throbber

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Miltenyi Ex. 1048 Page 1
`
`

`

`IFFICIAL JOURNAL OF INTERNATIONAL SOCIETY
`OR BIOLOGICAL THERAPY OF CANCER
`
`VOL. 32, NO. 7, Sep'l:ember 2009
`
`IDITOR-IN-CHIEF
`iteven A. Rosenberg, M.D., Ph.D.
`lethesda, Maryland
`
`lSSOCIATE EDITORS
`rlichael B. Atkins, M.D.
`!atrick Hwn, M.D.
`llichael T. Lotze, M.D.
`rancesco Marincola, M.D.
`1UDes J. Mule, Ph.D.
`11cholas P. Restifo, M.D.
`
`iJITORIAL COORDINATOR
`[annah Lee
`~one: 215-253-3551
`ax: 215-220-3450
`nail: joumalofimmunotherapy@gmail.com
`
`iITERNATIONAL SOCIETY FOR BIOLOGICAL
`THERAPY OF CANCER
`1tra Withington, Executive Director
`55 E, Wells Street
`lth Floor
`[ilwaukee, WI 53202-3823
`:I: (414) 271-2456; fax: (414) 276-3349
`nail: info@isbtc.org
`
`ffiLICATION STAFF
`1tvid Myers, Publisher
`1Dathan Pine, Senior Executive Editor
`
`EDITORIAL BOARD
`Michael Atkins
`Richard Barth
`Ernest Borden
`Alfred Chang
`Robert 0. Dillman
`Mary L. Disis
`Janice P. Dutcher
`Alexander Eggermont
`Soldano Ferrone
`Bernard A. Fox
`Michael S. Gordon
`Ronald B. Herberman
`Peter Hersey
`Yutaka Kawakami
`Ulrich Keilholz
`Steven K. Libutti
`Albert F. LoBuglio
`Kim A. Margolin
`James Lee Murray
`Elizabeth A. Repasky
`Licia Rivoltini
`Pedro Romero
`Michael Salgaller
`Noriyuki Sato
`Carmen Scheibenbogen
`Craig L. Slingluff, Jr.
`Paul M. Sondel
`Mario Sznol
`WalterUrba
`Louis Weiner
`Christine A. White
`Jon M. Wigginton
`
`· advertising inquiries, please contact Sherry Reed at sherry.reed@wolterskluwer.com, 4I0-528:8553.
`
`Miltenyi Ex. 1048 Page 2
`
`

`

`Journal of lmmunotherapy
`
`Volume 32
`
`Number 7
`
`September 2009
`
`Contents
`
`Basic Studies
`
`677
`
`689
`
`703
`
`713
`
`726
`
`737
`
`High-a,idity Autoreactive CD4+ T Cells Induce Host CTL, Overcome Tregs
`and Mediate Tumor Destruction
`Andrew G. Brandmaier, Wolfgang W: Leitner, Sung P. Ha, John Sidney, Nicholas P. Restifo,
`and Christopher E. Touloukian
`
`Construction and Preclinical Evaluation of an Anti-CD19 Chimeric Antigen Receptor
`James N. Kochenderfer, Steven A. Feldman, Yangbing Zhao, Hui Xu, Mary A. Black,
`Richard A. Morgan, Wyndham IL Wilson, and Steven A. Rosenberg
`
`Effect of Yeast-derived [3-glucan in Conjunction With Bevacizumab for the Treatment
`of Human Lung Adenocarcinoma in Subcutaneous and Orthotopic Xenograft Models
`Wangjian Zhong, Richard Hansen, Bing Li, Yihua Cai, Carolin.a Salvador, Grace D. Moore,
`and Jun Yan
`
`Inhibition of Tumor Growth by Targeted Toxins in Mice is Dramatically Improved by
`Saponinum Album in a Synergistic Way
`Christopher Bachran, Horst Diirkop, Mark Sutherland, Diana Bachran, Christian Muller,
`Alexander Weng, Matthias F. Melzig, and Hendrik Fuchs
`
`Genetic Modification of T Cells With IL-21 Enhances Antigen Presentation
`and Generation of Central Memory Tumor-specific Cytotoxic T-lymphocytes
`Anjum S Kaka, Donald R. Shaffer, Ryan Hartmeier, Ann Af. Leen, An Lu, Adham Bear,
`Cliona M Rooney, ';Ind Aaron E. Foster
`
`Activated T-cell-mediated Immunotherapy With a Chimeric Receptor Against CD38 in
`B-cell Non-Hodgkin Lymphoma
`Keichiro Mihara, Kazuyoshi Yanagihara, Misato Takigahira, Chihaya Imai, Akira Kitanaka,
`Yoshihiro Takihara, and Akiro Kimura
`
`( continued next page)
`
`r!PPincott
`tyilliams & Wilkins
`Nolters Kluwer
`Health
`
`Journal of lmmunotherapy (ISSN: #1524-9557) is published nine times a year in January, February, April, May, June, July,
`September, October, and November by Lippincott Williams & Wilkins, 16522 Hunters Green Parkway, Hagerstown, MD 21740-
`2116. Business and production offices are located at 530 Walnut Street, Philadelphia, PA 19106-3621. Periodicals postage paid at
`Hagerstown, MD and at additional mailing offices. Copyright (Q 2009 by Lippincott Williams & Wilkins.
`Annual Subscription Rates: United States- $600 Individual, $1585 Institution, $282 In-training. Rest of World-$700 Individual,
`$1867 Institution. $296 In-training.
`copy rate $206. All prices include a handling charge. Subscriptions outside of North
`America must add $14 for airfreight
`United States residents of AL, CO, DC, FL. GA, HI, IA, ID, IN, KS, KY, LA, MD,
`MO, ND, NM, NV, PR, RI, SC, SD, UT, VT, WA, V./V add state sales tax. The GST tax of7% must be added to all orders shipped to
`Canada (Lippincott Williams & Wilkins' GST Identification #895524239, Publications Mail Agreement #616346. Subscription
`prices outside the United States must be prepaid. Prices subject to change without notice. Visit us online at www.lww.com.
`Website: www.irnmunotherapy-journal.com
`Postmaster: Send address changes to Journal of Immunotherapy, P.O. Box 1550, Hagerstown, MD 21740.
`
`Miltenyi Ex. 1048 Page 3
`
`

