throbber
11SOU HUMVEE OLE
`
`Human gene therapy.
`23, no. 5 (2012
`RB155
`
`May)Hl ,
`
`Cl
`
`
`VI OEee
`www.liebertpub.com/hum
`
` i
`
`
`
`Miltenyi Ex. 1026 Page 1
`
`

`

`
`
`Human Gene Therapy
`
`European Society of Gene and Cell Therapy * British Society for Gene Therapy * French Society of Cell and Gene Therapy
`Irish Society for Gene and Cell Therapy * The Israeli Society of Gene & Cell Tharapy * German Society for Gene Therapy
`Spanish Society of Gene and Cell Therapy * Netherlands Society of Gene and Cell Therapy * Finnish Society of Gene Therapy
`Human Gene Therapyis the premier, multidisciplinary journal covering all aspects of gene therapy. The
`Journal publishes in-depth coverage of DNA, RNA,andcell therapies by delivering the latest break-
`throughsin research and technologies. Human Gene Therapyis a central forum forscientific and clinical
`information, including ethical, legal, regulatory, social, and commercial issues, which enables the ad-
`vancementandprogressof therapeutic procedures leading to improvedpatient outcomes,andultimately,
`to curing diseases.
`Information for Manuscript Submission: www.liebertpub.com/hum
`Submit manuscripts online: http://mc.manuscriptcentral.com/hum
`All papers, news, comments, opinions, findings, conclusions, or recommendations in Human Gene
`Therapy are those of the authors and do not constitute those of the Journal, its Publisher,its editorial staff,
`orits affiliated societies.
`Reprints: For permission to photocopy 99 copies or fewer of an article for internal purposes please
`request permission andpay the appropriate fee by contacting the Copyright Clearance Center,Inc.: (978)
`750-8400. If the number of copies is 100 or greater please contact the Publisher at (914) 740-2100 or
`reprints@liebertpub.com
`Advertising inquiries, with the exception of those coming from Europe and Australia, should be ad-
`dressed to the Publisher: (914) 740-2100; E-mail: advertising@liebertpub.com For advertising inquiries
`from Europe and Australia: +44-0-1875-825700; E-mail: impress@impressmedia.com All advertise-
`ments are subject to approval by the Publisher. The acceptance of advertisements doesnot constitute an
`endorsement of the product or service advertised.
`Indexed in: MEDLINE; Current Contents”/Life Sciences; Science Citation Index Expanded; Science
`Citation Index”; Biotechnology Citation Index”; Biological Abstracts; BIOSIS Previews; Journal Ci-
`tation Reports/Science Edition; Derwent Drug File; EMBASE/Excerpta Medica, EMBiology; Scopus;
`and Chemical Abstracts.
`Subscriptions should be addressed to the Publisher and are payable in advance, Bulk subscriptions are
`available upon request from the Publisher. No refunds may be issued after publication of a volume’sfirst
`issue. Visit our website (www.liebertpub.com/hum) or contact our Customer Service Department for
`current subscription rates and pricing information.
`Human Gene Therapy (ISSN: 1043-0342)is owned and published 12 times a year by Mary Ann Liebert,
`Inc. Periodicals postage paid at New Rochelle, NY, andat additional mailing offices. Mailed in Canada
`under CPC CPM #40026674. Copyright © 2012. Printed in the United States of America. Postmaster:
`Send address changes to Human Gene Therapy, c/o Subscription Department, Mary Ann Liebert, Inc.,
`140 HuguenotStreet, New Rochelle, NY 10801-5215.
`Mary Ann Liebert, Inc.
`140 HuguenotStreet
`New Rochelle, NY 10801-5215
`Telephone: (914) 740-2100
`E-mail: info@liebertpub.com
`
`MaryArs Liebert, ineepi
`
`www.liebertpub.com/hum
`
`Miltenyi Ex. 1026 Page 2
`
`Miltenyi Ex. 1026 Page 2
`
`

