell count of
`the patient
`treated with
`anti–CD19-CAR-transduced T cells is shown before treat-
`ment and at multiple time points after treatment. (F) The
`blood NK cell count of the patient treated with anti–CD19-
`CAR-transduced T cells is shown before treatment and
`at multiple time points after treatment. NK cells were
`measured by flow cytometry as CD3⫺, CD16⫹, CD56⫹
`cells. (E-F) Day 0 is the day of the second anti–CD19-
`CAR-transduced T-cell
`infusion, and the dashed line
`indicates the lower limit of normal. (G) The serum IgG
`level of the patient treated with anti–CD19-CAR-trans-
`duced T cells is shown before treatment and at multiple
`time points after treatment. Day 0 is the day of the second
`anti–CD19-CAR-transduced T-cell
`infusion. (H) Real-
`time polymerase chain reaction was performed with a
`primer and probe set that was specific for the anti-CD19
`CAR. Anti–CD19-CAR-transduced T cells were undetect-
`able in pretreatment blood samples. The anti–CD19 CAR
`transgene was detected in the peripheral blood of the
`patient who received anti–CD19-CAR-transduced T cells
`from 1 to 27 weeks after anti–CD19-CAR-transduced
`T-cell infusion.
`
`C
`
`E
`
`G
`
`Downloaded from http://ashpublications.org/blood/article-pdf/116/20/4099/1489743/zh804610004099.pdf by guest on 12 January 2022
`
`40
`24 32
`16
`8
`0
`Weeks after T cell infusion
`
`3000
`...1 2500
`-¢2000
`~ 1500
`o 1000
`I- 500
`o......,---r--~---..-----r--,
`0
`16
`24
`32
`40
`8
`Weeks after T cell infusion
`
`F
`
`250
`
`~200
`~ 150
`ai
`O 100
`::.::
`Z 50
`01-'---i----r--,--,------,---,
`0
`16
`24
`32
`40
`8
`Weeks after T cell infusion
`
`H
`
`i~ 150
`
`00
`0 c,1
`Cl> C
`co
`~o
`
`1/) -C ,._
`11:1 Cl)
`... 0.
`I-
`
`27
`18
`9
`1 4
`Weeks after T cell Infusion
`
`Weeks after T cell infusion
`
`same chemotherapy and IL-2 regimen as the patient described in this
`report along with T cells retrovirally transduced with receptors that
`recognized either NY-ESO or gp100 did not experience prolonged
`B-cell depletion (Figure 2D).
`Except for B cells and a mild thrombocytopenia, all blood cell
`counts, including neutophils, erythrocytes, T cells, and NK cells, of
`the patient treated with anti–CD19-CAR-transduced T cells recov-
`ered to normal levels by 9 weeks after treatment (Figure 2E-F).
`Thirty-six weeks after anti–CD19-CAR-transduced T cells were
`infused, CD19⫹ cells were absent from the bone marrow as measured
`by flow cytometry (Figure 1F) and immunohistochemistry (Figure
`1H-I). CD79a⫹ cells were undetectable in the bone marrow by
`immunohistochemistry 14 weeks after treatment (data not shown).
`CD79a⫹ cells were detected at greatly below normal frequency 36
`weeks after anti–CD19-CAR-transduced T-cell infusion (Figure 2B).
`CD79a is expressed earlier in B-cell development than CD19,25 so the
`presence of a small number of CD79a⫹ cells while CD19⫹ cells were
`absent suggests early recovery of B-lineage cells.
`A decrease in serum IgG levels occurred after treatment (Figure
`2G). Serum IgM was undetectable from 9 to at least 39 weeks after
`treatment. Serum IgA was 66.8 mg/dL before treatment. Serum IgA
`decreased to below the detectable limit of 10 mg/dL after treatment
`(supplemental Figure 3). Five months after treatment, the patient
`
`developed pneumonia of unknown etiology that required hospital-
`ization. After a course of antibiotics, the patient recovered com-
`pletely. The patient has subsequently received intravenous Ig
`replacement, and he has not had further infections.
`The anti-CD19 CAR transgene was detected in peripheral blood
`mononuclear cells from one to 27 weeks after anti–CD19-CAR-
`transduced T-cell infusion with a quantitative real-time polymerase
`chain reaction assay (Figure 2H).