`

`Journal of lmmunotherapy • Volume 32, Number 7, September 2009
`
`Contents (continued)
`
`744
`
`A Novel Mouse Model for Evaluation and Prediction of HLA-A2-restricted
`CEA Cancer Vaccine Responses
`Antonella Conforti, Daniela Peruzzi, Patrizia Giannetti, Antonella Biondo, Gennaro Ciliberto,
`Nicola La Monica, and Luigi Aurisicchio
`
`Clinical Studies
`
`755
`
`765
`
`HSCT Recipients Have Specific Tolerance to MSC but not to the MSC Donor
`Mikael Sundin, A. John Barrett, Olle Ringden, Mehmet Uzunel, Helena Lonnies,
`Asa-Lena Dack/and, Birger Christensson, and Katarina Le Blanc
`
`Vaccination of Renal Cell Cancer Patients With Modified Vaccinia Ankara Delivering
`the Tumor Antigen 5T4 (TroVax) Alone or Administered in Combination With
`Interferon-ix (IFN-a): A Phase 2 Trial
`Robert J. Amato, William Shingler, Madusha Goonewardena, Jackie de Belin, Stuart Naylor,
`Jaros/aw Jae, James Willis, Somyata Saxena, Joan Hernandez-McClain, and Richard Harrop
`
`This journal is listed in Index Medicus/MEDLINE Current Contents/Life Sciences Scisearch,
`Biomedical Database, BJOSIS, EMBASE/Excerpta Medica, Chemical Abstracts, and Current Awamess in Biological Sciences.
`
`Address for non-member subscription information, orders, or change of address: Lippincott Williams & Wilkins, P.O. Box 1580, Hagerstown, MD 21741-1
`phone 800-638-3030 (outside the United States 301-223-2300); fax 301-223-2400. ln Japan, contact LWW Igaku-Shoin Ltd., 3-23-14 Hongo, Bunkyo-ku, T,
`113-0033; phone 81-3-5689-5400; fax 81-3-5689-5402. In Bru,)gladesh, India, Nepal, Sri Lanka, and Pakistan, contact Globe Publications Pvt. B-13 3rd F
`A Block, Shopping Complex, Naraina V!har, Ring Road,ffiw Delhi, 1 !0028; phone 91-11-579-3211; fax 91-11-579-8876.
`-~✓
`
`Lippincott Williams & Wilkins cannot beJielore;~onsible for errors or for any consequences arising from the use of the information contained in this journal.
`appearance of advertising in this joµmaf does not constitute an endorsement or approval by Lippincott Williams & Wilkins for the quality or value of the pre
`advertised or of the claims)yade for it by its manufacturer.
`
`PERMISSION TO PHOTOCOPY ARTICLES: This publication is protected by copyright. Permission to photocopy must be secured in writing f
`Permission Dept, Lippincott Williams & Wilkins, 351 West Camden Street, Baltimore, MD 21201, FAX: 410-528-8550; or Copyright Clearance Center (C
`222 Rosewood Dr., Danvers, MA 01923; FAX: 508-750-4470; or UMI, Box 49, 300 North Zeeb Road, Ann Arbor, MI 48106-1346; FAX: 313-761-1203.
`
`Article and issue photocopies and 16-mm microfilm, 35-mm microfilm, and 105-mm microfiche are available from UMI, 300 North Zeeb F
`Ann Arbor, MI 48106-1346.
`
`Miltenyi Ex. 1048 Page 4
`
`