`

`Human Gene Therapy
`
`The Official Journal of the
`Furopean Society of Gene and Cell Therapy ° British Society for Gene Therapy * French Society of Cell and Gene Therapy
`irish Society for Gene and Cell Therapy »* The Israeli Society of Gene & Cell Therapy * German Society for Gene Therapy
`Spanish Society of Gene and Gell Therapy « Netherlands Society of Gene and Cell Therapy * Finnish Society of Gene Therapy
`Akseli Hemminki, M.D.
`Izumu Saito, M.D., Ph.D.
`University of Helsinki
`University of Tokyo
`
`Ulrich R, Hengge, M.D.
`Heinrich-Heine-University
`
`Roland W. Herzog, Ph.D,
`University of Florida
`Steven Howe, Ph.D.
`UCL Institute of Child Health
`
`Wenlin Huang, Ph.D, M.D.
`Sun¥ar-sen University
`
`Carl H, June, M.D,
`University of Pennsylvania Perelman
`School af Medicine
`
`Chang-Yuil Kang, Ph.D.
`Seoul National University
`Hans-Peter Kiem, M.D.
`University of Washington School ofMedicine
`David Klatzmann, M.D., Ph.D.
`INSERM
`
`Robert M. Kotin, Ph.D.
`National Heart, Lung, and Blood Institute
`
`Francois M. Lemoine, M.D., Ph.D.
`Université Pierre and Marie Curie
`
`You Lu, M,D,
`Sichuan University
`
`Patrick Midoux, Ph.D.
`INSERM-CNRS
`
`Eugenio Montini, Ph.D.
`San Raffuele Telethon Institute
`for Gene Therapy
`
`Philippe Moullier, M.D., Ph.D.
`INSERM-Genethon
`
`Rita Mulherkar, Ph.D,
`Tata Memorial Centre
`
`Nicholas Muzyezka, Ph,D,
`University of Florida
`
`Hiroyuki Nakai, M.D., Ph.D.
`Health & Science University Schaal ofMedicine
`
`Timothy O'Brien, M.D., Ph.D,
`National University of Ireland, Galway
`Richard Peluso, Ph.D.
`Merck & Co., Ine.
`
`Katherine Ponder, M.D.
`Washington University School of Medicine
`
`ZhiYong Qian, M.D.
`Sichuan University
`
`John E.J. Rasko, M.B.B.S., Ph.D.
`CenfenaryInstitute and Royal Prince
`Alfred Hospital
`
`Paul D. Robbins, Ph.D.
`University of Pittshurgh School
`of Medicine
`John J. Rossi, Ph.D.
`City of Hope
`
`Debi P. Sarkar, Ph.D.
`University of Delhi South Campus
`
`Ton Schumacher, Ph.D.
`The Netherlands Cancer Institute
`
`Arun Srivastava, Ph.D.
`Universiry of Florida College of Medicine
`
`George Stamatoyannopoulos, M.D,, D.Sc.
`University af Washington
`
`Mark Tangney, Ph.D.
`Cork Cancer Research Center
`
`Victor W. van Beusechem, Ph.D.
`VU University Medical Cenier
`
`Christof von Kalle, M.D., Ph.D.
`National Centerfor Tumor
`Diseases (NCT)
`
`Simon N, Waddington, Ph.D.
`Jmperial College London
`
`Gerard Wagemaker, Ph.D,
`Erasmus University Medical Center
`
`Matthew D, Weitzman, Ph.D.
`The Children's Hospital of Philadelphia
`
`John EL. Wolfe, V.M.D., Ph.D.
`University of Pennsylvania
`Jon A, Wolff, M.D.
`Roche Madison Inc.
`
`Joseph C. Wu, M.D,, Ph.D.
`Stanford University School of Medicine
`
`Xiao Xiao, Ph.D.
`University of North Carolina
`
`Rafael J. Vaiiez-Munoz, Ph.D.
`Royal Holloway, University of Landon
`
`Yiping Yang, M.D., Ph.D.
`Duke University Medical Center
`
`Seppo Yla-Herttuala, M.D,, Ph.D.
`University af Kuopia
`
`Yoshi Yonemiisu, M.D, Ph.D.
`Kyushu University
`
`Gideon Zamir, M.D.
`Halayseli-Hebrew University
`Medical Center
`
`Xia Zhao, M.D,
`Sichuan University
`
`Editorial Office
`Scientific Editors
`Sadik H. Kassim, Ph.D,
`Rebeca M. Tenney
`E-wail; AGThewswire@lieberipub,com
`
`Martha Turnitsa
`Editorial Manager
`Human Gene Therapy
`University of Pennsylvania
`125 §. Fist St, Ste. 2000 TRL
`Philadelphia, PA 19104
`E-mail: ngi@mail.med.wpenn.edu
`
`Founding Editor
`W. French Anderson, M.D.
`
`Editor-in-Chief
`James M. Wilson, M.D., Ph.D.
`Univ. of Fennsylvania Perelman School
`af Medione
`Gene Thenipy Program
`Dept of Pathalogy and Laboratory Medicine
`sy 2000, Translational Research