`This is the first patient treated on our trial and the only patient
`with long enough follow-up to evaluate B-cell depletion. The
`prolonged elimination of CD19⫹ cells in this patient indicates in
`vivo antigen-specific activity of anti–CD19-CAR-expressing T cells.
`Our findings should encourage continued study of anti–CD19-CAR-
`transduced T cells.
`
`Acknowledgments
`
`The authors thank Margaret Brown for flow cytometry, Manuel Van
`Deventer for Ig assays, Laura Devillier for T-cell preparation,
`and Hui Xu, Mary Black, and Zhili Zheng for
`technical
`assistance.
`
`Miltenyi Ex. 1045 Page 3
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/116/20/4099/1489743/zh804610004099.pdf by guest on 12 January 2022
`
`4102
`
`KOCHENDERFER et al
`
`BLOOD, 18 NOVEMBER 2010 䡠 VOLUME 116, NUMBER 20
`
`This work was supported by the Center for Cancer Research,
`National Cancer Institute, National Institutes of Health (intramural
`funding).
`
`Authorship
`
`Contribution: J.N.K. designed the protocol, provided patient care,
`conducted experiments, analyzed data, and wrote the paper;
`W.H.W., J.E.J., D.-A.N.N., and B.J.L. provided patient care,
`
`assisted protocol design, and edited the paper; S.A.F. and R.A.M.
`provided reagents and interpreted data; M.E.D. conducted experi-
`ments and edited the paper; M.S.-S., I.M., and M.R. conducted
`experiments, interpreted data, and edited the paper; and S.A.R.
`designed the protocol, interpreted data, and edited the paper.
`Conflict-of-interest disclosure: The authors declare no compet-
`ing financial interests.
`Correspondence: James N. Kochenderfer, National Institutes of
`Health, 10 Center Dr, CRC Rm 3-3888, Bethesda, MD 20892;
`e-mail: kochendj@mail.nih.gov.
`
`References
`
`3.
`
`1. Eshhar Z, Waks T, Gross G, Schindler DG. Spe-
`cific activation and targeting of cytotoxic lympho-
`cytes through chimeric single chains consisting of
`antibody-binding domains and the gamma or zeta
`subunits of the immunoglobulin and T-cell recep-
`tors. Proc Natl Acad Sci U S A. 1993;90(2):
`720-724.
`2. Kershaw MH, Teng MWL, Smyth MJ, Darcy PK.
`Supernatural T cells: genetic modification of
`T cells for cancer therapy. Nat Rev. 2005;5(12):
`928-940.
`Irving BA, Weiss A. The cytoplasmic domain of
`the T cell receptor zeta chain is sufficient to
`couple to receptor-associated signal transduction
`pathways. Cell. 1991;64(5):891-901.
`4. Sadelain M, Brentjens R, Riviere I. The promise
`and potential pitfalls of chimeric antigen recep-
`tors. Curr Opin Immunol. 2009;21(2):215-223.
`5. Vera JF, Brenner MK, Dotti G. Immunotherapy of
`human cancers using gene modified T lympho-
`cytes. Curr Gene Ther. 2009;9(5):396-408.
`6. Kowolik CM, Topp MS, Gonzalez S, et al. CD28
`costimulation provided through a CD19-specific
`chimeric antigen receptor enhances in vivo per-
`sistence and antitumor efficacy of adoptively
`transferred T cells. Cancer Res. 2006;66(22):
`10995-11004.
`7. Kochenderfer JN, Feldman SA, Zhao Y, et al.
`Construction and preclinical evaluation of an anti-
`CD19 chimeric antigen receptor. J Immunother.
`2009;32(7):689-702.
`8. Lamers CH, Langeveld SC, Groot-van Ruijven CM,
`Debets R, Sleijfer S, Gratama JW. Gene-modified
`T cells for adoptive immunotherapy of renal cell can-
`cer maintain transgene-specific immune functions in
`vivo. Cancer Immunol Immunother. 2007;56(12):
`1875-1883.
`9. Hwu P, Yang JC, Cowherd R, et al. In vivo antitu-
`mor activity of T cells redirected with chimeric an-
`tibody/T-cell receptor genes. Cancer Res. 1995;
`55(15):3369-3373.
`
`10. Brentjens RJ, Latouche JB, Santos E, et al.