`

`BASIC STUDY
`
`Construction ~nd Preclinical Evaluation of an Anti-CD19
`Chimeric Antigen Receptor
`
`James N. Kochenderfer,* Steven A. Feldman,* Yangbing Zhao,* Hui Xu,* Mary A. Black,*
`Richard A. Morgan,* Wyndham H. Wilson,t and Steven A. Rosenberg*
`
`Summary: T cells can be engineered to express the genes of
`chimeric antigen
`receptors
`(CARs)
`that
`recognize
`tumor(cid:173)
`associated antigens. We constructed and compared 2 CARs that
`contained a single chain variable region moiety that recognized
`CD19. One CAR contained the signaling moiety of the 4-IBB
`molecule and the other did not. We selected the CAR that did not
`contain the 4-IBB moiety for further preclinical development. We
`demonstrated that gammaretroviruses encoding this receptor could
`transduce human T cells. Anti-CD19-CAR-transduced cos+ and
`CD4+ T cells produced interferon-y and interleukin-2 specifically
`in response to CD19+ target cells. The transduced T cells
`specifically killed primary chronic lymphocytic leukemia (CLL}
`cells. We transduced T cells from CLL patients that had been
`previously treated with chemotherapy. We induced these T cells to
`proliferate sufficiently to provide enough cells for clinical adoptive
`T cell transfer with a protocol consisting of an initial stimulation
`with an anti-CD3 monoclonal antibody (OKT3) before trans(cid:173)
`duction followed by a second OKT3 stimulation 7 days after
`transduction. This protocol was successfully adapted for use in
`CLL patients with high peripheral blood leukemia cell counts by
`depleting CD19+ cells before the initial OKT3 stimulation. In
`preparation for a clinical trial that will enroll patients with
`advanced B cell malignancies, we generated a producer cell clone
`that produces retroviruses encoding the anti-CD19 CAR, and we
`produced sufficient retroviral supernatant for the proposed clinical
`trial under good manufacturing practice conditions.
`
`Key Words: chimeric antigen receptor, gene therapy, CDI9, T cell,
`gammaretrovirus, adoptive T cell therapy
`(J lmmunother 2009;32:689-702)
`
`Approximately 22,000 people die because of B cell
`
`malignancies each year in the United States. 1 Patients
`with some B cell malignancies
`including mantle cell
`lymphoma and chronic lympocytic leukemia (CLL) cannot
`be cured by therapies such as conventional chemotherapy
`and monoclonal antibodies,2•3 but some patients with these
`diseases can achieve prolonged disease-free survival after
`allogeneic stem cell
`transplantation.4-<i Unfortunately,
`
`Received for publication January 6, 2009; accepted April 21, 2009.
`From the •Surgery Branch of the National Cancer Institute; and
`tMetabolism Branch of the National Cancer Institute, National
`Institutes of Health, Bethesda, MD.
`This work was supported by intramural funding of the Center for
`Cancer Research, National Cancer Institute, NIH.
`Financial Disclosure: All authors have declared there are no financial
`conflicts of interest in regards to this work.
`Reprints: James N. Kochenderfer, Surgery Branch of the National
`Cancer Institute, NIH 10 Center Drive CRC Room 3-3888
`.Bethesda, MD 20892 (e-mail: kochendj@mail.nih.gov).
`Copyright © 2009 by Lippincott Williams & Wilkins
`
`allogeneic stem cell transplantation is limited by significant
`transplant-related mortality and a shortage of suitable
`donors. 2•6•7 In patients with B cell malignancies that relapse
`after allogeneic stem cell transplantation, infusion of
`allogeneic donor lymphocytes can induce remissions. 8- 10
`The effectiveness of these lymphocyte infusions provides a
`rationale for attempts to develop other cellular immuno(cid:173)
`therapies for B cell malignancies.
`Adoptive transfer of autologous T cells that are
`cultured from tumor infiltrating lympohocytes can cause
`regressions of advanced melanoma in humans. 11 •12 Because
`tumor-reactive T cells cannot be reliably cultured from
`most of human tumors, methods have been developed to
`engineer T cells to express genes encoding tumor antigen(cid:173)
`specific T cell receptors. 13•14 Adoptive transfer of these
`genetically modified T cells is a promising approach to
`cancer immunotherapy. 15 Another approach to adoptive
`T cell therapy is to engineer T cells to express chimeric
`antigen receptors (CARs). 16•17 CARs are made up of an
`antigen-recognizing receptor coupled to signaling molecules
`that can activate T cells expressing the CAR. 18- 20 The
`antigen-receptors most commonly incorporated into CARs
`are single chain variable region moieties (scFv) that consist
`of the light chain and heavy chain variable regions
`of a monoclonal antibody joined by a peptide linker.
`Murine models have shown that T cells transduced with
`retroviruses encoding CARs can protect mice from tumor
`challenges in vivo. 21 •22
`Our group has completed a phase I clinical trial in
`which patients with ovarian carcinoma were treated with
`T cells that were transduced with a CAR that was
`specific for
`the ovarian carcinoma-associated antigen
`ct-folate receptor. 23 No objective tumor regressions were
`seen. 23 The CAR used in this clinical trial incorporated the
`Fe receptor-y cytoplasmic signaling chain without any
`costimulatory molecules such as CD28 or 4-IBB. More
`recent work in mice has demonstrated that CARs contain(cid:173)
`ing the T cell receptor (TCR)-1; cytoplasmic signaling chain
`had superior in vitro function and in vivo antitumor efficacy
`than CARs containing the Fe receptor-y cytoplasmic
`signaling chain. 24 In addition, in vitro studies with human
`cells and murine
`in vivo studies have shown
`that
`incorporating the signaling domain of CD28 into CARs
`enhances function and in vivo antitumor efficacy. 22•25-27
`Signaling of the 4-1 BB costimulatory molecule has been
`shown to enhance T cell proliferation and persistence, 28•29
`and 4- I BB signaling enhanced the function of CARs in
`vitro. 30,31 Thus, significant advances in CAR design have
`occurred since our last clinical trial using CAR-transduced
`T cells.
`CD I 9 is a promising target for antigen-specific T cell
`therapies because CDl9 is expressed on most malignant
`
`J lmmunother • Volume 32, Number 7, September 2009
`
`www.immunotherapy-journal.com I 689
`
`Miltenyi Ex. 1048 Page 5
`
`