`Laboratories
`125 §. 34st Street
`Philadelphia, PA 19104-3403
`E-mail? wilsonjm@ mail, med.upenn.edu
`
`Deputy Editor
`Professor George Dickson
`Justitite of Biomedical & Life Sciences
`Shoal of Biological Sciences
`Roval Holloway - University of London
`Egium, Surrey, TW20 OEX, UK
`E-mail: g.dickson@rhul.ac.uk
`
`
`Methods Editor
`Thierry VandenDriessche, Ph.D.
`Head-Division of Gene Therapy
`& Regenerative Medicine
`Faculty of Medicine & Pharmacy
`Free University of Brussels (VUB)
`Laarbeeklaan 103
`Building A
`B-J090 Brussels
`Belgium
`E-mail: thierry.vandendniessche@ yub.ac.be
`
`Associate Editors
`Chiara Bonini, M.D.
`Experimental Hematology Unit
`Division af Regenerative Medicine,
`Gene Therapy and Stem Cells
`Son Raffaele Scientific Instinite
`Via Olgettina 58
`20132, Milano, laly
`E-niail: bommi.chiara@hsr.it
`
`Scientific Editorial Board
`Kari Airenne, Ph.D.
`University af Easter Finland
`
`Ramon Alemany, Ph.D.
`Institut Catald d'Oncologia-IDIBELL
`Tan E. Alexander, M.B.B.S,, Ph.D.
`University of Sydney
`
`Daniel G, Anderson, Ph.D.
`Massachusetts Institute of Technology
`
`Christopher Baum, M.D., Ph.D.
`Hannover Medical School
`
`Alessundra Biff, M.D.
`San Raffaele Scientific Institute
`
`Attilio Bondanza, M.D., Ph.D.
`San Raffaele Hospital
`Xandra O. Breakefield, Ph.D.
`Massachusetts General Hospital
`Juan A. Bueren, Ph.D.
`CIEMAT
`
`Hildegard Biining, Ph.D.
`Uniklinik Kain
`
`Barrie J. Carter, Ph.D.
`Carter BioConsulting
`
`Nathalie Cartier-Lacave, M.D.
`INSERM
`
`Toni Cathomen, Ph.D.
`Hanmaver Medical School
`
`Saswati Chatierjee, Ph.D.
`City of Hope Natl. Medical Center
`
`June-Key Chung, M.D,, Ph.D.
`Seayl National University
`
`Mary Collins, Ph.D.
`Royal Free and Universiry Callege
`Medical School
`
`Terence R, Flotte, M.D.
`Gene Therapy Center
`Depariments of Pediatrics and Microbiology
`& Physiologic Systems University
`of Massachusetts Medical School
`55 Lake Avenue North, Suite $1-340
`Worcester, MA O0/655
`E-mail: terry.flotte@umassmed.edu
`
`Kenneth Cornetta, M.D.
`Indiana University School of Medicine
`
`Frangois-Loic Cosset, Ph.D.
`Ecole Normale Supérieure de Lyon
`
`Gay M. Crooks, M.B,, B.S.
`University of California, Lox Angeles
`
`Ronald G. Crystal, M.D,
`Weill Comell Medical College
`
`Anja Ehrhardt, Ph.D.
`University of Munich
`
`John F. Engelhardt, Ph.D.
`University of Jowa
`
`Erik Falck-Pedersen, Ph.D,
`Weill Comel! Medical College
`
`Sarah Ferber, Ph.D.
`Sheba Medical Center
`
`Anne Galy, Ph.D.
`GENETHON
`
`Mark A, Kay, M.D., Ph.D.
`Departments of Pediatrics and Genetics
`Stanford University School of Medicine
`269 Campus Drive #2105
`Stanford, CA 94305
`E-mail: markay@stanford.edu
`
`Adrian J, Thrasher, M.D., Ph.D.
`Centre for Immunodeficiency
`Molecular Jmmunalogy Unit
`UCL Instinite of Child Health
`40) Guilford Street
`London WCIN LEH, UK
`E-mail: A.Thrasher@ich.ucl.ac.uk
`
`Yu-quan Wei, M.D., Ph.D.
`National Key Laboratory of Biotherapy and
`Cancer Center
`West China Hospital, West China Medical
`School, Sichuan University
`Guopeng Street, Keyuan Street Road 4, No. |
`Chengdu, Sichuan 61004)
`PR. China
`E-mail: yuquanwei@scu.edu,cn
`
`Guangping Gao, Ph.D.
`University of Massachusetts Medical School
`
`David W- Russell, M.D,, Ph.D.
`University of Washingron
`
`Manuel Grez, Ph.D.
`University of Frankfurt
`
`Stephen J. Russell, M.D., Ph.D.
`Maya Clinic College of Medicine
`
`Michel Sadelain, M.D., Ph.D.
`Yajun Guo, M.D., Ph.D.
`Shanghai Second Miltary Medical University Memorial Sloan-Kettering Cancer Cenier
`
`
`r *
`
`Marg Ams Liebert, Ine.
`
`poblishers
`
`Miltenyi Ex. 1026 Page 3
`
`Miltenyi Ex. 1026 Page 3
`
`