`Eradication of systemic B-cell tumors by geneti-
`cally targeted human T lymphocytes co-stimu-
`lated by CD80 and interleukin-15. Nat Med. 2003;
`9(3):279-286.
`11. Till BG, Jensen MC, Wang J, et al. Adoptive im-
`munotherapy for indolent non-Hodgkin lymphoma
`and mantle cell lymphoma using genetically
`modified autologous CD20-specific T cells.
`Blood. 2008;112(6):2261-2271.
`12. Pule MA, Savoldo B, Myers GD, et al.
`Virus-specific T cells engineered to coexpress
`tumor-specific receptors: persistence and antitu-
`mor activity in individuals with neuroblastoma.
`Nat Med. 2008;14(11):1264-1270.
`13. Milone MC, Fish JD, Carpenito C, et al. Chimeric
`receptors containing CD137 signal transduction
`domains mediate enhanced survival of T cells
`and increased antileukemic efficacy in vivo. Mol
`Ther. 2009;17(8):1453-1464.
`14. Brentjens RYR, Bernal Y, Riviere I, Sadelain M.
`Treatment of chronic lymphocytic leukemia with
`genetically targeted autologous T cells: a case
`report of an unforeseen adverse event in a phase
`I clinical trial. Mol Ther. 2010;18(4):666-668.
`15. Jensen MC, Popplewell L, Cooper LJ, et al. Anti-
`transgene rejection responses contribute to at-
`tenuated persistence of adoptively transferred
`CD20/CD19-specific chimeric antigen receptor
`redirected T cells in humans. Biol Blood Marrow
`Transplant. 2010;16(9):1245-1256.
`16. Nadler LM, Anderson KC, Marti G, et al. B4, a
`human B lymphocyte-associated antigen ex-
`pressed on normal, mitogen-activated, and malig-
`nant B lymphocytes. J Immunol. 1983;131(1):
`244-250.
`17. Pontvert-Delucq S, Breton-Gorius J, Schmitt C, et
`al. Characterization and functional analysis of
`adult human bone marrow cell subsets in relation
`to B-lymphoid development. Blood. 1993;82(2):
`417-429.
`
`18. Uckun FM, Jaszcz W, Ambrus JL, et al. Detailed
`studies on expression and function of CD19 sur-
`face determinant by using B43 monoclonal anti-
`body and the clinical potential of anti-CD19 immu-
`notoxins. Blood. 1988;71(1):13-29.
`
`19. Harada H, Kawano MM, Huang N, et al. Pheno-
`typic difference of normal plasma cells from ma-
`ture myeloma cells. Blood. 1993;81(10):2658-
`2663.
`
`20. Johnson LA, Morgan RA, Dudley ME, et al. Gene
`therapy with human and mouse T-cell receptors
`mediates cancer regression and targets normal
`tissues expressing cognate antigen. Blood. 2009;
`114(3):535-546.
`
`21. van Lochem EG, van der Velden VHJ, Wind HK,
`te Marvelde JG, Westerdaal NAC, van Dongen
`JJM. Immunophenotypic differentiation patterns
`of normal hematopoiesis in human bone marrow:
`reference patterns for age-related changes and
`disease-induced shifts. Cytometry B Clin Cytom.
`2004;60(1):1-13.
`
`22. Lucio P, Parreira A, van den Beemd MW, et al.
`Flow cytometric analysis of normal B cell differen-
`tiation: a frame of reference for the detection of
`minimal residual disease in precursor-B-ALL.
`Leukemia. 1999;13(3):419-427.
`
`23. Kawakami Y, Eliyahu S, Delgado CH, et al. Identi-
`fication of a human melanoma antigen recog-
`nized by tumor-infiltrating lymphocytes associ-
`ated with in vivo tumor rejection. Proc Natl Acad
`Sci U S A. 1994;91(14):6458-6462.
`
`24. Chen YT, Scanlan MJ, Sahin U, et al. A testicular
`antigen aberrantly expressed in human cancers
`detected by autologous antibody screening. Proc
`Natl Acad Sci U S A. 1997;94(5):1914-1918.
`
`25. Blom B, Spits H. Development of human lym-
`phoid cells. Annu Rev Immunol. 2006;24:
`287-320.
`
`Miltenyi Ex. 1045 Page 4
`
`

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