`

`Kochenderfer et al
`
`B cells, 32•33 and the only normal cells that express CD 19 are
`B cells and perhaps follicular dendritic cells. 33•34 Impor(cid:173)
`tantly, CD19 is not expressed on pluripotent hematopoietic
`stem cells. 35 Although destruction of normal B cells is a
`drawback to targeting CD19, destruction of normal B cells
`is well tolerated. In most patients that receive the widely
`the
`used anti-CD20 monoclonal antibody rituximab,
`number of normal peripheral blood B cells is severely
`depressed for several months, 36 yet patients that receive
`chemotherapy plus rituximab do not have an increased rate
`of infections when compared with patients that receive
`chemotherapy alone. 37 Finally, patients can be treated with
`intravenous infusions of IgG if necessary to increase IgG
`levels. 38
`We constructed 2 CARs that target CD19 and selected
`the one with the best in vitro function for further testing in
`preparation for a clinical trial. T cells that were transduced
`with gammaretroviruses encoding this CAR recognized
`in an antigen-specific manner
`CD 19-expressing cells
`and killed primary B cell CLL cells. We have optimized
`methods of T cell culture and transduction to generate
`highly active anti-CD19 CAR-expressing T cells from the
`blood of patients with CLL.
`
`MATERIALS AND METHODS
`Construction of the MSGV-FMC63-28Z and
`MSGV-FMC63-CD828BBZ Recombinant
`Retroviral Vectors
`The mouse stem cell virus-based splice-gag vector
`(MSGV)-FMC63-2SZ recombinant retroviral vector encodes
`the MSGV retroviral backbone and the FMC63-1SZ CAR.
`The FMC63-2SZ CAR consists of an anti-CD19 scFv that
`wa~ derived from the FMC63 mouse hybridoma, 39 a
`portion of the human CD2S ·molecule, and the intracellular
`component of the human TCR-~ molecule. The exact
`sequence of the CD2S molecule included in the FMC63-2SZ
`CAR corresponds to Genbank identifier NM_006139. The
`sequence includes all amino acids starting with the amino
`acid sequence IEVMYPPPY and continuing all the way
`to the carboxy-terminus of the protein. To encode the
`anti-CD19 scFv component of the vector, we designed a
`DNA sequence which was based on a portion of a
`previously published CAR. 40 This sequence encodes the
`following components in frame from the 5' end to the
`3' end: an XhoI site, the human granulocyte-macrophage
`colony-stimulating factor receptor ~-chain signal sequence,
`the FMC63 light chain variable region, 39 a linker peptide, 40
`the FMC63 heavy chain variable region, 39 and a Notl site.
`This sequence was synthesized by GeneArt AG (Regensburg,
`Germany), and a plasmid encoding this sequence was
`digested with XhoI and Notl (New England Biolabs). To
`form the MSGV-FMC63-2SZ retroviral vector, the XhoI
`and Natl-digested fragment encoding the FMC63 scFv was
`ligated into a second XhoI and Natl-digested fragment that
`encoded the MSGV retroviral backbone 14 as well as part of
`the extracellular portion of human CD2S, the entire
`transmembrane and cytoplasmic portion of human CD2S,
`the cytoplasmic portion of the human TCR-~
`and
`molecule. 41
`We also designed a second recombinant retroviral
`vector called MSGV-FMC63-CDS2SBBZ that consists of
`the following components in order from 5' to 3': the MSGV
`retroviral backbone, the FMC63 scFv, the hinge and
`
`690 I www.immunotherapy-journal.com