`

`Human Gene Therapy
`
`
`
` Volume 23 Number 5 May 2012
`
`
`
`EDITORIAL
`
`A New Open Access Partner
`JM. Wilson
`
`COMMENTARY
`
`Double-Blinded, Placebo-Controlled, Randomized Gene Therapy Using Surgery
`for Vector Delivery
`R.G. Crystal, 8.M. Kaminsky, N.R. Hackett, and T.K. Rosengart
`
`GENE THERAPY BRIEFS
`A. Philippidis
`
`RESEARCH ARTICLES
`
`Infusing CD19-Directed T Cells to Augment Disease Control in Patients
`Undergoing Autologous Hematopoietic Stem-Cell Transplantation for Advanced
`B-Lymphoid Malignancies
`P. Kebriaei, H. Huls, B. Jena, M. Munsell, R. Jackson, D.A. Lee, P.B. Hackett, G. Rondon,
`E. Shpall, R.E. Champlin,-and L.J.N. Cooper
`
`Kebriaei and colleagues report on their phase 1 protocol to examine the safety and feasibility of administering
`genetically modified autologous T cells expressing a CD19-specific chimeric antigen receptor (CAR)to patients
`with high-risk B-lymphoid malignancies undergoing autologous hematopoietic stem cell transplantation, CAR
`was introduced to T cells by nonviral gene transfer, using the Sleeping Beauty system and signals through
`chimeric CD28 and CD3-t.
`
`AAVrh.10-Mediated Expression of an Anti-Cocaine Antibody Mediates Persistent
`Passive Immunization That Suppresses Cocaine-Induced Behavior
`J.B. Rosenberg, M.J, Hicks, B.P. De, O. Pagovich, E. Frenk, K.D. Janda, S. Wee, G.F. Koob,
`N.R. Hackett, S.M. Kaminsky, S. Worgall, N. Tignor, J.G. Mezey, and R.G. Crystal
`
`Rosenberg and colleagues construct an AAV serotype rh.10 vector that encodes the heavy andlight chains
`of the high-affinity anti-cocaine monoclonal antibody GNC92H2. They demonstrate that intravenous administration
`of this vector leads to high serum levels of cocaine-specific antibodies that block administered cocaine from access
`to ihe brain, prevent cocaine-induced hyperlocomotoractivity, and lead to anticocaine passive immunity in mice.
`
`Immunodominant Liver-Specific Expression Suppresses Transgene-Directed
`Immune Responsesin Murine Pompe Disease
`P. Zhang, B. Sun, T. Osada, R. Rodtiguiz, X.Y. Yang, X. Luo, A.R. Kemper, T.M. Clay,
`and D.D. Koeberl
`
`Zhang and colleagues investigate the mechanism for immunomodulatory gene therapy by evaluating two AAV
`vectors encoding human acid a-glucosidase (hGAA), including both a tolerogenic vector containing theliver-specific
`promoter (LSP) and an immunogenic vector containing the ubiquitously active cytomegalovirus enhancer and chicken
`B-actin promoter (CB) regulatory cassette. The authors find that simultaneous administration of tolerogenic
`and immunogenic vectors induces immunetolerance in mice and leads to higher levels ofliver transduction,
`in comparison with either vector alone,
`
`Characterization of an HIV-Targeted Transcriptional Gene-Silencing RNA
`in Primary Cells
`A.-M.W. Turner, A.M. Ackley, M.A. Matrone, and K.V. Morris
`
`437
`
`438
`
`442
`
`444
`
`451
`
`460
`
`473
`
`Turner and colleagues demonstrate that transcriptional gene silencing (TGS) can reduce viral transcription
`in primary human CD4*Tcells, although increasing viral burden resulted in the loss of this antiviral effect.
`Studies into off-target effects additionally identified a novel potential interaction between the small nucleolar
`RNA pathway and the TGS-based antisense RNA, resulting in activation of p53.
`
`(continued)
`
`Miltenyi Ex. 1026 Page 4
`
`Miltenyi Ex. 1026 Page 4
`
`