`
`J lmmunother • Volume 32, Number 7, September 2009
`
`transmembrane regions of the CDS molecule, the cytoplasmic
`the cytoplasmic
`portions of CD2S and 4- lBB, and
`component of the TCR-~ molecule. MSGV-FMC63-
`CDS2SBBZ was constructed using a multistep strategy.
`A fragment encoding the CDS, CD2S, 4-lBB, and TCR-~
`components was generated using an overlapping polymerase
`chain reaction (PCR) method42 using the following primers:
`1. Notl-CDSF: 5' -acgGCGGCCGCA ttcgtgccggtcttcctgc-3';
`2. 2Scyto-CDSR: 5'- gcctgctcctcttactcctgttcctgtggttgcagta
`aag-3';
`3. CDS-2ScytoF: 5'- ctttactgcaaccacaggaacaggagtaagaggag
`caggc-3';
`4. BamHI-zetaR: 5'- ttat GGATCC ttagcgagggggcagggcc-3'.
`The Notl-CDSF and 2Scyto-CDSR oligonucleotide
`primers were used to generate a PCR product that encoded
`the hinge and transmembrane region of CDS by using human
`CDS~ cDNA as a template. An overlapping fragment
`encoding the cytoplasmic portions of CD2S, 4-1 BB, and
`TCR-~ components in the order of CD2S-4-1BB-CD3~
`was generated using forward primer CDS-2ScytoF and
`the BamHI-zetaR reverse primer. The 2 PCR products were
`combined and the full-length construct generated using
`the Notl-CDSF and the BamHI-zetaR primers. DNA
`encoding this full-length construct and DNA encoding the
`into the MSGV retroviral
`FMC63 scFv were ligated
`backbone to form the MSGV-FMC63-CDS2SBBZ retro(cid:173)
`viral vector.
`the SP6 scFv43 was kindly
`A plasmid encoding
`provided by Z. Eshhar. This scFv was cloned into the
`MSGV retroviral vector along with a portion of the CD2S
`gene and the gene for cytoplasmic portion of the CD3~
`the MSGV-SP6-2SZ plasmid. This
`to form
`molecule
`plasmid encoded a receptor that is referred to as SP6-2SZ
`in this paper. The SP6-2SZ receptor recognizes the hapten
`2, 4, 6-trinitrobertzenesulfonic acid and served as a negative

`control.
`
`Culture Media
`T cells were always cultured in T cell medium
`which consisted of AIM V medium plus 5% AB (blood
`type AB) serum (Gemini Bioproducts, Woodland, CA),
`100 U /mL penicillin, 100-µg/mL streptomycin, and 1.25 µg/
`mL amphotericin B. Rl0 medium consisted of Roswell
`Park Memorial Institute 1640 medium plus 10% fetal
`bovine serum (FBS), 100 U/mL penicillin, 100 µg/mL
`streptomycin and 2 mM L-glutamine. D 10 medium con(cid:173)
`sisted of Dubecco Modified Eagle Medium, 10% FBS,
`100 U /mL penicillin, 100 µg/mL streptomycin, nonessential
`amino acids, and 2 mM L-glutamine. SupB I 5 medium
`consisted of Iscove Modified Dulbecco's Medium plus
`20% FBS, 100 U /mL penicillin, 100 µg/mL streptomycin,
`2 mM L-glutamine, and 0.055 mM 2-mercaptoethanol.
`Cytotoxicity medium was phenol red-free Roswell Park
`Memorial Institute medium plus 5% AB (blood type AB)
`serum (Gemini Bioproducts), 100 U/mL penicillin, and
`100 µg/mL streptomycin. All cell culture media ingredients
`except the AB (blood type AB) serum were from Invitrogen
`(Carlsbad, CA). Recombinant human interleukin (IL)-2
`was obtained from Chin;m (Emeryville, CA).
`Transient Retrovirus Production
`To transiently produce retroviruses, 293GP packaging
`cells44 were transfected with either the MSGV-FMC63-2SZ
`plasmid, the MSGV-FMC63-CDS2SBBZ plasmid, or the
`MSGV-SP6-2SZ plas~id along with a plasmid encoding
`
`© 2009 Lippincott Williams & Wilkins
`
`Miltenyi Ex. 1048 Page 6
`
`