`

`re
`
`*
`=
`
`ie
`A
`a
`
`byl
`
`
`
`
`Retargeting Vesicular Stomatitis Virus Using Measles Virus Envelope Glycoproteins
`C. Ayala-Breton, G.N. Barber, S.J. Russell, and K.-W. Peng
`Ayala-Breton and colleaques use the measles virus (MV) hemagglutinin (H) and fusion (F) envelope glycoproteins
`ta redirect vesicular stomatitis virus (VSV) entry and infection specifically to tumor-associated receptors
`such as epidermal growth factor receptor,folate receptor, and prostate membrane-specific antigen. /n vivo
`expression ofthe all retargeted VSV was restricted to receptor-positive human tumor xenograits.
`
`Development and Validation of Novel AAV2 Random Libraries
`Displaying Peptides of Diverse Lengths and at Diverse Capsid Positions
`M. Naumer, Y. Ying, S. Michelfelder, A. Reuter, M. Trepel, O.J. Muller, and J.A. Kleinschmidt
`Naumer and colleagues expand the combinatorial AAV2 display system with a panelof novel AAVlibraries displaying
`peptides of 5, 7, 12, 19, or 26 amino acids in length at capsid position 588 or 7-mer peptides at position 453.
`Characterization of vector selectivity by transduction of nontarget cells and comparative genetransduction analysis
`using a panel of 44 human tumorcell lines revealed that insertion of different-length peptides allows targeting
`of distinct cellular receptors for cell entry with similar efficiency, but different selectivity.
`
`484
`
`A492
`
`Silencing of miR20a Is Crucial for Ngn1-Mediated Neuroprotectionin Injured
`Spinal Cord
`M.K. Jee, J.S. Jung, Y.B. Im, S.J. Jung, and S.K. Kang
`Jee and colleagues demonstrate that small interfering RNA (siRNA)-mediatedinhibition of microRNA 20a (miR20a)
`leads to increased expression of a key neuronaltranscription factor, neurogenin-1 (Ngn1). They show that siRNA-
`mediated knockdown of miR20a enhances neural cell survival and neurogenesis and leads to recovery of motor
`function in a mouse model of traumatic spinal cord injury.
`Asymmetric siRNA: New Strategy to Improve Specificity and Reduce Off-Target
`Gene Expression
`Z, Yuan, X. Wu, C. Liu, G. Xu, and Z. Wu
`
`508
`
`521
`
`In this study, Yuan and colleagues demonstrate that asymmetrically shortened siRNAs cansilence their targets
`as efficiently as their traditional 19+ 2 counterpartsin vitro. Moreover, the authors suggestthat shortening siRNAs
`from their 3‘ end improves the loading specificity of the RNA-induced silencing complex and results in feweroff-target
`gene expression effects.
`
`BRIEF REPORT
`
`Hepatic Gene Transfer in Neonatal Mice by Adeno-Associated Virus
`Serotype 8 Vector
`L, Wang, H. Wang, P. Bell, D. McMenamin, and J.M, Wilson
`Wang and colleagues conducta series of preclinical animal studies examining the kinetics of AAV gene transfer.
`They demonstrate that self-complementary AAV8results in robust andefficient hepatic genetranster in neonatal
`mice. Yet, this transduction quickly decreases over a few weeks because ofvectordilution caused by rapid cell
`proliferation in the liver of growing young mice.
`
`
`533
`
`British Society for Gene Therapy 2012
`March 9, 2012
`UCL Institute of Child Health, London
`
`pp A1-A22
`ABSTRACTS AVAILABLE ONLINE ONLY
`
`
`Instructions for Authors can be found on our website at www.liebertpub.com/hum
`
`Cover: Adeno-associated virus type 2 (AAV2) capsid protein structures showing two peptide insertion
`sites at the top of the spikes around the threefold axes, enabling the development of AAV2 random
`libraries displaying peptides at two different positions. Insertion sites are marked together with two
`neighboring amino acids in purple (arginine 588) and cyan (glycine 453) in the side and top view of a
`capsidprotein trimer. For further details, see the article by Naumeref a/. on page 492.
`
`MaryArn Liebert, ee
`
`www.liebertpub.com/hum
`
`Miltenyi Ex. 1026 Page 5
`
`Miltenyi Ex. 1026 Page 5
`
`

`

`
`
`HUMAN GENE THERAPY 23:444-450 (May 2012)
`© Mary Ann Liebert, Inc.
`DOI: 10.1089/hum.2011 .167
`
`ResearchArticles
`
`Infusing CD19-Directed T Cells to Augment Disease Control
`in Patients Undergoing Autologous Hematopoietic Stem-Cell
`Transplantation for Advanced B-Lymphoid Malignancies
`
`Partow Kebriaei’ Helen Huls2 Bipulendu Jena? Mark Munsell! Rineka Jackson? Dean A. Lee??
`Perry B, Hackett? Gabriela Rondon! Elizabeth Shpall! Richard E. Champlin! and Laurence J.N. Cooper?*
`
`Abstract
`Limited curative treatment options exist for patients with advanced B-lymphoid malignancies, and new ther-
`apeutic approaches are needed to augmenttheefficacy of hematopoietic stem-cell transplantation (HSCT).
`Cellular therapies, such as adoptive transferofT cells that are being evaluated to target malignant disease, use
`mechanisms independent of chemo- and radiotherapy with nonoverlapping toxicities. Gene therapy is em-
`ployed to generate tumor-specific T cells, as specificity can be redirected through enforced expression of a
`chimeric antigen receptor (CAR) to achieve antigen recognition based on the specificity of a monoclonal anti-
`body. By combining cell and gene therapies, we have opened a new Phase | protocolat the MD Anderson Cancer
`Center (Houston, TX) to examine the safety and feasibility of administering autologous genetically modified T
`cells expressing a CD19-specific CAR (capable of signaling through chimeric CD28 and CD3-() into patients with
`high-risk B-lymphoid malignancies undergoing autologous HSCT. The T cells are genetically modified by
`nonviral gene transfer of the Sleeping Beauty system and CAR™Tcells selectively propagated in a CAR-de-
`pendent manneron designerartificial antigen-presenting cells. The results of this study will lay the foundation
`for future protocols including CAR” T-cell infusions derived from allogeneic sources.
`
`Introduction
`
`for patients with
`oe TREATMENT OPTIONS exist
`B-lymphoid malignancies whorelapse after autologous or
`allogeneic hematopoietic stem-cell transplantation (HSCT). Re-
`garding non-Hodgkin lymphoma (NHL), 50% of patients who
`relapse after conventional chemotherapy may be salvaged by
`autologous HSCT. Amongthose whoare transplanted,residual
`disease at the time oftransplantation predicts an increased rate
`of relapse (Mills et al., 1995; Caballero et al., 2003), Allogeneic
`HSCT, which may deliver a potent graft-versus-tumor (GVT)
`effect mediated by donor-derived immunocompetentcells, can
`be potentially curative in 30-50%of patients (Ratanatharathorn
`et al, 1994; Verdonck, 1999). However, because of the high
`treatment-related mortality (TRM) associated with allogeneic
`HSCT,the GVT effect is not always correlated with improved
`overall survival (Peniket et al., 2003). Thus, novel therapeutic
`approaches are urgently needed for these patients.
`
`T-cell therapy can specifically target disease, employing
`anti-tumor mechanisms that are nonoverlapping with the
`conditioning regimen used for HSCT. Therefore, investiga-
`tors have developed adoptive immunotherapy as a strat-
`egy to augment
`the GVT effect. To overcome immune
`tolerance to tumor-associated antigens (TAAs)investigators
`have developed and implemented immunotherapies infus-
`ing T cells genetically modified to express a chimeric antigen
`receptor (CAR)(Paulos etal., 2008;Jenaet al., 2010). CARs are
`typically engineered to provide the genetically modified T
`cell with the specificity of a murine monoclonal antibody
`and, on binding a cell surface TAA, can specifically activate
`the T cell for proliferation, cytokine production, and cytolysis
`(Eshharef al., 1993; Brentjenset al., 2003; Cooper et al., 2003;
`Park et al,, 2007; Brenner and Heslop, 2010; Porter et al.,
`2011). CARs are “universal” in that they bind TAAs inde-
`pendent of major histocompatibility complex (MHC), and
`thus one receptor construct can be used to treat a population
`
`
`Ipivision of Cancer Medicine, M.D. Anderson Cancer Center, Houston, TX 77005.
`2Division of Pediatrics, M.D. Anderson Cancer Center, Houston, TX 77005.
`3Graduate Schoolof Biomedical Sciences, University of Texas, Houston, TX 77005.
`“Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455.
`
`444
`
`Miltenyi Ex. 1026 Page 6
`
`Miltenyi Ex. 1026 Page 6
`
`