`

`J lmmunother • Volume 32, Number 7, September 2009
`
`Construction and Evaluation of an Anti-CD/ 9 CAR
`
`the RD 114 envelope protein45 using Lipofectamine 2000
`(lnvitrogen). The transfected cells were incubated at 37°C
`for 6 to 8 hours in DI0 medium without antibiotics. The
`medium used for transfection was then replaced with fresh
`D10 medium and the cells were incubated for another
`36 to 48 hours. During and after transfection, the 293GP
`cells were cultured on poly-n-lysine coated dishes (BD
`Biosciences, San Jose, CA). Supernatant containing retro(cid:173)
`viruses was removed from the dishes and centrifuged to
`remove cellular debris. The supernatant was snap frozen
`on dry ice and stored at - 80°C. Transiently produced
`retroviruses were us~d in the experiments described in
`
`Figures l and 2 and in Table 1. In addition, all retroviruses
`encoding the SP6-28Z CAR were produced transiently.
`
`Generation of the MSGV-FMC63-28Z PG 13
`Producer Cell Clone, H3
`PG 13 packaging cell clones were generated using
`the PG I 3 gibbon ape leukemia virus packaging cell line
`(A TCC, Manassass, VA), and
`the human ecotropic
`packaging cell line, Phoenix Ecotropics (kindly provided
`by Gary Nolan, Stanford University, Stanford, CA). All
`cells were cultured in D 10 medium without antibiotics.
`Cells were maintained at 37°C and 5% CO2•
`
`A
`
`8
`
`C
`
`D
`
`E
`
`FMC63 single chain Fv
`
`CD28 TCR-1;
`
`Isotype
`
`0.01
`
`Anti-Fab
`
`0.10
`
`...
`
`CDJ
`Isotype
`
`o.oo
`
`o.o
`
`0.22
`
`(DJ
`Anti-Fab
`
`19.3
`
`CDJ
`
`o.oo
`
`CDJ
`Anti-Fab
`
`0.2
`
`0.03
`
`II
`
`C. a J!!
`
`3
`
`(DJ
`CDJ
`FIGURE 1. A comparison of the design and transduction efficiency of 2 anti-CD19 chimeric antigen receptors. A, A diagram of the
`recombinant retroviral vector MSGV-FMC63-28Z is shown (LTR, long terminal repeat; Fv, variable regions; CD28, part of the
`extracellular region and all of the transmembrane and intracellular regions of CD28; TCR-1;, the entire cytoplasmic region of the TCR-~
`molecule). B, A diagram of the recombinant retroviral vector MSGV-FMC63-CD828BBZ is shown (LTR, long terminal repeat; Fv, variable
`regions; CDS, CDS hinge region; CD28, CD28 cytoplasmic region; 4-1 BB, 4-1 BB cytoplasmic region; TCR-~ the cytoplasmic region of
`the TCR-~ molecule. C, PBMC were started in culture with OKT3 and IL-2 on day 0. The cells were transduced with retroviruses encoding
`FMC63-28Z on days 2 and 3. On day 8, expression of FMC63-28Z was detected on 45% of T cells when the cells were stained with anti(cid:173)
`Fab antibodies and anti-CD 3. Staining with isotype-matched control antibodies and anti-CD3 is also shown. D, A PBMC culture from the
`same donor as in (C) was initiated on day 0. Cells were transduced with retroviruses encoding FMC63-CD828BBZ on days 2 and 3, and
`stained with either anti-Fab antibodies or isotype-matched control antibodies on day 8 in the same manner as in (C). Expression of
`FMC63-CD828BBZ was detected on 19% of T cells. E, A PBMC culture from the same donor as in (C) and (D) was initiated on day 0. The
`cells were not transduced. On day 8 the cells were stained with either anti-Fab or isotype-matched control antibodies in the same
`manner as in (C) and (D). This.experiment was performed using cells from the same cultures that were tested in the experiments
`described in Table 1 and Figure 2. The results presented in (C), (D), and (E) are representative of 6 experiments that used cells from 6
`different donors. MSGV indicates mouse stem cell virus-based splice-gag vector; PBMC, Peripheral blood mononuclear cell; TCR, T cell
`receptor.
`
`© 2009 Lippincott Williams & Wilkins
`
`www.immunotherapy-journal.com I 691
`
`l
`
`Miltenyi Ex. 1048 Page 7
`
`