`

`DISEASE CONTROL AUGMENTED WITH CD19-DIRECTED T CELLS
`
`445
`
`lymphoma (SLL), chronic lymphocytic leukemia (CLL), fol-
`licular lymphoma, or mantle cell lymphoma;that are beyond
`first relapse or primary refractory to conventional treatment.
`Additionaleligibility criteria include adequate organ func-
`tion, a Zubrod performancestatus of 0 or 1, no evidence of
`uncontrolled infection, and negative serology for hepatitis B
`(HBV), hepatitis C (HCV), and HIV. Patients with active
`CNSdisease are excluded. In addition, patients with known
`allergy to bovine or murine products will be excluded, Fur-
`thermore, patients must not be taking systemic corticoste-
`roids within 3 days before T-cell infusion or experiencing any
`new significant toxicity within 24hours before T-cell infu-
`sion. Last, to be eligible for [L-2 injections after administra-
`tions of T cells, patients must not experience any new
`significant adverse events (AEs) probably or definitely at-
`tributed ta the adoptively transferred T cells.
`
`T cell manufacture and release
`
`of patients with the same TAA* tumors. Multiple CAR de-
`signs have emerged to dock with multiple TAAs and to
`provide a fully competentactivation signal in desired T-cell
`subsets (Berry et al., 2009; June etal., 2009; Jena et al., 2010).
`CAR* T cells have been tested in multiple trials at multiple
`centers, as we have reviewed (Jena etal., 2010).
`Here we describe a new PhaseI cell and gene immuno-
`therapy trial administering autologous ex vivo-expanded
`genetically modified T cells expressing a CD19-specific CAR
`into patients with high-risk B-lymphoid malignancies un-
`dergoing autologous HSCT.Therationale for redirecting the
`specificity of T cells for CD19, using a CAR,is based on the
`following: (1) CD19 is a B-lineage cell surface TAA expressed
`on lymphoid malignancies; (2) CD19 is not expressed on
`hematopoietic stem cells (HSC) and (3) soluble CD19 is ap-
`parently not shed into the circulation (to act as a competitor
`for binding CAR to CD19 on tumorcells). Preliminary
`studies demonstrated the safety, feasibility, and efficacy of
`infusing CD19-specific CAR therapy for lymphoid malig-
`The two SB DNAplasmids (manufactured by Waisman
`nancies (Kochenderferet al., 2010). Our results on infusing a
`Biomanufacturing, Madison, WI), encoding the CAR (des-
`first-generation CD19-specific CAR into patients with re-
`ignated CD19RCD28) transposon (Cooper, 2007; Singhetal.,
`current follicular lymphoma after a preparative regimen
`2008; Jena et al., 2010) and SB11 transposase (Singh et al.,
`consisting of rituximab and fludarabine demonstrated limited
`2008; Manuri et al., 2009), will be simultaneously electro-
`persistence due to mcomplete signaling through CD3-{ and an
`transferred into T cells with Nucleofector device (Lonza
`immune response to immunogenic transgenes coexpressed
`Group, Basel, Switzerland). The genetically modified T cells
`with CAR inTcells (Jensen, 2007). Therefore, we developed a
`will be selectively propagated in a CAR-dependent manner
`new platform for the genetic modification of T cells based on
`on y-irradiated K562 cells that have been genetically modi-
`(1) a second-generation CAR that signals through both chi-
`fied by transduction with lentivirus (in collaboration with C.
`meric CD28 and CD3-{ (Singh, 2008), (2) electrotransfer of the
`June at the University of Pennsylvania, Philadelphia, PA)
`Sleeping Beauty (SB) system to improve integration efficiency
`and cloned by limiting dilution to function as CD19* aAPC.
`of CAR transgene, and (3) outgrowth of CAR* T cells on
`The CD19RCD28 transgene is a second-generation CAR that
`designerartificial antigen-presenting cells (aAPC)to select for
`activates T cells via chimeric CD28 and CD3-f. The aAPC
`T cells with proven proliferative potential.
`(clone #4) were manufactured as a Master Cell Bank by
`Production Assistance for Cellular Therapies (PACT) under
`the auspices of the National Heart, Lung, and Blood Institute
`(NHLBI, Bethesda, MD). Subsequently, a Working Cell Bank
`of clone #4 was derived at MDACC and used to support
`numeric expansion of CAR* T cells as cultured in the pres-
`ence of soluble recombinant human IL-2 and IL-21. The
`validation studies describing phenotype and function of the
`CAR* T cells manufactured in compliance with current good
`manufacturing practice (CGMP) are being published (Singh
`et al., 2012).
`In the event
`there is an overgrowth of
`CD3"°®CD56* lymphocytes early in the culturing process,
`these cells will be removed using CD56-specific monoclonal
`antibody and paramagnetic selection. The electroporation
`and propagation ofclinical-grade T cells will occur at the
`MDACC in compliance with cGMP for Phase I/II trials.
`The release criteria, undertaken in compliance with Clinical
`Laboratory Improvement Amendments (CLIA),
`for
`the
`manufactured T cells are (1) sterility (bacteria, fungi, myco-
`plasma, endotoxin),
`(2) chain of custody (low-resolution
`MHCclass I typing), (3) phenotype (presence of T cells [CD3
`expression], presence of transgene [CAR expression], ab-
`sence of aAPC [CD32 expression], absence of B cells [CD19
`expression]), (4) safety (absence of autonomous cell growth),
`and (5) viability.
`
`Clinical Trial Design
`
`Regulatory approvals
`
`The clinical trial is open only at the MD Anderson Cancer
`Center (MDACC, Houston, TX) as IRB #2007-0635 (IBC
`#RM0508-070). Approval was obtained from the NIH Office
`of Biotechnology Activities (OBA, #0804-922) and U.S. Food
`and Drug Administration (FDA) (IND #14193). Thetrial is
`listed at ClinicalTrials.gov (identifier: NCT00968760).
`
`Objectives
`
`The primary objective is to assess the safety, feasibility,
`and persistence of autologous ex vivo-expanded genetically
`modified CD19-specific CAR®* T cells intravenously admin-
`istered to patients with persistent or relapsed CD19* lym-
`phoid malignancies, The secondary objectives will include
`(1) screening for the developmentof host immune responses
`against the CD19-specific CAR, (2) describing the ability of
`the infused T cells to hometo sites of disease, such as bone
`marrow and lymph nodes, (3) assessing the impact of in-
`terleukin (IL)-2 on T-cell persistence, and (4) assessing dis-
`ease response.
`
`Patient eligibility
`
`Patients are eligible if they are between 18 and 60 years of
`age with advanced CD19* lymphoid malignancies, includ-
`ing non-Hodgkin's lymphoma (NHL), small
`lymphocytic
`
`Peripheral blood stem cell mobilization and collection
`
`The methods of peripheral blood stem cell (PBSC) mobi-
`lization, collection, storage, and infusion have been described
`
`Miltenyi Ex. 1026 Page 7
`
`Miltenyi Ex. 1026 Page 7
`
`