`

`Kochenderfer et al
`
`J lmmunother • Volume 32, Number 7, September 2009
`
`A
`
`FMC63-28Z-transduced
`NGFR-K562
`
`CD19-K562
`
`5.73
`
`48.45
`
`U.18
`
`FMC63-CD828BBZ-transduced
`NGFR-K562
`CD19-K562
`
`3.37
`
`3.01
`
`19.61
`
`U.UB
`
`0.2
`
`CDS
`
`CDS
`
`B
`
`FMC63-28Z-transduced
`NGFR-K562
`
`CD19-K562
`
`FMC63-CD828BBZ-transduced
`NGFR-K562
`CD19-K562
`
`U.86
`
`2.
`
`a.11
`
`U.1
`
`1
`
`N
`
`~
`
`..
`J
`.....
`••,: :•• .:
`
`.
`~ :-·
`:!! ~•1.·
`
`•
`
`. I
`
`•
`
`FIGURE 2. A comparison of cytokine production by T cells transduced with different anti-CDl 9 chimeric antigen receptors. Peripheral
`blood mononuclear cell cultures were initiated on day O and transduced on days 2 and 3 after culture initiation. On day 11 after the
`cultures were initiated, cells that were transduced with either FMC63-28Z or FMC63-CD828BBZ were stimulated with either
`CDl 9-K562 cells or NGFR-K562 cells for 5 hours and intracellular staining for IFNy (A) and IL-2 (B) was performed. The transduced
`T cells produced IFNy and IL-2 in a CDl 9-specific manner. The plots are gated on CD3+ lymphocytes, and the percentage of cells in each
`quadrant is shown on the plots. This experiment was performed using cells from the same cultures that were tested in the experiments
`described in Figur_e 1 and Table 1. This experiment is representative of 6 experiments that used cells from 6 different donors.
`added to each well followed by centrifugation (2000 x g) at
`32°C for 3 hours. After centrifugation, supernatant was
`removed and 5 x 105 PG13 cells were added to each well,
`and the plates were centrifuged (1000 g) for 10 minutes at
`32°C. The transduction was repeated the next day, and then
`PG 13 producer cell clones were generated by limiting
`dilution cloning. Owing to lack of a selectable marker, high
`titer clones were identified by RNA dot blot as described
`previously. 14,46 Retroviral supernatant from the 6 highest
`titer clones was generated. Briefly, 175 cm2 tissue culture
`
`A PG 13 retroviral producer cell clone was generated as
`described previously 14 with the following changes. Phoenix
`Ecotropic cells were transfected with 9 µg of plasmid DNA
`(MSGV-FMC63-28) using the Lipofectamine 2000 trans(cid:173)
`fection reagent (Invitrogen). After 48 hour, supernatant
`was harvested and used to transduce retroviral packaging
`cell line, PGl3. Nontissue culture treated 6-well plates were
`coated with 20 µg/mL recombinant fibronectin fragment
`(RetroNectin) as described by the manufacturer (Takara,
`Madison, WI). Retroviral vector supernatant (4mL) was
`
`TABLE 1. Interferon y Enzyme Linked lmmunosorbent Assay Comparing FMC63-28Z and FMC63-CD828BBZ
`CD19-Targets
`CD19+Targets
`A549
`SupB15
`
`bv173
`
`CLL
`
`MDA231
`
`CCRF-CEM Effectors Alone
`
`Effector cells
`7
`10
`30
`2970
`301
`6150
`17450
`FMC63-28Z
`7
`20
`7
`640
`15
`2700
`13500
`FMC63-CD828BBZ
`8
`15
`6
`16
`57
`17
`118
`Nontransduced
`100,000 effector cells were cultured overnight with 100,000 target cells, and an interferon-y ELISA was performed. Effector cells were T cells that were
`transduced with the FMC63-28Z CAR, T cells that were transduced with the FMC63-CD828BBZ CAR, or nontransduced T cells from the same donor that
`were cultured in the same manner. This assay was performed on day 8 after initiation of cultures. Transductions were performed on day 2 and day 3 after
`culture initiation. All values are pg/mL of IFN-y (mean of duplicate wells). CLL refers to primary CLL cells. These data are representative of the results
`obtained in 2 separate experiments with cells from 2 different donors. The cells used in this experiment were from the same cultures used in the experiments
`reported in Figures 1 and 2.
`Bold values indicate specific recognition of CD 19-expressing target cells.
`
`692 I www.immunotherapy-journal.com
`
`© 2009 Lippincott Williams & Wilkins
`
`Miltenyi Ex. 1048 Page 8
`
`

`

`J lmmunother • Volume 32, Number 7, September 2009
`
`Construction and Evaluation of an Anti-CD] 9 CAR
`
`flasks (Nunc, Cole-Parmer, Vernon Hills, IL) were seeded
`at 4 x 104 cells/cm2. A medium exchange (30 mL) was
`performed on day 3. Supernatant was harvested 24-hour
`later and aliquoted. The supernatant was stored at - 80°C
`until further use. Supernatant from each clone was
`evaluated by transducing T cells as described below
`under "Retroviral Transductions" and measuring CAR
`expression on the surface of transduced T cells as described
`below under "CAR Detection on Transduced T cells". The
`ability of the transduced T cells to produce interferony
`(IFNy) in a CD19-specific manner was measured in an
`enzyme-linked immunosorbent assay (ELISA). One clone,
`which was designated H3, was selected for production of a
`master cell bank and subsequent production of retroviral
`supernatant under Good Manufacturing Practice (GMP)
`conditions.
`
`MSGV-FMC63-28Z Retrovirus Production Using
`PG13 Producer Cell Clone H3 Under GMP
`Conditions
`A total of 26 1700 cm2 expanded surface roller bottles
`were seeded on day 0 at a cell density of 4 x 104 clone
`H3 cells/cm2 in 200mL of Dl0 medium. On day 3, the
`medium was exchanged and replaced with 120mL DlO
`medium. Medium containing the retroviral vector was
`harvested daily with bottles being refed with 120 mL of
`medium. Glucose levels were monitored daily using Roche's
`Accu-check system (Roche, Basal, Switzerland). If glucose
`levels dropped below 2 g/L, the volume of the medium
`exchange was doubled to 240mL/roller bottle for all
`subsequent harvests. All harvests were aliquoted and stored
`at - 80°C until further use. All clinical products were
`subjected to an extensive biosafety testing program m
`accordance with current regulatory guidelines. 47- 50
`
`Patient Samples and Cell Lines
`Nonleukemic peripheral blood mononuclear cell
`(PBMC) samples were obtained from melanoma patients
`that were enrolled on institutional review board-approved
`protocols in the Surgery Branch of the National Cancer
`Institute. Leukemic PBMC were obtained from patients
`with CLL that were enrolled on National Cancer Institute
`protocol number 1997-C-0! 78. PBMC were cryopreserved
`in 90% FBS plus JO¾ dimethyl sulfoxide (Sigma, St Louis,
`MO). The following CD19-expressing immortalized cell
`lines were used: bv 173 ( chronic myeloid leukemia in
`lymphoid blast crisis, a kind gift of Dr A. Wiestner
`National Heart Lung and Blood Institute, Bethesda,
`MD), NALM-6 (acute lymphoid leukemia from DSMZ,
`Braunschweig, Germany), SupB15
`[acute
`lymphoid
`leukemia from American Type Culture Collection (A TCC),
`Manassass, VA], and Toledo (B cell diffuse large cell
`lymphoma from A TCC). The following CD 19 - cell lines
`were used: A549 (lung carcinoma, from ATCC), CCRF(cid:173)
`CEM (T cell leukemia from A TCC), K562 ( chronic myeloid
`leukemia from A TCC), MDA23 l (breast carcinoma from
`ATCC), TC71 (Ewing sarcoma, a kind gift of Dr M. Tsokos,
`National Cancer Institute, Bethesda, MD), and 624
`(melanoma, derived in Surgery Branch, National Cancer
`Institute). All cell lines were maintained in RIO medium
`except for SupB15. SupB15 was cultured in SupB15
`medium. When CLL PBMC were used as targets in assays,
`the cells were cultured in RIO medium for 12 to 18 hours
`before the assay.
`
`T Cell Culture
`PBMC were thawed and washed once in T cell
`medium. PBMC were suspended at a concentration of
`I x I 06 cell/mL in T cell medium containing 50 ng/mL
`of the anti-CD3 monoclonal antibody OKT3 (Ortho,
`Bridgewater, NJ) and 300 IU /mL of IL-2. Twenty micro(cid:173)
`liters of this suspension were added to 75 cm2 culture flasks
`(Corning, Corning, NY). The flasks were cultured upright
`at 37°C and 5% CO2• This method of T cell stimulation in
`which OKT3 was added directly to media is referred to as
`solubilized OKT3 and was used for the initial stimulation
`of PBMC in all experiments repor

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