`

`446
`
`KEBRIAEI ET AL.
`
`TABLE 1, RESEARCH PARTICIPANT ASSIGNMENT
`to T Ceti Dosinc CoHoRTS
`
`
`
` Dose cohort Single T cell dose* IL-2
`
`
`
`No
`>10’/m? but <5x10’/m?
`Dose level X
`No
`>5x107/m?* but <5 10°/m?
`Dose level A
`No
`>5x108/m? but <5x10°/m?
`Dose level B
`Yes
`>5x107/m? but <5 10°/m?
`Dose level C
`
`
`>5x10°/m? but <5 10°/m?Dose level D Yes
`
`*Tcell dosing, starting at Dose level A, is calculated on the basis of
`prefteeze counts, and infused over a 2-day split.
`
`istered as a single daily injection at 0.3 x 10° [U/m*beginning
`within 2 days of the last split of the T-cell infusion and
`continuing for up to 14 daily doses. Patients may be pre-
`medicated with acetaminophen and diphenhydramine be-
`fore each injection. The [L-2 dose will be reduced by 50%if
`the patient develops a new adverse event of grade >2 (CTC,
`version 4) involving cardiopulmonary, hepatic (excluding al-
`bumin), gastrointestinal, neurological, or renal toxicity prob-
`ably or definitely attributed to IL-2 administration.IL-2 will be
`stopped in patients with persistent grade >2 (CTC,version 4)
`toxicity despite IL-2 dose reduction. Patients taking IL-2 with
`a grade 4 adverse event will stop IL-2. If the adverse event
`does not improve to grade <3 within 36 hr of stopping IL-2,
`then corticosteroids will be started. Isolated injection site skin
`toxicity attributable to IL-2 administration may be an indica-
`tion to discontinue IL-2, but not to ablate T cells.
`
`Supportive care
`Institutional HSCT guidelines for antimicrobial, antifun-
`gal, and antiviral prophylaxis will be followed. Patients will
`not receive filgrastim to enhance neutrophil recovery unless
`there is concern for serious infection. Packed red bloodcells
`will be administered to maintain hemoglobin levels 2>8 g/dl.
`Platelet transfusions will be administered to keep platelet
`counts 210x10°/liter. All blood products are filtered and
`irradiated, All research participants who are able to have
`children mustpractice effective birth control while on study.
`
`Study assessments
`
`Miltenyi Ex. 1026 Page 8
`
`Patients will have an initial assessment to include HLA
`typing, serology for cytomegalovirus (CMV), HIV, human T-
`lymphotropic virus (HTLV)-1, HCV, and HBV, as well as
`peripheral blood (PB) collected before manufacture of CAR*
`T cells and analyzed for protein expression and genetic
`profiling. Before initiating the conditioning regimen for
`HSCT,
`the patients will undergo institutional standard-of
`care evaluations to validate adequate organ functions and
`standard restaging studies, including bone marrow biopsy
`and positron emission tomography/computed tomography
`(PET/CT)scans, as clinically indicated. Immediately after the
`T-cell infusion (last split dose) and then weekly for 2 weeks,
`and then at 1, 3, 6, and 12 months, the research participants
`will receive physical and laboratory evaluations, such as PB
`for protein expression and genetic profiling, including PCR
`After we have established safety and feasibility of the
`analyses for the presence ofinfusedTcells. Skewing ofthe T-
`T-cell dosing in the first two cohorts of the study (Table 1),
`cell receptor repertoire may indicate that someinfusedTcells
`we will assess whether low-dose IL-2 can support the sur-
`can preferentially survive and thus the PB will be serially
`vival and/or numeric expansion of CAR* T cells in the last
`analyzed for emergenceofoligoclonal or clonal population(s)
`two cohorts ofthe trial. IL-2 will be subcutaneously admin-
`
`(Hosing et al., 2006). Patients will receive nonpurged autol-
`ogous HSC collected after mobilization with filgrastim and
`chemotherapy. The target progenitor cell dose is 410°
`CD34" cells/kg with a minimal acceptable dose of 2x 10°
`CD34* celis/kg. Patients failing to reach that target cell
`number can undergo bone marrow harvestat the discretion
`of the treating physician. Bone marrow will be obtained by
`multiple aspirations from the right and left iliac crest under
`general anesthesia, with a target total nucleated cell dose of
`3x 108 cells/kg. All products are cryopreserved according to
`standardinstitutional techniques.
`
`Conditioning regimen for HSCT
`
`We will use our standard-of-care conditioning regimen
`as a means to lympho-deplete the patients in an effort to
`improve the persistence of CAR* T cells. The conditioning
`regimen consists of intravenous carmustine on day —6 ata
`(300 mg/m?) infused over 2hours, fo

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